analysis of teashirt mutants affecting cell...

255
UNIVERSIDADE DE LISBOA Faculdade de Ciências Departamento de Biologia Vegetal Analysis of teashirt mutants affecting cell proliferation in Drosophila melanogaster Silvia Alexandra Barbosa Jacome Pimentel DOUTORAMENTO EM BIOLOGIA (Genética) Lisboa 2010

Upload: others

Post on 03-Jul-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

UNIVERSIDADE DE LISBOA

Faculdade de Ciências

Departamento de Biologia Vegetal

Analysis of teashirt mutants affecting cell

proliferation in

Drosophila melanogaster

Silvia Alexandra Barbosa Jacome Pimentel

DOUTORAMENTO EM BIOLOGIA (Genética)

Lisboa 2010

Page 2: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal
Page 3: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

UNIVERSIDADE DE LISBOA

Faculdade de Ciências

Departamento de Biologia Vegetal

Analysis of teashirt mutants affecting cell

proliferation in

Drosophila melanogaster

Silvia Alexandra Barbosa Jacome Pimentel

DOUTORAMENTO EM BIOLOGIA (Genética)

Supervisors: Rui Gomes

Laurent Fasano

Lisboa 2010

Page 4: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II

Page 5: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Por ti, Alexandre.

The most important thing in life is not the triumph but the struggle.

Page 6: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

IV

Page 7: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

This PhD work was support by my PhD grant (SFRH/BD4861/2001 and

SFRH/BD/13233/2003) from the POCI 2010 (Fundo Social Europeu_ FSE) approved by

Fundação para a Ciência e a Tecnologia (Portugal), and also by the collaboration between the

Rui Gomes lab and Laurent Fasano labs (CNRS and the Association pour la Recherche Contre le

Cancer and La Ligue, France).

V

Page 8: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

VI

Page 9: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Acknowledgements

I would like to thank to all the people that directly or indirectly contributed for the

concretisation of this work:

Ao meu Orientador, Professor Rui Gomes, por me ter acolhido no seu grupo, pela

oportunidade que me deu em realizar este trabalho no seu laboratório, pelo seu esforço em

arranjar alternativas que permitissem ultrapassar as nosssas limitações financeiras e técnicas,

por todas as sugestões e discussões. E pela sua boa disposição e humor.

Au Laurent Fasano, je lui remercie pour m’avoir accueilli dans son labo, pour toute sa

disponibilité et patience avec moi, pour me faire confiance et me donner confiance dans mes

résultats, pour toutes les suggestions et discussions et pour me montrer une autre forme de

faire sience.

À Fundação para a Ciência e Tecnologia (FCT), pelo apoio financeiro, sem o qual teria

sido impossível a realização deste trabalho.

To the Centro de Genética e Biologia Molecular (CGBM) and to the Institut de Biologie

du Développement du Luminy (IBDML) for giving me conditions for the development of this

work.

To all the people that contributed with the necessary reagents needed for the

elaboration of this work, mainly to Dr. Alvaro Tavares, Dr. Claudio Sunkel, Dr.Roger Karess,

Dr. Steve Kerridge, Dr. Stephen Cohen, Dr. Armel Gallet, Dra. Ruth Lehmann, Dr. E. McCarthy

and to Bloomington Drosophila stock Centres.

Aos meus colegas do CGBM, pela entre-ajuda, pela amizade, pelo convívio e pelas

“brincadeiras”, que tornaram o nosso dia-a-dia mais agradável e produtivo. Um OBRIGADO

especial para os colegas de grupo, Cristina e Paulo, e para a minha “mamã” científica, Isabel,

que me revelou o maravilhoso mundo das moscas e me fez dar os primeiros passos no ciclo

celular, com muita paciência e enorme boa disposição.

À toutes les personnes qui m’ont aidé dans l’IBDML, sans oublier les personnes des

plate-formes techniques qui ont facilité énormément mon séjour à Marseille. Un gros MERCI à

tous les membres de l’équipe KF que j’ai connus (Laurent, Natalie, Xavier, Christine, Pierre,

Virginia, Hervé, Elise, Steve, Delphine, Mylene, Bernard, Sébastien, Lucile et Fred), pour votre

Page 10: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Acknowledgements

accueil et patience d’essayer entendre mon luso-français, pour tous les conseils et discussions

et pour votre amitié. Je ne peux pas oublier de vous remercier pour tous les bons moments,

les blagues, les rigolades et les petites fêtes qui m’ont fait rester presque un année de plus

que le prévu! Je vous remercie la magnifique soirée que vous avez organisée avant mon

départ et que je ne l’oublierai jamais. Je veux exprimer ma gratification à Laurent et à

Christine pour tout le temps dépensé à regarder et à discuter mes résultats et surtout me

papier, à Delphine pour tous les protocoles, les anticorps, les solutions et à Steve pour tous les

stocks de moches. Un Gros Merci à vous tous !

Ao Dr. Álvaro Tavares e a todos os membros da sua Equipe que tive o prazer de

conhecer (à Susana, à Célia, à Mariana, à Cláudia e ao Paulo), gostaria de agradecer o facto de

nos terem aberto a porta sempre que precisámos, inclusive para a realização das experiências

de RNAi.

A toda a minha família e amigos, inclusive aos que infelizmente já partiram, um

obrigado pelos telefonemas e mails que encurtaram a distância e pelo apoio que prestaram.

Um agradecimento aos meus pais e irmão por gostarem de mim e pelo carinho que sempre me

deram, cada um à sua maneira. Um agradecimento aos quatro avos do Alex por todas as

vezes que ficaram com ele para que eu terminasse a tese. E à minha prima Sara pelas

enumeras vezes que fiquei em sua casa, nesta minha vida de viagens.

Finalmente, porque os últimos são os primeiros, obrigada César, por me aturares, pelo

teu apoio, por estares sempre a meu lado, pelo teu amor e ao Alex, apesar de todo o trabalho

e desgaste associado, obrigado por teres nascido e seres meu filho. Os teus sorrisos e

beijinhos são a motivação para não desistir e continuar lutando para ultrapassar a pouco e

pouco os obstàculos que vão surgindo.

VIII

Page 11: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Objectives, Methodological Approach and Structure of the Thesis

The main goal of this thesis was to clarify the role of the transcription factor Teashirt

(Tsh) during cell cycle progression and proliferation. Previous work in our lab showed that the

late larval lethal mutants tshA3-2-66 and tshNG1 present atrophied brains and strong reduced

imaginal discs with a strong mitotic arrest in a metaphase-like stage (Salazar, 2000). We

performed a better characterisation of the mitotic phenotype by immunostaining and by the

analysis of the phenotype of tsh in double mutant constructs. The depletion of tsh in S2 cells

by the interference RNA technique (RNAi) was performed in order to compare with the

phenotype observed in tsh mutant neuroblasts. The analysis of the expression pattern in

mutant brains by microarrays was also performed in order to identify an obvious Tsh target

among cell cycle genes. Unfortunately, no canonical cell cycle target gene was found to be

particularly upregulated or downregulated. Furthermore, our detailed characterisation of the

mitotic phenotype revealed only a few structural abnormalities or defects, not enough to cause

such a strong mitotic arrest. All these results are presented in chapter II (part IV) of this

manuscript.

In order to prove that tsh is the gene responsible for the mitotic arrest observed in

tshA3-2-66 neuroblasts we performed P-element excision mutagenesis to get a revertant. During

this mutagenesis scheme, we obtained news alleles and, among themselves, we identified a

potential tsh female sterile mutations causing atrophied ovaries and which do not lay eggs.

This interesting phenotype led us to study the tsh role during Drosophila oogenesis. The 5’

region of tsh gene was sequenced in three of the six female sterile lines obtained. It was

characterised the ovary phenotype and functional studies have been performed in vivo using

the UAS/Gal4 system. The mutagenesis was made in Rui Gomes’ lab and the characterisation

of the sterile female mutants was mostly performed in Laurent Fasano lab. All the results are

presented in chapter II (part I, II and III) of this manuscript.

Curiously, these independent studies of tsh function had given convergent results. We

concluded that tsh is required for the normal progression of the cell cycle in L3 neuroblasts and

depleted S2 cells, as in the proliferation of the follicle stem cells during oogenesis (this is

detailed discussed in chapter III).

During all this PhD work, the main concern was to prove that the tsh gene was required

for cell proliferation since the progression of the cell cycle was affected in the three Drosophila

systems (mutant neuroblasts, S2 cells and ovaries). Thus, the introduction of this manuscript

will start with a description of the tsh gene and their reported functions during Drosophila

development (Chapter I part I). The introduction will be followed by a overview of oogenesis,

with special incidence in the principal pathway required for the proliferation and maintenance

of the three type of stem cells found in the Drosophila germarium (germline stem cells, follicle

Page 12: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Objectives, Methodological Approach and Structure of the Thesis

stem cells and escort stem cells; chapter I part II). Finally, a brief review about the cell cycle

focalised in the metaphase/ anaphase transition will end the introduction (chapter I, part III).

The results will not be presented by chronological order. The major results were

obtained during the study of tsh in the follicle stem cells proliferation. Thus, chapter II

(results) will start with P-element excision mutagenesis performed to obtain the new tsh alleles

(part I). Next, it will be characterised and described the tsh function in follicle stem cells (part

II). A reference about the mutual repression between tsh and their paralog tiptop during

oogenesis will be presented afterwards (part III). The results will end with the presentation of

all the evidence implicating tsh in mitosis progression (part IV).

A final discussion will be presented in the chapter III. Here, the major conclusion will be

discussed, as well as the new questions raised with the results presented in this PhD work.

In this manuscript, I did not dedicated one chapter exclusively to the material and

methods used during my PhD work, since they are presented in each part of the results

(chapter II).

All the references mentioned in chapters I, II and III are presented in the end of this

manuscript.

Objective

The main goal of this thesis was to clarify the role of Tsh in cell cycle progression and to

identify putative cell cycle target. For that, it was studied the requirement of Tsh to the

progression of cell cycle using three Drosophila model systems: (1) ovaries to study follicle

stem cells proliferation during oogenesis and, (2) neuroblasts and (3) S2 cells to analyse the

progression of mitosis onset. In addition, during the development of this work, I proposed to

characterise, for the first time, the Tsh function in Drosophila oogenesis.

The results presented in this manuscript give evidence that Tsh is required for cell

proliferation in Drosophila neuroblasts, S2 cells and ovaries. Furthermore, it was showed that

Tsh acts through the Hedgehog pathway to mediate follicle stem cells proliferation in ovaries.

Finally, we demonstrated that both in neuroblasts or S2 cells the absence of Tsh arrest cells in

metaphase/ anaphase transition.

X

Page 13: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Table of Contents

Acknowledgements ....................................................................................................................................... VII

Objectives, Methodological Approach and Structure of the Thesis ........................................................................ IX

Table of Contents .......................................................................................................................................... XI

Liste of Figures............................................................................................................................................XIII

Liste of Tables.............................................................................................................................................. XV

Liste of Abbreviatures .................................................................................................................................. XVI

Resumo..................................................................................................................................................... XVII

Palavras Chave: ........................................................................................................................................... XX

Abstract .................................................................................................................................................... XVII

Key Word: .................................................................................................................................................. XXI

CHAPTER I_ INTRODUCTION

I- The teashirt (tsh) gene............................................................................................................................ 3

I_1- Characterisation of the tsh gene .................................................................................................................3

I_2- Expression patterning of the tsh mRNA and protein in Drosophila ...................................................................4

I_3- Modulators of the tsh expression during embryogenesis ................................................................................5

I_4- Tsh functions during Drosophila development...............................................................................................5

I_4.1- Homeotic function of Tsh: the identity of the trunk and the specification of the prothoracic identity..................6

I_4.2- Tsh in the segmental polarity: modulation of the Wingless pathway and regulation of the hedgehog pathway

target genes ...................................................................................................................................................8

I_4.3- Tsh and the midgut morphogenesis........................................................................................................11

I_4.4- Tsh in the Drosophila Pax6/ So/ Eya network during eye development .......................................................13

I_4.5.2- tsh promotes the identity of the proximal structures of wing and halteres discs ........................................16

I_5- tiptop as a tsh paralog ............................................................................................................................18

I_6- The vertebrate tsh family.........................................................................................................................20

II- Overview of Drosophila Oogenesis ....................................................................................................... 23

II_1- The Drosophila reproductive system ........................................................................................................23

II_2- Drosophila germarium offers the unique opportunity of to study three types of stem cells and their niches .......25

II_3- Origin of GSC, TF and CC........................................................................................................................27

II_4- Ovary stem cells and their molecular mechanisms of regulation ..................................................................28

II_4.1- Genes and Pathways that control GSC self-renewal, proliferation and differentiation ...................................28

II_4.1.1- BMP, Piwi and Yb from the niche to regulate the maintenance and division of the GSC..............................29

II_4.1.2- External factors: Nutrition.................................................................................................................32

II_4.1.3- Intrinsic factors in GSC maintenance and differentiation .......................................................................33

II_4.1.3.1- Translational factor: Pumilio (Pum) , Nanos (Nos), Vasa (Vas) and Pelota (Pelo) ...................................33

II_4.1.3.2- MicroRNA (miRNA) and repeat-associated small interfering RNA (rasiRNA) is important for GSC proliferation

and maintenance...........................................................................................................................................33

II_4.1.3.3- Junctional proteins: DE-cadherin, Armadillo (Arm) and Zero population growth (Zpg)............................34

II_4.1.3.4- Cell cycle regulators: Cyclin B (CycB) and Imitation switch (ISWI) ......................................................35

II_4.1.3.5- Differentiation promoting factors: Bag of marbles (Bam), Benign germ cell neoplasm (Bgcn), Orb, Sex

lethal (Sxl) and ovarian tumor (Otu) ................................................................................................................36

II_4.2- Genes and Pathways that control FSC self-renewal, proliferation and differentiation....................................38

II_4.2.1- Molecular mechanisms shared by GSC and FSC for self-renewal and proliferation ....................................38

II_4.2.2- Hedgehog (Hh) and Wingless (Wg), two critical signals for FSC maintenance and proliferation...................38

Page 14: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Table of Contents

II_4.2.2.1- Wingless (Wg) signalling is essential for FSC maintenance and proliferation .........................................40

II_4.2.2.2- Hedgehog (Hh) signalling is essential for FSC maintenance and proliferation ........................................40

II_4.3- Genes and Pathways controlling the maintenance of ESC and EC..............................................................43

II_5- Drosophila ovary as a model system in stem cells studies. .........................................................................43

III- Overview of Drosophila Cell Cycle....................................................................................................... 44

III_1- The Cell Cycle......................................................................................................................................44

III_2- M-phase..............................................................................................................................................45

III_2.1- Mitosis Promoting Factor (MPF) and Mitosis entry ..................................................................................48

III_2.1.1- CdK1 family ...................................................................................................................................49

III_2.1.2- Cyclin A.........................................................................................................................................50

III_2.1.3- Cyclin B.........................................................................................................................................51

III_3- The bipolar spindle and the kinetocore-microtubule attachment until the formation of the metaphase plate.....53

III_3.1- The Kinetochore ................................................................................................................................55

III_4- The anaphase promoting complex/ Cyclosome (APC/C).............................................................................56

III_4.1- The APC/C activators and substrate recognition.....................................................................................57

III_4.2- APC/C regulation ...............................................................................................................................57

III_5- The regulation of the cell cycle: the checkpoints ......................................................................................58

III_5.1- DNA damage checkpoint ....................................................................................................................59

III_5.2- Metaphase or Spindle assembly checkpoint (SAC) .................................................................................61

III_5.2.1- Models for spindle assembly checkpoint sense the defects ...................................................................63

III_5.2.1.1- Microtubule attachment model.......................................................................................................64

III_5.2.1.2- Tension model .............................................................................................................................65

III_5.2.2- Inhibition of APC/C by the spindle assembly checkpoint.......................................................................66

III_5.2.2.1- Recruitment of spindle checkpoint proteins to unattached/ untensed kinetochores ...............................68

CHAPTER II_ RESULTS

Part I: ........................................................................................................................................................73

New teashirt alleles obtained by P-element excision mutagenesis ........................................................................75

Part II: .......................................................................................................................................................91

Regulation of cell proliferation and patterning in Drosophila oogenesis by Hedgehog signalling depends on teashirt ...93

Part III:.................................................................................................................................................... 117

Teashirt and Tiptop repress each other in Terminal Filament Cells during Drosophila oogenesis............................. 119

Part IV: .................................................................................................................................................... 137

The knock-down of the teashirt gene causes a metaphase-like arrest in Drosophila larval neuroblasts and S2 cells.. 139

CHAPTER III_ DISCUSSION AND PERSPECTIVES

1- Tsh and its paralog Tio repress each other in the tip of the Drosophila ovary, and both are able to regulate En

expression.................................................................................................................................................. 173

2-Tsh regulates the expression of Hh and En at the Tip of the Drosophila germarium........................................... 174

3-Tsh mediating Hh pathway is required for FSC proliferation ........................................................................... 175

4-The metaphase/anaphase transition is compromised in the absence of Tsh...................................................... 177

5- Tsh/Hh signalling from flies to vertebrates.................................................................................................. 179

Concluding Remarks: ................................................................................................................................... 180

REFERENCE.............................................................................................................................................. 183

APPENDIX ............................................................................................................................................... 211

XII

Page 15: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

List of Figures

CHAPTER I_ INTRODUCTION

Figure I_1- Tsh expression patterning during Drosophila embryogenesis ..............................................................4

Figure I_2- Tsh as a Hox co-factor in the segment identity of the trunk................................................................7

Figure I_3- Summary of gene interactions in the cells surrounding the middle constriction ...................................12

Figure I_4- Schematic representation of Drosophila leg imaginal discs and the adult leg.......................................15

Figure I_5- Schematic representation of Drosophila wing imaginal discs and the adult wing ..................................17

Figure I_6- Embryonic expression of Tio and Tsh.............................................................................................19

Figure I_7- The Teashirt family in Drosophila and vertebrates ...........................................................................21

Figure II_1- Diagram and a Drosophila ovary and ovarioles ..............................................................................24

Figure II_2- Drosophila germarium cross section showing the locations of germ line stem cells (GSC), follicle stem

cells (FSC), and their niches............................................................................................................................26

Figure II_3- Diagrams of the identified signalling pathways and intrinsic factors that regulate GSC maintenance and

differentiation in the Drosophila ovary..............................................................................................................31

Figure II_4- Diagrams of the identified signalling pathways and intrinsic factors that regulate follicle stem cells (FSC)

and escort stem cells (ESC) maintenance and differentiation in the Drosophila ovary .............................................39

Figure II_5- The Hedgehog pathway based on what is known in Drosophila ........................................................41

Figure III_1- Cell cycle phases. ....................................................................................................................45

Figure III_2- Diagram of mitosis in animal cells ..............................................................................................47

Figure III_3-The regulation of the MPF activity by phosporilation ......................................................................48

Figure III_4- Schematic representation of the G2 checkpoint............................................................................60

Figure III_5- Relationship of the SAC with the cell-cycle machinery...................................................................62

Figure III_6- The attachment process............................................................................................................63

Figure III_7- A Model for the Regulation of APC/CCdc20 by the Spindle Checkpoint.............................................69

CHAPTER II_ RESULTS

Part I

Figure 1- Diagram of the flies matting during P-element excision mutagenesis....................................................79

Figure 2- No mitotic anomalies were detected in neuroblasts from 8 P-element excision lines L3 lethal. ..................82

Figure 3- No mitotic anomalies were detected in neuroblasts from P-element excision lines lethal during pupae stage.

...................................................................................................................................................................82

Figure 4- Adult ∆P48/tshA3-2-66 and ∆P127/tshA3-2-66 ovaries showed a very strong and early arrest in oogenesis........83

Figure 5- Amplified and sequenced regions of the PlacW in tshA3-2-66 and ∆P126. ..................................................86

Figure 6- Morphologic aspects of ∆P126/ tshA3-2-66 flies. .....................................................................................88

Part II

Figure 1- en co-localises with a tsh-subset domain in Drosophila ovaries........................................................... 102

Figure 2- Expression level variation of tsh in ∆126/tshA3-2-66 ovarioles. .............................................................. 103

Figure 3- ∆126/ tshA3-2-66 ovaries display abnormal germline cyst encapsulation. ............................................... 105

Figure 4- Cell cycle progression is affected in ∆126/tshA3-2-66 ovarioles. ............................................................. 107

Figure 5- Tsh is required for the proper expression of En and Hh in the germarium. ........................................... 109

Figure 6- Ectopic expression of hh and tsh induces similar effects. ................................................................... 110

Figure 7- Tsh rescues the imperfect proliferation phenotype of ∆126/tshA3-2-66 mutants and allows the progression of

oogenesis until stage S5/S6.......................................................................................................................... 111

Supplementary Figure 1- Rescue of the ∆126/tshA3-2-66 proliferation phenotype by ectopic Hh............................ 116

Page 16: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

List of Figures

Part III

Figure 1- Tsh and Tio co-repress each other. ................................................................................................. 124

Figure 2- Tsh and Tio rescue the imperfect proliferation phenotype of the ∆126/tshA3-2-66 mutant and allow the

progression of oogenesis until stage S5/S6..................................................................................................... 126

Figure 3- Localisation of En in wild-type, ∆126/tshA3-2-66 and tio473 germaria....................................................... 127

Figure 4- Ectopic expression of hh, tsh and tio induces a similar phenotype....................................................... 129

Supplementary Figure 1- Tsh and En in wild-type, ∆126/tshA3-2-66, tio473 and ∆126/tshA3-2-66;UAS-tio/hsGAL4

germaria. ................................................................................................................................................... 135

Part IV

Figure 1- Molecular organisation in tshNG1 and tshA3-2-66 mutants. ...................................................................... 150

Figure 2- Expression analysis of Tsh. ............................................................................................................ 151

Figure 3- Mitotic phenotypes in tsh mutant larval brains. ................................................................................ 152

Figure 4- Mitotic figures in preparations of larval CNS cells from wild-type and tsh homozygotes. ........................ 153

Figure 5- Cyclin A and B levels in wild-type and tsh mutant larval CNS cells. ..................................................... 154

Figure 6- Distribution of centromere CID and kinetochore components Bub1 and Rod in wild-type and tshNG1

neuroblast cells. .......................................................................................................................................... 156

Figure 7- Time course analysis of Tsh RNAi in Drosophila S2 Cells. ................................................................... 158

Figure 8- Abnormal mitotic figures observed in S2 TSH-RNAi cells.................................................................... 159

Supplementary Figure 1- Immunolocalisation of the Tsh protein in larval CNS cells.......................................... 166

Supplementary Figure 2- In situ cell death detection.................................................................................... 167

Supplementary Figure 3- Cuticules phenotype............................................................................................. 168

Supplementary Figure 4- Immunolocalisation of Polo in larval CNS cells ......................................................... 169

Supplementary Figure 5- Analisys of Polo expression by western blot............................................................. 169

Supplementary Figure 6- A CNS from L1, L2 and L3 larvae instars expressing tshlacZ stained with X-Gal. .......... 170

CHAPTER III_ DISCUSSION AND PERSPECTIVES

Figure 1- Tsh signalling to Follicle Stem cells (FSC) proliferation mediating Hh expression................................... 175

Figure 2- Proposed model to explain Tsh role in cell proliferation, mediating Hh expression signal in FSC and

neuroblasts................................................................................................................................................. 183

XIV

Page 17: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

List of Tables

CHAPTER II_ RESULTS

Part I

Table1- Counting of F2 male that lost P-element based in the colour of the eyes. .................................................81

Table 2- Classification of the P-element excision line based in their lethality in trans-heterozygous with tshA3-2-66. ....81

Table 3- Fertility of the seven P-element excision lines with viable escapers, in Trans with the tshA3-2-66. .................84

Table 4- Fertility of the combination of the seven P-element excision lines with viable escapers in Trans between

them............................................................................................................................................................85

Table 5- List of the tsh alleles crossed with ∆P126 and tshA3-2-66 to test for the viability and fertility of the adult female.

...................................................................................................................................................................87

Supplementary table 1- PlacW excision results obtained in two independents mutagenesis from the tshA-3-2-66 allele

by Salazar (2000, refered in chapter IV of the results of this manuscript) and Pimentel (presented in this chapter). ..90

Part II

Table 1- List of the tsh alleles crossed with P{lacW}A3-3-66∆126 and tshA3-2-66 to test for the viability and fertility of the

adult female. .............................................................................................................................................. 104

Table 2- Quantification of the ∆126/ tshA3-2-66and the ∆126/ tshA3-2-66;UAS-tsh13/hs-Gal4 ovaries phenotype. ........ 112

Supplementary Table 1- Quantification of the positive P-Histone H3 FC in the first 5 egg chambers of wild-type (n=69)

and ∆126/ tshA-3-2-66(n=98) ovaries................................................................................................................ 116

Part IV

Table 1- Mitotic index (number of mitotic cells per optic field) and percentage of anaphase figures in the larval CNS of

wild-type, tsh, rod x5, bub1, tsh rodx5 and tsh bub1 mutants........................................................................... 157

Table 2- List of the most significant gene culters altered in tshNG1 homozygous neuroblats microarrays with the

respectived P value and total number of genes include. .................................................................................... 160

Table 3- The most up- and down-regulated genes in tshNG1 homozygous neuroblats microarrays, the putative Tsh

target genes are also listed with the respective lower bound of fold change (LBFC), The positive LBFC represent the up

regulated genes while the negative LBFC represent the downregulated genes. .................................................... 161

APPENDIX

Table I- Results of the up- or down-regulation genes in tshNG1 homozygous neuroblats which have been ordered by

lower bound of fold change (LBFC), after using a cut-off of 1.3. The positive LBFC represent the up regulated genes

while the negative LBFC represent the downregulated genes. Only the genes with the absolute value of LBFC upper

than 1.3 are represented. ............................................................................................................................. 213

Table II- Results of the gene ontology clusters with a P-value <0.001 in tshNG1 homozygous neuroblats microarrays

after using a cut-off of 1.3. The positive lower bound of fold change (LBFC) represents the up regulated genes while

the negative LBFC represent the downregulated genes. Only the genes with the absolute value of LBFC upper than 1.3

are represented........................................................................................................................................... 227

Page 18: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Liste of Abbreviatures

µg microgram µl microliter

µm micrometer APC/C Anaphase promoting complex/ cyclosome

bp base pair CC Cap cells

Cdc genes Cell division cycle genes Cdk Cyclin-dependent kinase CNS Central nervous system

D. melanogaster Drosophila melanogaster DAPI 4’,6’-Diamidino-2-phenilindole DNA Desoxiribonucleic acid

dsRNA double-stranded RNA en engrailed

ESC Escort stem cells EC Escort cells

FSC Follicle stem cells FC Follicle cells

FITC Flurescein isothiocyanate GSC Germline stem cells

hh hedeghog hs heat-shock promoter

IGS Inner germarial sheath cells ihog interference hedgehog KDa Kilo-Dalton (1KDa=1000 Da) MPF Mitosis promoter factor

mRNA messenger Ribonucleic acid MT Microtubule

NEBD Nuclear envelope breakdown PBS Phosphate buffer saline pH=7.4

PBST PBS with 0,1% Triton X-100 PCR Polymerase chain reaction

PPH3 Phospho-histone H3 RNA Ribonucleic acid RNAi RNA interference SAC Spindle assemby checkpoint

SC Stem cells SDS Sodium dodecyl sulphate Ser Serine TF Terminal Filament cells

Thr Threonine tio tiptop tsh teashirt Tyr Tyrosine

UAS Upstream activator sequence X-Gal X-gal-5-bromo-4-chloro-3-indolyl-beta-D-galactopiranosideo

WT Wild-type

Page 19: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Resumo

O gene teashirt (tsh) de Drosophila codifica para uma proteína de 116 KD com três

motivos atípicos do tipo Zinc finger (Cx2Cx12HMx4H) espaçados. Este gene foi, inicialmente,

referido como sendo necessário para a correcta especificação dos segmentos do tronco,

durante a embriogénese de Drosphila, em colaboração com os genes Hox. Adicionalmente, em

Drosophila, o gene tsh foi implicado na morfogénese do intestino médio, durante o

desenvolvimento da parte proximal dos apêndices e durante a especificação dos olhos no

adulto.

Este trabalho teve como principal objectivo clarificar a função do gene tsh na

proliferação celular em Drosophila. Para tal, estudou-se o efeito da perda de função tsh em

três contextos celulares de Drosophila: (1) nos ovários, com o intuito de estudar a proliferação

das células estaminais foliculares (FSC) do germarium; (2) nos neuroblastos e (3) nas células

S2, com o fim de analisar a progressão da mitose propriamente dita. Caracterizou-se as

anomalias apresentadas em neuroblastos e ovários de mutantes hipomorficos tsh e em células

S2 defectivas para o mesmo gene através da técnica de interferência de RNA (RNAi). A análise

do padrão de expressão de cérebros mutantes para tsh foi obtida, utilizando a técnica de

microarrays.

Durante este trabalho, observou-se que o gene tsh é co-expresso com o gene

hedgehog (hh) na extremidade anterior do germário de Drosophila: nas células do filamento

terminal (TF) e nas células cap (CC), ambas são localizadas adjacentemente às células

estaminais germinais (GSC) e são descritas como parte integrante do nicho das GSC e FSC.

Nos ovários de Drosophila, a via de sinalização Hh é essencial para a auto-renovação e

proliferação das FSC. Uma nova mutação tsh, causadora da paragem precoce em oogénese

(no estado S2/S3), permitiu mostrar que a proteína Tsh regula a expressão dos genes

engrailed (en) e hh na extremidade do germário, tornando-se, desta forma, crucial para a

normal proliferação das FSC. De acordo com esta observação, as experiências de sobre

expressão do gene tsh nos ovários, originaram um fenótipo que se asssemelha ao obtido

aquando da sobre expressão do gene hh. As FSC proliferam de forma excessiva, originando

um excesso de descendência que se acumula entre as câmaras do ovo, formando as estruturas

semelhantes a stalks gigantes. Em adição, nos mutantes defectivos para o tsh, a reposição

deste gene, usando o sistema UAS/ hsGal4, permitiu recuperar parcialmente o fenótipo

associado à proliferação anormal das FSC, possibilitando a progressão da oogénese até ao

estado S5/S6. Consistentemente, a expressão do gene hh através do mesmo sistema UAS/

Gal4, também promoveu uma melhoria do fenótipo nos mutantes tsh, mas de forma menos

eficiente.

Page 20: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Resumo

Assim, conclui-se que o Tsh desempenha um papel crítico na regulação da expressão do

hh, contribuindo, desta forma, para a proliferação e especificação da identidade das FSC nos

ovários de Drosophila. Estes resultados estão de acordo com as interacções entre o gene tsh e

a via de sinalização Hh, previamente descritas em embriões de Drosophila. Infelizmente, os

resultados obtidos não permitiram concluir se o Tsh actua directamente na regulação da

expressão dos genes hh e en, ou se por outro lado, se trata de uma regulação indirecta. Mais,

continua por se clarificar, neste caso, se o gene tsh actua como um repressor ou um activador.

Recentemente, o gene tiptop (tio) foi descrito como sendo um novo membro da família

tsh em Drosophila. Tio codifica para uma proteína de 1024 amino ácidos, a qual apresenta um

quarto motivo, tipo zinc finger na região C-terminal. Durante a embriogénese, foi demonstrado

que os dois membros da família tsh em Drosophila se reprimem mutuamente. Durante a

oogénese, observou-se uma complementaridade na expressão de tsh e tio nas TF: Tio localiza-

se nas TF apicais, enquanto o Tsh é preferencialmente detectado nas TF proximais. No

entanto, a expressão de tio é ectópica em todas as celulas do TF em ovários ∆126/tshA3-2-66,

assim como a expressão de tsh é generalizasa a todas as TF células em ovários tio473

homozigotas. A expressão de hsGal4>UAS-tioFL permitiu a recuperação do fenótipo dos

ovários ∆126/tshA3-2-66, tal como foi acima mencionado para o hsGal4>UAS-tsh13. De forma

idêntica, a expressão ectópica de tio induz efeitos similares aos acima descritos para a sobre

expressão de tsh, especialmente num contexto mutante para tsh. Desta forma, tal como foi

descrito durante a embriogénese, tsh e tio reprimem-se mutuamente na extremidade apical do

germário. Sugere-se ainda que Tio desempenha uma função redundante, completamente

restituída pela presença de Tsh nos ovários tio473.

Durante este trabalho, reuniu-se várias evidências que indicam que o gene Tsh é

indispensável para a progressão em mitose. Duas mutações hipomórficas no gene tsh (tshNG1

and tshA3-2-66) causam, em neuroblastos, uma paragem em metaphase, com a maioria das

figuras mitóticas exibindo cromossomas altamente condensados e associados a fusos mitóticos

aparentemente normais e a centrossomas regulares. A análise dos esfregaços destes

neuroblastos, com proteínas específicas do ponto de controlo do fuso (como Bub1 e Rod),

revelou a normal localização destas proteínas. A imunomarcação com anticorpos contra a

proteína CID (homologa da CENP-A) excluiu a ocorrência da separação prematura de

cromatídeos irmãos. Mais ainda, nos neuroblastos parados em mitose, a ciclina A aparece

normalmente degradada, enquanto que a a ciclina B continua a ser detectada. Duplos

mutantes para tsh rod e tsh bub1 apresentam um fenótipo semelhante ao observado nos

mutantes tsh simples. Assim, as mutações rod e bub1 não conseguem ultrapassar a forte

paragem imposta pela perda de função tsh, continuando inibida a progressão para anáfase.

Um papel geral para o Tsh é desconhecido, considerando o seu padrão de expressão localizado

no sistema nervoso central (CNS) e as suas bem documentadas funções durante a

especificação de estruturas particulares. No entanto, não foi identificado nenhum tipo de

XVIII

Page 21: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Resumo

regionalização específica dos defeitos mitóticos no CNS. Além disso, o silenciamento do gene

tsh em células S2 pela técnica de RNAi provoca uma paragem em mitose, imitando o fenótipo

descrito em neuroblastos mutantes para o tsh. Durante as experiências de RNAi, a maioria das

células pararam numa configuração semelhante à metafase, o que originou um incrível

aumento no índice mitótico. Assim, estes resultados levam a propor que o gene tsh

desempenha um papel indispensável na transição metafase/anafase, uma vez que na ausência

deste gene, o ponto de controlo do fuso e/ou a actividade do complexo promotor da anafase/

ciclossoma (APC/C) são pertubados. A análise do padrão de expressão de cérebros mutantes

para tsh, comparativamente ao selvagem, não permitiu identificar um óbvio gene mitótico

alvo. Curiosamente, foi possível estabelecer uma ligação putativa com a via de sinalização Hh

através do interference–hedgehog gene (ihog), o terceiro gene com níveis de downregulation

mais significativos. IHog é uma proteína transmembranar intermediária na resposta ao sinal

activo de Hh, actuando como amplificador deste sinal upstream do receptor negativo desta via

de sinalização: Patched (Ptc). O mecanismo, pelo qual o Hh induz a proliferação, depende do

tecido em causa, mas inclui a indução e a regulação de componentes do ciclo celular como o

das ciclinas: D1, D2, B1 e E, a Cdc25 e o N-Myc. Teoricamente, a Cdc25 (homóloga do String)

pode também ser um alvo do Hh nos neuroblastos, uma vez que, de todos os alvos mitóticos

conhecidos para a via Hh, a Cdc25 é o unico que se encontra ligeiramente downregulated nos

mutantes tsh. No entanto, o sincronismo de paragem em metafase, observado nos

neuroblastos tsh, está mais de acordo com alterações nos níveis de expressão do gene polo

(um alvo mitótico com níveis de downregulation idênticos aos registados para a Cdc25). De

facto, a observação dos neuroblastos tsh faz lembrar o fenótipo mitótico apresentado por

mutantes polo (por exemplo homozigotas para o alelo polo9). No entanto, a redução dos níveis

de expressão de polo não é por si só suficiente, para explicar a forte paragem em metafase

apresentada pelos mutantes tsh e, por outro lado, o polo nunca foi implicado como sendo um

alvo da via Hh. Neste ponto, os resultados aqui expostos falham para identificar um alvo

mitótico, no entanto, os significantes níveis de downregulation registados para o iHog sugerem

uma interligação entre a forte paragem em mitose e a via de sinalização Hh, fazendo lembrar

os resultados observados durante a oogénese. Assim, propõe-se que o gene tsh é necessário

para a transcrição do iHog e que este ligando está implicado na modulação da sinalização Hh

durante a mitose.

Com o trabalho aqui apresentado, pretende-se demonstrar que o gene tsh afecta a

proliferação das FSC através da regulação da expressão do en e hh. Em adição, poder-se-á

especular que em neuroblastos e células S2, o Tsh poderá também afectar a progressão da

mitose através da sinalização Hh, uma vez que, pelo menos em neuroblastos, pela técnica dos

microarrys, verificou-se que o gene ihog é o terceiro mais donwregulated. Assim, propõe-se

uma nova função para o gene tsh na proliferação celular, através da via de sinalização Hh.

XIX

Page 22: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Resumo

Palavras Chave: teashirt; tiptop; via de signalização do hedgehog; células

estaminais somáticas; neuroblastos; mitose, ponto de controlo do fuso; transição metafase/

anafase, complexo promotor da anafase/ ciclossoma (APC/C).

XX

Page 23: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Abstract

Drosophila teashirt (tsh) encodes a zinc finger protein essential during Drosophila

development.

My PhD aimed to clarify the tsh role in cell proliferation studying tsh function in three

Drosophila systems: follicle stem cells (FSC), neuroblasts and S2 cells.

I found that Tsh and Hh were co-expressed in the Drosophila terminal filament and cap

cells. A new tsh female sterile mutation exhibiting early oogenesis arrest allowed me to show

that Tsh acts via Hh-mediated signalling pathway to control FSC proliferation; tsh

overexpression mimics the hh overexpression associated phenotype. Additionally, in tsh

background, the hsGal4 driven expression of tsh or hh partially rescues the abnormal

proliferation phenotype allowing oogenesis progression. These results suggest that Tsh is

critical to control hh expression contributing for the regulation of FSC proliferation and

specification of somatic cell identity.

In neuroblasts, hypomorphic tsh mutation leads to a metaphase-like arrest with highly

condensed chromosomes associated with apparently normal mitotic spindles and centrosomes.

Additionally, checkpoint proteins Bub1 and ROD were localised normally and CID

imunodetection excluded an eventual premature sister chromatid separation. Furthermore,

cyclin-A appears normally degraded, whereas cyclin-B remains detectable. Double tsh rod and

tsh bub1 mutants phenotypes resemble the tsh single mutant. Finally, tsh depletion in S2 cells

mimics the tsh mutant phenotype in neuroblasts. These results suggest that Tsh is critical

during mitosis progression and I propose that Tsh loss maintains active the spindle checkpoint

and/or unable the APC/C activity.

We proposed a tsh acts through Hh signalling in cell proliferation. During oogenesis, tsh

affects FSC proliferation mediating Hh expression. In neuroblasts, tsh could acts through Hh

signalling, since expression of interference hedgehog, a positive mediator of Hh signal, was

severely downregulated in microarrays-based analysis.

Key Word: teashirt; tiptop; hedgehog signalling; follicle stem cells; neuroblasts;

mitosis, spindle assembly checkpoint; metaphase-anaphase transition, anaphase-promoting

complex/ cyclosome (APC/C).

Page 24: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

XXII

Page 25: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

CHAPTER I

INTRODUCTION

Page 26: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

2

Page 27: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

I- The teashirt (tsh) gene

I_1- Characterisation of the tsh gene

The tsh gene was firstly identified by the enhancer trap technique, in a screen for β-

galactosidase activity during embryogenesis, (Fasano et al., 1991). The tsh gene is located at

the left arm of chromosome 2 in the cytogenetic region 40A5.

Two size classes of tsh transcripts were detected in Drosophila: a predominant class

consisted of transcripts of 5.4 kb, which are expressed throughout development, and a large

size class of 8.5 kb transcripts that are detected predominantly, though not exclusively, during

embryogenesis (Fasano et al., 1991). The differential splicing is indicated as the responsible,

at least in part, for these two different classes of transcript.

Tsh codes for a protein of 993 amino acids, with an estimated molecular mass of 106

KDa (Fasano et al., 1991). The tsh protein (Tsh) is characterised by the presence of three

atypical widely-spaced C2-H2 zinc finger motifs, corresponding to the consensus sequence

CX2CX3FX5(L,M)X2HMX4H, in which the two histidines are separated by five amino acids and

the size of the loop between the second C and the first H is of twelve residues in all Tsh zinc

fingers (Fasano et al., 1991). In addition, the protein contains a consensus motif (PLDLS) for

the interaction with the C terminal Binding Protein (CtBP) in a complex with Brinker (Brk) and

three domains rich in alanine, in the N-terminal (Fasano et al., 1991; Manfroid et al., 2004;

Saller et al., 2002). Finally, the acidic rich domain, identified in Tsh, is required for their

interaction with Scr (Sex combs reduced) for the identity of the prothorax (Taghli-Lamallem et

al., 2007).

These structural properties are compatible with a role on the modulation of the

chromatin. In fact, the gene Su(var)3-7, which encodes a protein with seven zinc of the Tsh

zinc finger type, play a role in the modification of the chromatin structure (Cleard and Spierer,

2001). It was shown that Tsh is able to bind to DNA behaving like a transcription factor. In

vitro, Tsh binds specifically to a fragment on the promoter of the gene modulo (mod) and

recognises two specific binding sites in this mod fragment (Alexandre et al., 1996). The mod

expression is repressed by Tsh in the trunk (Alexandre et al., 1996). Moreover, cell culture

analyses showed that tsh acts as a transcriptional repressor of mod (Waltzer et al., 2001).

Recently, Tsh was identified as a protein that interact with FE65 (Kajiwara et al., 2009). FE65

is an adapter protein that binds to the amyloid protein precursor and this complex can

modulated gene expression (Duilio et al., 1991). FE65 could simultaneously recruit SET (a

component of the inhibitor of acetyl transferase) and Tsh (that in turn recruit histone

deacetylase to produced a powerful gene silencing complex; Kajiwara et al., 2009). The

primate specific Caspase 4 (CASP 4) was identified as a target of this FE65/ SET or FE65/ Tsh

complex. In addition, chromatin immunoprecepitation showed a direct interaction between

FE65 and Teashirt3 with the promoter region of the CASP 4 (Kajiwara et al., 2009).

Page 28: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

I_2- Expression patterning of the tsh mRNA and protein in Drosophila

The tsh transcripts are first detected at the beginning of cellularisation (stage 4-5), in a

central ring of cells corresponding to the primordium of the thorax (Fig. I_1; Fasano et al.,

1991). At the end of cellularisation/ beginning of gastrulation, tsh expression is observed in

five bands transient in the central region of the embryo (the three posterior bands are weaker

then the two anterior ones). During gastrulation (stage 6), the striped pattern gives a more

uniform labelling in a domain of cells that correspond to the presumptive trunk region of the

embryo (Fasano et al., 1991). At the beginning of the gastrulation, tsh present a transient

expression in 5 bands of the embryo trunk. Than, at stage 9, tsh transcripts present a

homogeny distribution in all the presumptive domain of the trunk (to parasegments 3 to 13).

This staining is maintained until the end of the embryogenesis, with the tsh transcripts

presenting a particular segmental pattern: they are stronger expressed in the posterior region

of each trunk segments (Fasano et al., 1991).

Figure I_1- Tsh expression patterning during Drosophila embryogenesis (in <flybase.bio.indiana.edu

/reports/FBgn0003866.html>).

At stage 4-5 tsh transcripts are first dectect in the primordium of the thorax.

At stage 6 (beginning of gastrulation) tsh expression is observed transiently in five band in the central region of the

embryo; that correspond to the presumptive trunk region of the embryo.

At stage 9 (the extended germband stage), cells specific to parasegments 3 to 13 are labelled, that correspond to the

presumptive domain of the trunk. The trunk staining is maintained until the end of the embryogenesis with the tsh

transcripts stronger expressed in the posterior region of each trunk segments.

After stage 15 of embryogenesis, tsh expression is also observed in the ventral nervous cord, in the anterior part of

the pharynx, in the visceral mesoderm, in the anal opening and in stellate cells of the Mapighi tubes.

Tsh transcripts are also detected in ventral nervous cord, with a strong intensity in the

three thoracic segments, in the anterior part of the pharynx, in the visceral mesoderm and in

the anal opening, after the 15 stage of embryogenesis (Fasano et al., 1991).

More recently, other domains of tsh expressions have been reported. The tsh transcript

is also detected in stellate cells, a group of specific cells in the Mapighian tubes (Denholm et

4

Page 29: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

5

al., 2003). Tsh is also present in the proximal region of the imaginal discs of the halteres,

wings and legs (Bhojwani et al., 1997; Erkner et al., 1999).

During embryogenesis, the tsh transcript and protein present sensibly the same

distribution. The protein is detected within the cytoplasm or the nucleus during the first stages

of development. After stage 7 of embryogenesis, the localisation is predominantly in the

nucleus (Alexandre et al., 1996).

I_3- Modulators of the tsh expression during embryogenesis

tsh expression is regulated during embryogenesis by Hox genes in ectoderm and

mesoderm derivatives (Roder et al., 1992; Mathies et al., 1994). Moreover, some of these

homeodomain transcription factors, including Antennapedia (Antp), Ultrabithorax (Ubx) and

abdominal-A (abd A), bind directly to specific enhancers in the tsh regulatory region

(McCormik et al., 1995). However, Hox proteins are not required for initiation of the tsh

expression, but rather modulate and maintain the expression pattern in a segment-specific

manner. Tsh in turn regulates the expression of Hox genes active in the gnathal segment and

head: Sex combs reduced (Scr), Deformed (Dfd) and labial (lab; Roder et al., 1992). Thus, tsh

works at the same level as Hox.

spalt (sal) and grunge (gug, homologue of vertebrates atrophine genes), are known to

regulate tsh expression (Erkner et al., 2002; Röder et al., 1992). Sal protein acts like a

transcriptional repressor of tsh: in the absence of sal, tsh is expressed ectopically in

parasegment 2, in the labial and in the tail region (parasegments 14-15, stage 10-11; Röder et

al., 1992). Grunge acts positively to regulate teashirt expression in proximoventral parts of the

leg. Grunge has other regulatory functions in the leg, including the patterning of ventral parts

along the entire proximodistal axis and the proper spacing of bristles in all regions (Erkner et

al., 2002). In Gug loss-mutants, the embryo segmentation is affected via a failure in the

repression of at least four segmentation genes known to regulate tsh [hunchback (hb), Krüppel

(Kr), knirps (kni) and fushi tarazu (ftz)]; in these embryos, tsh appears in bands pattern in the

dorsal region, and the ventral tsh expression is lost (Coré et al., 1997; Erkner et al., 2002;

Röder et al., 1992).

I_4- Tsh functions during Drosophila development

Tsh is crucial for the patterning of the trunk identity in collaboration with the Hox genes

(Fasano et al., 1991; Roder et al., 1992). Tsh also acts on the Wingless (Wg) and Hedgehog

(Hh) pathways (Angelats et al., 2002; Gallet et al., 1998, 1999) for the specification of the

naked cuticule during embryogenesis. In addition, tsh function is required for the midgut

morphogenesis (Mathies et al., 1994) and for the development of adult appendages (Bessa et

al., 2002; Erkner et al., 1999; Pan and Rubin, 1998; Soanes et al., 2001; Wu and Cohen,

Page 30: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

6

2000). The molecular mechanisms underlying tsh function are poorly understood. However, in

vitro experiments have shown that, when bound to DNA, Tsh could repress transcription

(Alexandre et al., 1996; Waltzer et al., 2001; Saller et al., 2002). Tsh binds to a specific

enhancer of the modulo (mod) gene, a known target of Scr and Ubx (Graba et al., 1994). Tsh

inhibits mod expression in the epidermis of the prothorax (or T1, the first thoracic segment)

(Alexandre et al., 1996). In the midgut mesoderm, tsh is required for the transcriptional

repression of Ubx that is mediated by high levels of Wg in collaboration with the co-repressors

Brk and CtBP (Waltzer et al., 2001; Saller et al., 2002). In this case, no sequence similar to

the mod enhancer bound by Tsh has been identified. Instead, Brk binds the Ubx enhancer and

then recruits Tsh and CtBP into a ternary repressor complex (section I_4.3 of this chapter).

I_4.1- Homeotic function of Tsh: the identity of the trunk and the specification

of the prothoracic identity

The absence of any single homeotic gene results in a change in the identity of a specific

segment or segments into another type, generating a typical homeotic transformation. Trunk

morphology in the Drosophila embryo also depends on the normal function of the tsh gene.

However, tsh is different from the classical homeotic genes in at least two aspects: (1) it codes

for a zinc finger protein and (2) the analysis of the phenotype of mutations at this locus affect

the entire trunk (Roder et al., 1992).

tsh is critically required for the identity of the T1 segment and globally for segmental

identity throughout the entire trunk, whereas the “classical” homeotic genes have more

specific roles (Fig. I_2; de Zulueta et al., 1994; Fasano, 1991; Roder et al., 1992).

The specific identity of the anterior prothorax is determined by the simultaneous

activities of the tsh and Scr gene products. First, in the absence of the Hox genes from the

trunk [Scr, Antp, Ubx, abdA and abdominal B (AbdB)], each thoracic and abdominal segment

adopts a T1 identity conferred by the presence of tsh, which is active in all this region (Struhl,

1983). Moreover, in tsh mutants, the T1 segment is transformed in labial, since Scr is unable

to promote the T1 identity in the absence of tsh (de Zulueta et al., 1994; Fasano et al., 1991).

In the absence of tsh, the Scr expression is affected: Scr is still present in the labial and the

dorsal part of T1 segment, but it is ectopic in the ventral region of this T1 segment, showing

that tsh controls the posterior limit of the Scr expression in the ventral ectoderm (Fasano et

al., 1991).

Page 31: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

Figure I_2- Tsh as a Hox co-factor in the segment identity of the trunk (in Laugier, 2005 modified

from Robertson et al., 2004).

In the trunk segments, the Tsh protein repress the expression of disco and target the segments for a trunk identity.

Sal (Spalt protein) defines the boundary between head and trunk (broken line) repressing tsh expression in the

gnathal segments. In the head segments, disco is expressed and activates the acquisition of gnathal fate. Thus, the

distribution of Tsh and Disco regionalize the embryo. The combination of these proteins with Hox genes determines the

segment identity. Scr (Sex combs reduced) is expressed in the gnathal domain as well as in the trunk, and the

acquisition of different segment identities depend on the co-factor present. Dfd (Deformed) can establish the maxilar

ou mandibular identity in function of the presence of the protein cap-n-collar (Cnc; Mohler et al., 1995). Ma:

mandibular segment; Mx: maxilar segment; Lb: labial segment; T1: first thoracic segment (or prothorax) and T2:

second thoracic segment.

In contrast, the ectopic expression of tsh induces the inverse: the head-to-trunk

transformation. The labial segment is transformed in T1 and the maxilar and antennal

segments acquire a trunk identity, illustrated by the presence of naked cuticule, without clear

segmental identity (de Zulueta et al., 1994). In this genetic context, Scr expression remains

unchanged. When tsh and Scr are simultaneously ectopically expressed, the labial, maxilar and

antennal segments acquire a T1 phenotype.

Recently, there has been a new insight into the mechanism by which Tsh, in concert

with Scr, specifies the prothoracic identity (Taghli-Lamallem et al., 2007). It was shown that

Tsh interacts directly with Scr, and this interaction depends, at least in part, on the presence

of a short “acidic domain” located on the N-terminal half of Tsh. In vivo, the expression of full

length Tsh can rescue the tsh null phenotype throughout the trunk, whereas Tsh lacking the

Scr interacting domain rescues all the trunk defects except in the prothorax (Taghli-Lamallem

et al., 2007). Thus, direct interaction between tsh and Scr is indispensable to define the T1

identity (de Zulueta et al., 1994; Taghli-Lamallem et al., 2007).

7

Page 32: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

8

Since tsh activity is required for segment identity of the entire trunk region, it has

characteristics in common with the “region specific” homeotic genes spalt and fork head

(Jürgens, 1988; Jürgens and Weigel, 1988).

It has been shown that Split ends (Spen), tsh and Antp function in a combinatorial

manner to repress the development of head-like sclerites and to promote the development of

thoracic identity (Wiellette et al., 1999).

In tsh Antp double mutant embryos, the first two thoracic denticle belts are completely

absent and replaced by cuticle typical of the head skeleton (Roder et al., 1992). Conversely,

ectopic expression of Antp induces a strong transformation of T1 to T2 (Gibson and Gehring,

1988; Gibson et al., 1990). In vitro, it was recently shown that Tsh interacts directly with the

Hox protein Antp (Taghli-Lamallem et al., 2007).

If Tsh is a co-factor for Hox proteins in the trunk, how can different Hox proteins

establish different segment identities with the same co-factor (for example, Scr and Antp with

Tsh)?

One role for the Tsh–Scr and Tsh–Antp complexes could be to control specific subset of

target genes, in order to specify the identity of the T1 and T2 segments. Moreover, the

capacity of Tsh∆acid (defective for the acidic domain) to rescue the T2 defects of a tsh8 null

mutant suggests that the collaborative activity of Tsh/Scr and Tsh/Antp does not involve the

same domain of Tsh (Taghli-Lamallem et al., 2007).

I_4.2- Tsh in the segmental polarity: modulation of the Wingless pathway and

regulation of the hedgehog pathway target genes

The segment-polarity genes, which encode a diversified group of proteins including

transcription factors and components of the Hedgehog (Hh; Ingham, 1998) and Wingless (Wg)

signal transduction pathways (Klingensmith and Nusse, 1994; Van den Heuvel et al., 1989;

Willert and Nusse, 1998) are required for intrasegmental patterning (Baker, 1988; Bejsovec

and Martinez-Arias, 1991; DiNardo et al., 1988; Dougan and DiNardo, 1992; Heemskerk et

al., 1991; Martinez-Arias et al., 1988; Noordermeer et al., 1992; Perrimon, 1994).

The wg gene (Wnt in vertebrates), encodes secreted glycoproteins (Klingensmith and

Nusse, 1994; Kühl and Wedlich, 1997; Miller and Moon, 1996; Van den Heuvel et al., 1989;

Willert and Nusse, 1998). A conserved signal transduction pathway transmits the signal

promoted by Wg following its secretion (Klingensmith and Nusse, 1994; Kühl and Wedlich,

1997; Miller and Moon, 1996; Willert and Nusse, 1998). At the end of this pathway, two

proteins are responsible for Wg output: Armadillo (Arm; homologue to vertebrate β-catenin;

Kühl and Wedlich, 1997; Miller and Moon, 1996; Peifer and Wieschaus , 1990; Peifer et al.,

1991; Riggleman et al., 1990; Willert and Nusse, 1998), and the Drosophila T-cell factor

(dTcf), which is also known as Pangolin and is analogous to the vertebrate T-cell factor (Tcf) or

lymphocyte enhancer factor-1 (Lef-1; Behrens et al., 1996; Brunner et al., 1997; Huber et al.,

Page 33: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

9

1996; Riese et al., 1997; van de Wetering et al., 1997). Tcf is a member of the family of high

mobility group (HMG) DNA-binding proteins required for DNA architecture and gene regulation.

Both, Arm and dTcf, seem to be crucial for the transmission of all known Wg signalling events.

Two pools of Arm exist within cells: one at the cell membrane that is required for cell adhesion

(Orsulic and Peifer, 1996; see also section II_4.1.3.3 of this chapter), and which is

independent of the Wg signal, and the other, a cytoplasmic pool for transmission of the Wg

signal (Pai et al., 1997; Peifer et al., 1991; Riggleman et al., 1990). In the plasmic membrane,

Arm binds to E-cadherin and α-catenin in order to maintain the cellular adhesion (Willert and

Nusse, 1998). In the cytoplasm, upon reception of Wg, Arm is stabilized and recruited for the

wg transduction signalling (Peifer, 1995).

In the absence of Wg, cytoplasmic Arm is ubiquitinated and degraded by the

proteossome, thereby blocking the transmission of the Wg signal (Aberle et al., 1997). In Wg-

receiving cells, cytoplasmatic Arm is stabilised and associated with dTcf. The Arm–dTcf

complex is transported to the nucleus, where it is thought to regulate the transcription of

genes required for cellular patterning (Brunner et al., 1997; Riese et al., 1997; van de

Wetering et al., 1997).

Tsh acts like a modulator of the Wg signalling pathway, being required to maintain the

expression of the late target (like wg itself), but not the early target (like engrailed, in stages

7–10 of embryogenesis (the cell-stabilisation phase; Gallet et al., 1998). Moreover, ectopic Tsh

expression maintains the Wg expression in the gnathal labial and maxillary segments at the

12-13 stage, whereas at this stage, in a wild embryo, wg is no longer detected in these two

segments (Gallet et al., 1998; Manfroid et al., 2004). In addition, tsh mutants present a

phenotype similar to the one observed in absence of the late components of the Wg pathway

and, in fact, wg is not maintained ventrally in the trunk segments of the embryo (Gallet et al.,

1998).

Epistasis tests and in vitro interactions showed that Tsh modulates Wg signalling by a

direct interaction with the C-terminal domain of Arm (Gallet et al., 1998; 1999). The

expression pattern of both proteins is modulated in a similar way depending on Wg. In fact,

around stage 10/11, both (Tsh and Arm proteins) accumulate to a high level in the nuclei of

posterior cells, receiving Wg signal that will form the naked cuticle. By contrast, in the anterior

part of the segment, where Wg does not signal, lower levels of nuclear Tsh are detected, which

in part contribute to the patterning of denticles (Gallet et al., 1998). Very high-level of Tsh

replaces denticles with naked cuticle. Moreover, the maintenance of wg expression is

controlled by tsh in the ventral part of the trunk (Gallet et al., 1998). The intracellular

localisation of Tsh depends on phosphorylation: Tsh is hyperphosphorylated in the nucleus and

hypophosporylated in the cytoplasm. This phosphorylation and consequent nuclear

accumulation seems to depend partially on Wg signalling. Probably, the stabilization of Arm by

Wg occurs first and favours interaction between Arm and Tsh in the cytoplasm prior to the

Page 34: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

10

translocation of Tsh to the nucleus. In fact, the accumulation of Arm precedes Tsh

accumulation in nuclei (Gallet et al., 1999).

Recently the interaction between Tshz and Wnt signalling has been demonstrated in

Xenopus (Koebernick, et al., 2006). XTsh1 is not expressed in the hindbrain, but was found to

influence spatially restricted hindbrain genes. Embryos injected with XTsh1 antisense

morpholino oligonucleotides (MO) showed reduced Wnt-4 expression in brain and spinal cord.

However, ectopic expression of XTsh1 in Xenopus had no effect on Wnt-4 expression

(Koebernick, et al., 2006).

The alternate expression of wg, hh, patched (ptc) and rhomboid (rho) is responsible for

cellular identity and polarity of the embryonic segments (Alexandre et al., 1999; Gritzan et al.,

1999). Hh is required to maintain the expression of wg in a single row of cells (Ingham and

Hidalgo, 1993; Hidalgo and Ingham, 1990). The stabilisation of wg expression allows the

maintenance of hh transcription in the adjacent cell row and, thus, Hh and Wg reinforce each

others expression. In a later step, these secreted molecules will specify the cell fate choices.

Wg is directly responsible for the naked cell fate with the requirement of Tsh (as mentioned

above), whereas Hh will indirectly govern some of the denticle identities by activating rho

transcription in rows of cells posterior to the Hh-secreting cells (Sanson et al., 1999; Gritzan et

al., 1999). rho encodes a transmembrane protein implicated in EGF signal activation and it is

required for denticle diversity during embryogenesis (Szuts et al., 1997; O’Keefe et al., 1997).

In addition, to transduce the Hh signal, a third target gene expression, the Hh receptor Ptc,

must be maintained by Hh on both sides of its expression domain, thus limiting the range of

Hh action by restricting its diffusion (Chen and Struhl, 1996; Hidalgo and Ingham, 1990;

Hooper and Scott, 1989; Nakano et al.,1989).

Loss of hh function abolishes wg, rho and ptc expression and gives rise to larvae

without any naked cuticle and none denticle diversity. By contrast, hh overexpression

promotes the expansion of wg, rho and ptc expression and the larvae displays mirror image

duplication of their denticle belts (Alexandre et al., 1999, Gallet et al., 2000; Ingham, 1993;

Tabata and Kornberg, 1994).

Cubittus interruptus (Ci; belonging to the vertebrate Gli family) is the most downstream

component so far identified in the Hh pathway (reviewed by Aza-Blanc and Kornberg, 1999). It

encodes a zinc-finger transcription factor that mediates the majority of the Hh cellular

responses in the imaginal discs and embryo. In the presence of Hh, Ci is converted into an

active form (155-kDa form, full-length), while the absence of Hh promotes the cleavage of Ci

into a repressive form (75-kDa form; Alexandre et al., 1996a; Aza-Blanc et al., 1997;

Dominguez et al., 1996; Hepker et al., 1997; Méthot and Basler, 1999; Ohlmeyer and

Kalderon, 1998; Von Ohlen et al., 1997; Wang and Holmgren, 1999; see also section II_4.2.2

of this chapter). Thus, Ci is required for both activation and repression of Hh target genes.

However, Ci does not mediate all Hh functions during embryogenesis: it is only required for ptc

Page 35: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

11

regulation and for late wg maintenance, but not for early neither for rho expression (Gallet et

al., 2000).

It appears that Tsh is also an actor in Hh signalling, controlling the expression of certain

target genes in the embryonic epidermis (Gallet et al., 2000). Moreover, before stage 11, Tsh

and Ci are necessary in a redundant way, for the regulation of Wg dependent on Hh (Gallet et

al. 2000). In addition, the expression of the gene rho requires the presence of Tsh.

Different epistasis tests showed that different combinations of the three proteins (Tsh,

Ci, and Arm) are required for the specification of the naked cuticle at different positions along

the anterior–posterior (A/P) axis (Angelats et al., 2002). Biochemical analyses revealed the

presence of complexes including the proteins Tsh, Ci and Arm (Angelats et al., 2002). In vitro,

it was previously shown that Ci is able to directly interact with the hyperphosphorylated form

of Tsh, while hypophosphorylated Tsh specifically immunoprecipitates with Arm (Gallet et al.,

1999). In addition, Arm does not bind directly to Ci, at least in vitro. Therefore, it was

suggested that Arm, Tsh, and Ci form protein complexes, with Tsh interacting directly with

both Ci and Arm (Angelats et al., 2002).

Tsh takes part in the regulation of target genes of two essential pathways, responsible

for the correct polarisation patterning of the ventral part of the segments in the embryo trunk,

appearing as an essential regulator for the development of the ventral epidermis. Tsh is a good

candidate to link segmental polarity and homeotic segmental identity in the trunk, since Tsh

participates in these both aspects of embryogenesis.

I_4.3- Tsh and the midgut morphogenesis

The Drosophila midgut is formed late in embryogenesis and is a tube consisting of two

cell layers: an inner endodermal layer and an outer mesodermal layer, encasing the yolk

(Skaer, 1993). The gut becomes divided into several compartments through the constriction of

the midgut in three places and four tubes.

Drosophila midgut development is regulated by four homeotic genes expressed in the

visceral mesoderm, where two of their identified target genes encode secreted proteins (Bienz

and Tremml, 1988; Tremml and Bienz, 1989; Reuter and Scott, 1990). The Ubx gene activates

transcription of the decapentaplegic (dpp) gene, while in adjacent mesoderm cells the abd-A

gene activates transcription of the wg gene. Dpp is a secreted molecule, homologue to TGFβ

(Transforming growth factor β) familly and it is implicated in some processes of proliferation

and differentiation (Hoodless et Wrana, 1998). The homeotic genes Antp and Scr act in more

anterior midgut regions. Both target genes, dpp and wg, are transcribed in the mesoderm, but

their secreted signalling molecules move into the endoderm (van den Heuvel et al., 1989;

Panganiban et al., 1990b; Reuter et al., 1990). In the endoderm, both are required for normal

expression of the homeotic gene labial (lab; Immerglück et al., 1990; Reuter et al., 1990;

Tremml and Bienz, 1992).

Page 36: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

Figure I_3- Summary of gene interactions in the cells surrounding the middle constriction (adapted

from Mathies et al., 1994).

Ubx and abd-A are expressed in adjacent cells in parasegments parasegment 7 and parasegment 8 respectively. abd-A

represses the transcription of Ubx in the cells posterior to parasegment 7, and so no cells express both Ubx and abd-A

(Tremml and Bienz, 1989). Ubx activates expression of dpp in parasegment 7, while abd-A activates expression of wg

in parasegment 8. abd-A prevents Ubx from activating dpp in the cells posterior to parasegment 7 (Reuter et al.,

1990). wg and dpp are both required for activation of tsh in the cells overlying and posterior to the central

constriction. dpp influences tsh expression through the activation of wg.

During embryogenesis, perturbations in the tsh expression affect the formation of the

midgut. Tsh is required for the formation of the anterior and central structures of the midgut

(the 1st and 2sd midgut constrictions) in the visceral mesoderm (Fig. I_3; Mathies et al., 1994).

In this domain, tsh transcription is controled by the Hox genes Antp, Ubx and abdA, and by the

Wg and Dpp signalling pathways (Mathies et al., 1994; McCormick et al., 1995). Antp activates

tsh in anterior midgut mesoderm, while in the central midgut mesoderm Ubx, abd-A, dpp, and

wg are required for proper tsh expression (Mathies et al., 1994). During embryogenesis,

perturbations in the tsh expression affect the formation of the midgut. Tsh is required for the

formation of the anterior and central structures of the midgut (the 1st and 2sd midgut

constrictions) in the visceral mesoderm (Fig. I_3; Mathies et al., 1994). In this domain, tsh

transcription is controled by the Hox genes Antp, Ubx and abdA, and by the Wg and Dpp

signalling pathways (Mathies et al., 1994; McCormick et al., 1995). Antp activates tsh in

anterior midgut mesoderm, while in the central midgut mesoderm Ubx, abd-A, dpp, and wg

are required for proper tsh expression (Mathies et al., 1994).

In the embryonic midgut, Ubx and labial are stimulated by low levels of Wg and

repressed by high levels of Wg (Bienz 1997). It was shown, that Tsh is also required for this

transcriptional repression of Ubx. In response to high levels of Wg, Brinker (Brk) binds to the

regulatory sequences of Ubx and then recruits Tsh to form a repressor complex; after that, the

12

Page 37: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

13

two proteins can recruit the corepressor C-terminal Binding Protein (CtBP) and repress the Ubx

expression (Waltzer et al., 2001; Saller et al., 2002).

I_4.4- Tsh in the Drosophila Pax6/ So/ Eya network during eye development

The eyes of the fly arise from a larval structure called the eye-antennal imaginal disc.

tsh is also crucial for eye development, which is controlled by the ‘eye selector’ genes: the

Pax6 paralogs eyeless (ey) and twin of eyeless (toy) (Czerny et al., 1999; Gehring, 2002).

Both ey and toy have the ability to induce ectopic eye formation when ectopically expressed

during larval development (Halder et al., 1995; Czerny et al., 1999).

The eye primordium specification starts with the expression of the early retinal genes:

eyes absent (eya), sine ocullis (so) and Dachshund (Dac). Their coexpression is necessary to

lock-in the eye fate within the eye field, possibly by acting together as a transcriptional

complex (Desplan, 1997; Kenyon et al., 2003; Kumar and Moses, 2001; Pichaud et al., 2001).

Then, retinogenesis is triggered by hh and the Hh target (Dpp/Bmp4) signals that are

produced by the surrounding posterior margin cells, at the so-called ‘firing point’ (Treisman

and Heberlein, 1998). The expression of hh and hh-dependent dpp transcription, at the

posterior margin of the disc, is a key for the definition of the eye primordium, since they

activate the expression of eya and so. The eye-inducing functions of dpp, also include the

posterior repression of wg, that by they turn, could repress eye development by promoting the

alternative head-capsule fate (Dominguez and Casares, 2005).

Retinal differentiation begins in the posterior region of the eye primordium and

proceeds as a wave in a posterior-to-anterior direction (Curtiss and Mlodzik, 2000; Dominguez

and Hafen, 1997; Heberlein et al., 1993). During wave progression, undifferentiated cells are

anterior to the morphogenetic furrow (MF; an indentation of the main epithelium that mark the

border of differentiated/ undifferentiated cells), while differentiating cells are posterior to it

(Treisman and Heberlein, 1998). The progression of the MF is driven by the joint action of dpp,

expressed within the furrow, and by hh, expressed in cells posterior to the furrow. The

induction of the proneural gene atonal (ato) by hh, is the first step towards the definition of

the R8 photoreceptor, the founder neuron of the mature eye units or ommatidia (Dominguez,

1999; Dominguez and Hafen, 1997).

tsh expression starts during L2 and is restricted to one of the two epithelial layers that

compose the disc (the ME). tsh expression in L3 eye discs is very similar to that of ey/toy,

whose expression is activated anterior to the MF and repressed posterior to it (Bessa et al.,

2002; Fasano et al., 1991). The tsh territory can be further subdivided into two domains: a

domain far from the MF, in which Homotorax (hth, a transcription factor member of Meis

family homeobox gene) expression maintains cells in an undifferentiated state and represses

retinal selector gene expression (such as eya); and a domain more close to the MF, in which

hth is repressed leading to eya upregulation (Bessa et al., 2002). The latter is also known as

Page 38: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

14

the pre-proneural domain, since it precedes the onset of retinal differentiation (Greenwood and

Struhl, 1999). Note that Tsh is not expressed in the two rows of cells immediatly anterior to

the MF, which stop to divide and are synchronized to start differentiation once the MF has

moved on (Bessa et al., 2002).

tsh overexpression in the eye disc can induce ectopic eye development or block its

normal formation, depending on the Gal4-promoter used (Manfroid et al., 2004; Pan and

Rubin, 1998; Singh et al., 2004; Singh et al., 2002).

It has been postulated that Tsh is required to prevent the premature expression of

downstream transcription factors: So, Eya and Dac (Bessa et al., 2002). In addition, tsh and

ey, mutually induce each others expression in the ventral region in the antenna disc (Pan and

Rubin, 1998; Bessa et al., 2002; Singh et al., 2002). This genetic pathway seems conserved

during the evolution: the vertebrate Pax6 genes (homologue to toy and ey) can mimic the

overexpression of toy and ey promoting the formation of ectopic eyes in the Drosophila

(Quiring et al., 1994).

These asymmetric Tsh functions depend on the presence of other factors implicated

early in the dorso-ventral regionalisation of the eye. Thus, the expression of Iroquois complex

(Iro-C) and Delta (Dl) genes in the dorsal region of the discs are necessary and sufficient for

dorsal function of tsh, while ventral tsh role in the eye suppression requires Wg and Serrete

(Ser) (Singh et al., 2004).

More recently it was shown that tsh induces eye specification, at least in part, by

allowing the activation of eye specification genes by the Wg and Dpp signalling pathways

(Bessa and Casares, 2005).

I_4.5- Tsh during the development of proximal region of adult’s appendixes

The appendages of Drosophila developed from invaginations of the larval ectoderm to

form imaginal discs. In leg and wing imaginal discs the distal-most tip of the appendage is

formed by cells in the center of the disc, with peripheral tissue contributing to more proximal

structures. Proximal-distal (PD) axis must be created de novo for each appendage during

embryogenesis.

I_4.5.1- tsh promotes the identity of the proximal structures of legs

The Drosophila leg is divided in: the proximal leg segments (coxa and trochanter) and

the distal region, which forms the remaining leg segments (femur, tibia, basitarsus and tarsus;

Fig. I_4).

Page 39: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

Figure I_4- Schematic representation of Drosophila leg imaginal discs and the adult leg (Schubiger,

1968; modified from Fristrom and Fristrom, 1993).

(A) A dorsal–ventral (d-v) cross section through a third-instar leg imaginal disc, which is made up of a folded

epithelium inside the larva, attached to larval structures (black shading). The peripodial membrane (pm) overlays the

distal epithelium (arrowhead) and corresponds to the most proximal structure of the disc, which will form parts of the

future body wall. The green-shaded regions of the epithelium will give rise to the body wall, coxa, and trochanter. The

dotted line represents the focus on the P-D boundary (between trochanter and femur).

(B) Third-instar imaginal disc with an apical view (under the peripodial membrane). The arrowhead indicates the distal

tip. The proximal regions (green) derive from the ring on the periphery at the disc. The dotted line corresponds to the

anterior(a)–posterior(p) compartment boundary. The blue region represents the expression domain of wg, in the

ventral anterior sector. Wg is secreted on either side of its expression domain to organize ventral and proximal distal

(P-D) patterning.

(C) A Drosophila adult leg representing the different leg segments along the P-D axis: the proximal segments (green)

are the coxa (co) and the trochanter (tr). The distal segments are called the femur (fe), tibia (ti), and tarsus (ts). The

ventral region is shown by the blue shading. The leg is attached by the coxa to the body wall.

The leg PD axis is elaborated by Wg and Dpp, which induce the expression of

downstream transcription factors at different positions along the PD axis in the leg imaginal

discs (Lecuit and Cohen 1997; Abu-Shaar andMann 1998). The secreted signaling proteins Wg

and Dpp instruct cells to adopt distal identity. Distal leg segments are lost in wg or dpp

mutants (Diaz-Benjumea et al., 1994; Held et al., 1994). By mechanisms that are still not well

understood, high levels of both Dpp and Wg are required to activate Distal-less (Dll)

expression, while lower levels of the same two signals activate another transcription factor:

dachshund (dac) (Lecuit and Cohen 1997). Dll protein is crucial for the formation of specific

distal parts of the legs (Cohen, 1990), as loss of function gives rise to an excess of proximal

leg tissue at the expense of distal patterns. The result of the Wg/ Dpp signalling is the

formation of three domains of gene expression along the PD axis: Dll in distal cells, dac in

medial cells, and hth/ tsh in proximal cells (Lecuit and Cohen 1997; Abu-Shaar and Mann

1998).

15

Page 40: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

16

Tsh expression is restricted to a proximal ring and the peripodial membrane (Gonzalez-

Crespo and Morata, 1996) destined to make the body wall, coxa, and trochanter. Studies

making use of hypomorphic mutations (Erkner et al., 1999; Wu and Cohen, 2000) confirmed

that tsh function is required for the normal development of proximal leg segments. The

hypomorphic mutants show a reduction in the size of the proximal structures of the leg (coxa,

where lacks most of the bristles and sense organs, trochanter and femur), while in stronger

mutant combination the trochanter is no longer detectable as a discrete segment and the coxa

appears to articulate directly with the femur, with both (coxa and femur) reduced in size and

no sense organs recognised on the coxa and trochanter rudiment (Erkner et al., 1999; Wu and

Cohen, 2000).

It was shown a complex regulatory relationship between Hth and Tsh, in which Hth acts

both directly and indirectly to modulate Tsh levels and to define the distal limit of tsh

expression (Wu and Cohen, 2000). Although, hth and tsh influence each others expression, tsh

mutants produce quite different phenotypes from hth mutants in the leg. Hth limits the

proximal extent of dac expression and is required for the affinity boundary between trochanter

and femur. Tsh limits the proximal extent of Dll expression and is required for proper growth

and differentiation of proximal segments, but does not appear to have a role in PD boundary

formation (Wu and Cohen, 2000).

More recently, it was shown that Tsh plays a role, not only in proximal leg

morphogenesis, but also in the boundary formation between Tsh-expressing and -

nonexpressing cells; as hypomorphic mutants affect, not only the development of the proximal

leg parts, but also the morphology of the region just adjacent to Tsh-expressing cells (Erkner

et al., 1999;2002).

In the leg, Dll acts as a repressor of tsh in Wg active cells, which keeps the boundary

intact in ventral positions (Erkner et al., 1999). In contrast, it seems that Hth acts as tsh

activator in a cell autonomic manner (Azpiazu et Morata, 2002).

I_4.5.2- tsh promotes the identity of the proximal structures of wing and

halteres discs

In the Drosophila wing imaginal disc, different domains along the PD axis are present as

concentric regions, each with a unique genetic address. The centre cells of the disc originate

the distal region of the mature wing, while the peripheral tissues contribute to the formation of

the more proximal structures (Fig. I_5).

The tsh function is required for the proper development of the proximal structures of

the wing and halteres. In fact, the tsh loss of function mutations cause an abnormal wing

posture phenotype called aeroplane (ae): the wings present a paralysis and the halteres are

falling. This is the consequence of the the proximal ventral radius fusion of the wing hinge to

Page 41: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

the pleural wing process of the thorax (Soanes and Bell, 1999 ; Soanes et al., 2001; Soanes et

Bell, 2001).

tsh and hth are also co-expressed in all wing disc. However, tsh and hth expression, in

the early of the second and third instars, are restricted to the proximal cells in order to allow

the correct development of the distal region (Azpiazu et Morata, 2000; Wu et Cohen, 2002).

Although tsh and hth showed the same expression pattern in the wing disc, they don’t share

the same mechanisms of regulation. In the wing disc, like in the leg disc, Wg and Dpp signal

transduction also appear to be necessary to complete hth repression in the pouch.

Nevertheless, wg activity is only required for hth repression in a subset of the wing pouch

(Azpiazu and Morata, 2000). The relationship between Dpp signaling and tsh regulation

remains unclear. Dpp pathway in early-induced clones only represses tsh in the most lateral

regions of the disc (Wu and Cohen, 2002) and hypomorphic dpp mutant larvae still show some

tsh repression in the distal wing disc (Cavodeassi et al., 2002).

Figure I_5- Schematic representation of Drosophila wing imaginal discs and the adult wing (in

Laugier, 2005).

(A) Schematic representation of a wing disc. The dorsal region and the ventral wing pouch are represented by (d) and

(v), respectively, from each size of the dorsoventral boundary. (n) Represent the notum and (h) the hinge. The dotted

line indicate the limit of the (A) anterior and (P) posterior compartments.

(B) The adult wing with the indication of the region correspondent to the (n) notum and the (h) hinge.

Wg is required for the early establishment of tsh repression in distal wing disc (Ng et

al., 1996). In contrast, later, Notch-dependent expression of wg at the Dorsoventral (D/V)

boundary is dispensable for tsh repression (Wu and Cohen, 2002). Instead, the maintenance of

tsh repression requires Polycomb Group (PcG) mediated gene silencing (Zirin and Mann,

2004).

17

Page 42: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

18

Soon after tsh is repressed, distal wing disc cells activate nubbin (nub), which encodes

a POU domain transcription factor required for wing patterning and growth (Cifuentes and

Garcia-Bellido, 1997; Ng et al., 1995). nub expression is indirectly induced by vestigial (vg) in

the wing pouch, but extends more proximally into the wing hinge (Liu et al., 2000;Rodriguez

et al., 2002). tsh expression is incompatible with distal appendage development. Ectopic Tsh

expression in distal cells blocks nub activation and proximalises the adult wing (Azpiazu and

Morata, 2002; Casares and Mann, 2000;Wu and Cohen, 2002; Zirin and Mann, 2007). Thus,

Tsh acts as a repressor of wg expression in the hinge region of the wing disc (Zirin and Mann,

2004).

Recently, it was proposed the division of the wing disc in three domains, whose

boundaries restrict the pattern of cell divisions along the PD axis. These three regions are

comprised of cells that are: Nub+ and Tsh− (the Nub domain), Nub− and Tsh− (the gap

domain), and Nub− and Tsh+ (the Tsh domain; Zirin and Mann, 2007). In this model, tsh

repressed state does not define a compartment using the strict definition of this term (lack

their stringent and immobile boundaries), but the authors defend an analogy with

vein/intervein divisions in the wing disc (de Celis et al., 1995; Gonzalez- Gaitan et al., 1994),

and the rhombomere divisions in the development of the vertebrate brain (Fraser et al., 1990;

Kiecker and Lumsden, 2005).

I_5- tiptop as a tsh paralog

The tiptop (tio), a paralogue of tsh, was identified by a bioinformatics approach. In

addition to the three zinc finger domain common to tsh, tio has a supplementary zinc finger

type Cx2Cx12Hx4H domain in the C-terminal (Laugier et al., 2005). Curiously, the zinc finger

domains organisation present in tio is more closely related to the other tsh member family

than to tsh itself. The mosquito (Anopheles gambiae) and the bee (Apis mellifera), only

present a unique tsh member that codes for a protein containing four zinc fingers domains,

closer to Tio than to Tsh. These various results suggest that in the arthropods, the duplication

event responsible for the generation of the genes tsh and tio occurred within the drosophilae

and that the tsh prototypical gene was closely related to tio (Laugier et al., 2005). The best tsh

and tio sequences homology (51%) is located into the zing finger domain (Laugier et al.,

2005).

Tio is first detected at stage 10 (Campos-Ortega and Hartenstein, 1985) in two

posterior regions of the embryo: Malpighian tubule primordia and a sub-region of the hindgut

primordia (Fig. I_6A). At stage 11, Tio is found in the Malpighian tubule buds and part of the

hindgut (Fig. I_6B). In the head domain, the expression is initiated in the clypeolabrum, in two

bilaterally symmetric clusters of cells (Fig. I_6B). At stage 12, Tio expression appears in the

central nervous system (CNS) of the trunk and the epidermis (Fig. I_6C). The staining in the

hindgut is maintained throughout embryogenesis (black arrowhead).

Page 43: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

Figure I_6- Embryonic expression of Tio and Tsh (in Laugier et al., 2005).

Lateral (A, C– F), dorsal (B, H) and ventral (G) views. (A–F) Tio staining. Arrows: Malpighian tubules (MTs);

arrowhead: part of the hindgut; black asterisk: part of the clypeolabrum corresponding to future pharyngeal cells;

gray asterisk: neurons in the brain; white arrowheads: second constriction of the gut. (G, H) Tio (red) and Tsh (green)

immunostainings. Arrowhead: second constriction of the gut; arrow: central nervous system (G) and stellate cells of

the MTs (H). In all panels, the anterior is towards the left and the dorsal towards the top.

At stage 13, Tio staining is present in elongating Malpighian tubules (Fig. I_6D). The

anterior staining is detected in cells that invaginate into the stomodeum (Fig. I_6D) and by

stage 15 onwards, in cells close to the pharynx (Figs. I_6E, F). Tio is also detected in cells of

the brain, the second constriction of the gut, the trunk epidermis, the anterior segments of the

CNS (the three thoracic and the first two abdominal segments) and in the Malpighian tubules

(Figs. I_6E, F; Laugier et al., 2005). Thus, tsh and tio initiate and maintain exclusive

expression domain, however later they present common expression patterns in certain tissues.

At the functional level, the ectopic expression of tsh or tio induce similar effects: both promote

a homeotic transformation of the head into trunk, but tio is less effective than tsh to induce

19

Page 44: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

20

the naked cuticle identity. Moreover, the ectopic expression of tsh is more qualified than the

one of tio for the modelling of the trunk, because it restores the diversity of the denticules

more effectively than tio, in a tsh mutant (Laugier et al., 2005).

The simultaneous loss of tsh and tio has a stronger effect than the tsh loss alone,

showing that tio is active in absence of tsh and can partially replace the activity of tsh in the

trunk. In fact, tio and tsh repress each other: in the clypeolabrum, tio represses tsh

expression, while in the trunk, tsh functions as a negative regulator of tio (Laugier et al.,

2005). Consequently, the crucial role of tsh in the patterning of the ventral skin of the trunk is

partly masked by the ectopic expression of tio in absence of tsh (Laugier et al., 2005).

Recently, the interaction between the two paralogs was described during imaginal

development (Bessa et al., 2009; Datta et al., 2009). In the imaginal discs, tsh and tio have

coincident expression domain and in addition both genes show similar functional properties,

when expressed ectopically. Furthermore, tio is able to rescue the development of tsh

mutants, indicating that both genes are functionally equivalent during imaginal development

(Bessa et al., 2009). In addition, these genes share the ability of being able to induce ectopic

eye formation when forcibly expressed in the antennae tissue (Datta et al., 2009). Curiously,

tio appears to be a more effective inducer of retinal formation than tsh (Datta et al., 2009).

Interestingly, the transcriptional regulation of tio and tsh is linked by a negative feedback loop,

which is required to maintain a tight control on the total levels of Tio/Tsh and could explain the

apparently maintenance of a dispensable gene in Drosophila (Bessa et al.,2009).

I_6- The vertebrate tsh family

Tsh plays an essential role during the Drosophila development. As it was explained

before (sections I_4.1 to I_4.5.2 of this chapter), Tsh acts as a co-factor of Hox proteins, that

have been highly conserved during evolution, but Tsh also functions as a modulator of Wg and

Hh signallings, which are homologue to the vertebrates Wnt and Hh proteins, respectivelly.

Three Tshz-related genes, Tshz1, Tshz2 and Tshz3, have been isolated in vertebrates

(chick, mouse and human) based on sequence homology. The conservation, between

Drosophila and murine Tsh amino acid sequences is very low (35%) and essentially restricted

to the region of the three atypical zinc-finger motifs (Cx2Cx12HMx4H) and the acidic domain

towards the N terminus. The murine Tsh proteins possess in addition, two more classical

(Cx2Cx12Hx3-4H) zinc-finger motifs at their C-terminal end, which are not found in Tsh (Fig.

I_7; Manfroid et al., 2004). Thus, murine Tsh proteins are structurally more close to Tio than

to Tsh protein.

Page 45: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

Figure I_7- The Teashirt family in Drosophila and vertebrates

The two representants of teashirt family in Drosophila: Teashirt (Tsh) and Tiptop (Tio) are characterized by the

presence of 3 zinc finger motifs (blue box ZnF) in the N-terminus region and an additional Zinc finger motifs (blue box

ZnF) in the C terminus of Tio. The 3 members of Tsh family in vertebrates presents a structure more close to Tio, with

an extra Zinc fingers (3 Zinc fingers in the N-terminus and 2 Zinc fingers in the C terminus represented by the blue

box ZnF) and a acid domain (pink box HD) in the C terminus. The N-terminus is to the left and the C-terminus is to

the right. The numbers in the right correspond to the predict total amino acids number.

The expression patterns of Tshz1 and Tshz2, at different stages of mouse

embryogenesis, are reminiscent of tsh as they appear in restricted regions of the trunk, the

gut and the limbs (Caubit et al., 2000; Coré et al., 2007). Tshz3 is detected in a temporal and

spatial pattern that is similar, though not identical, to Tshz1 (Caubit et al., 2005; Caubit not

published data). These preliminary observations suggest a conserved function for tsh genes, in

the patterning of the anterior/posterior axis.

Murine tsh-related genes are in many aspects functionally equivalent to Drosophila tsh.

The ectopic expression of the three mouse Tsh proteins in Drosophila, like tsh, results in

homeotic transformation of the labial head segment into the T1 trunk segment (Manfroid et

al., 2004). Following ectopic Tshz (or tsh) expression, one or two other head segments take a

trunk identity, as they differentiate denticles and naked cuticle in place of certain head

structures. More importantly, Tshz genes efficiently (although, not completely in the T1

segment) rescue the phenotype of tsh8 null mutants in a very similar way to that described for

tsh: the segment polarity and the T1 to labial homeotic phenotypes are rescued (de Zulueta et

al., 1994; Manfroid et al., 2004). Both murine and Drosophila tsh, also affect the expression of

wg in the ventral labium at stage 11 and repress the expression of the direct tsh target gene

mod (Manfroid et al., 2004). Moreover, Tshz proteins behave as transcriptional repressor: they

21

Page 46: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

22

can reduce mod expression and they inhibit transcription in a heterologous test in mammalian

cells (Manfroid et al., 2004; Waltzer et al., 2001).

The ability of tsh and Tshz genes to induce T1 identity indicates that both collaborate

with the same genes and/or proteins that determine segment identity. The Drosophila tsh

directly interacts with the Hox gene Scr to promote the T1 identity (see section I_4.1 of this

chapter; de Zulueta et al., 1994; Röder et al., 1992; de Zulueta et al., 1994; Taghli-Lamallem

et al., 2007). The fact that, ectopic expression of Tshz1, Tshz2 or Tshz3 can transform the

labial segment into T1, provides strong evidence that murine Tshz can specify T1 identity in

cooperation with Scr. Interestingly, the analysis of the skeleton of HoxA5 (an orthologue of

Scr) mutant mice reveals that the most frequent morphological abnormality is the posterior

transformation of the last cervical vertebra (C7) into the likeness of a thoracic vertebra

(Jeannotte et al., 1993). Curiously, this position corresponds to the anterior limit of Tshz2

expression, suggesting its requirement to restrict the domain of activity of HoxA5 in the

somites and to set a morphological border. Even more, ectopic expression of HoxA5 in

Drosophila embryos activates ectopic expression of salivary gland target genes, just like its

functional homologue Scr (Zhao et al., 1993a). This ectopic expression leads also to the

transformation of the larval thoracic segments T2 and T3 toward T1 (Zhao et al., 1993a),

suggesting that, the mammalian protein not only recognizes the correct Scr target sites on

DNA, but also interacts with the proper Scr cofactors. Given that, the interaction between Tsh

and the Hox protein Scr in Drosophila seems to be conserved in vertebrates, at least in the

trunk patterning.

All together these data support that, in flies, the three putative mouse Tshz genes

behave in a similar way as the fly tsh gene (Manfroid et al., 2004) suggesting that, the results

obtained during Drosophila embryogenesis could be applicable to vertebrates embryogenesis.

In addition, the two murine genes Tshz1 and Tshz2 show regionalised patterns of expression,

in the trunk, in the developing limbs and the gut of mice, reforcing the hypothesis that

Teashirt function may have been conserved for patterning the primary axis and specification of

limb structures (Caubit et al., 2000). Thus, it seems that, at least some of the functions

promoted by tsh in Drosophila are conserved in vertebrates

The human tsh genes are localised at the 18q22.3-23 (TSHZ1), 20q13.2 (TSHZ2) and

19q13-11 (TSHZ3). In a screen to identified potential genes implicated in the colon cancer, a

partial cDNA, correspondent to TSHZ1, was isolated (Scanlan et al., 1998). Interestingly, the

human TSHZ1 (SDCCAG33) gene maps in the critical interval (18q22.3–18q23) frequently

reported with chromosomal anomalies in patients with congenital aural atresia (CAA), a rare

developmental anomaly of the ear, which is characterised by a conductive hearing loss with

variable degree (Cody et al., 1999; Core et al., 2007; Cremers et al., 1988; Keppler-Noreuil et

al., 1998; Kline et al., 1993; Strathdee et al., 1995). In addition, the inactivation of Tshz1 in

mice leads to a severe middle ear phenotype, mimicking defects observed in the CAA

syndrome in humans, suggesting that TSHZ1 is a good candidate gene for CAA syndrome

Page 47: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

I-The teashirt (tsh) gene

(Core et al., 2007). In the region of TSHZ2, many genes coding for proteins with zinc finger

motifs (for example, ZNF217) are overexpressed in the breast carcinoma (Nonet et al., 2001).

Rencently, it was demonstrated that Tshz3 plays a critical role in mammalian ureteric

development, by controlling smooth muscle (SM) differentiation (Caubit et al., 2008). Tshz3 is

expressed in mesenchymal cells during ureter development, within the SM cells and stromal

cell layers. Furthermore, homozygous Tshz3 null mutant mice display dilatation of the fetal

renal pelvis and proximal ureter and this phenotype correlates with the early, aberrant down-

regulation of genes encoding contractile SM cells markers. In fact, myocardin (Myocd)

expressed in developing wild-type ureteric SMCs, it is downregulated in Tshz3lacZ/lacZ ureters,

suggesting that Tshz3 may control SM differentiation by regulating Myocd. Thus, Tshz3 null

mutant mice have congenital hydronephrosis and hydroureter affecting the proximal section of

the ureter only. This model of ‘functional’ urinary obstruction resembles human congenital

pelvi-ureteric junction obstruction, which occurs in 0.5% gestations, suggesting that TSHZ, or

related, gene variants may contribute to this disorder (Caubit et al., 2008). The implication of

Tshz3 in urinary tract in mice could reflect also a conserved tsh function. In Drosophila it was

previously reported the presence of tsh and tio in the stellate cells, sub-population of the renal

(or Malpighian) tubules, the major excretory and osmoregulatory organs in Drosophila (Laugier

et al., 2005).

Thus, Tsh and Tsh-related are zinc finger protein implicated in the same several

processes both in Drosophila and in vertebrates.

II- Overview of Drosophila Oogenesis

During my PhD work, we identified and analysed some tsh mutants that revealed

problems during oogenesis progression, specifically in follicle cell stem cells (FSC) proliferation.

Thus, we take advantage of the Drosophila ovary to study FSC proliferation.

Drosophila oogenesis represents an excellent system to analyse regulatory intercellular

signalling, it produces ovarian follicle continually throughout adult life and it involves a high

degree of coordination between the proliferation and differentiation programs of two lineages

cells: soma and germ line (Narbonne-reveau et al., 2006).

II_1- The Drosophila reproductive system

The internal reproductive system of the adult female of Drosophila melanogaster is

comprised of the paired ovaries, the efferent genital duct system and its diverticula (King,

1970). Each ovary consists of a cluster of parallel ovarioles (Fig.II_1A). The average number of

23

Page 48: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

ovarioles is relatively constant (15-20). Each ovarioles is differentiated into an anterior,

sausage-shape germarium and a posterior vetellarium.

In the germarium take a place the consecutive mitosis to produce a cluster of 16-cell

cyst which one is enveloped by follicle cells. However, the major growth of the ovary and its

accompanying cells occurs within the vitelarium (King, 1970).

Figure II_1- Diagram and a Drosophila ovary and ovarioles (in King, 1970).

(A) A dorsal view of the internal reproductive system of a Drosophila adult female. Two ovarioles have been pulled

loose from the left ovary. Sperm are stored in the ventral seminal receptacle (which is drawn and uncoiled) and in

paired spermathecae. The uterus is drawn expanded as it would be when contains a mature egg.

(B) A Diagram of a single ovariole and its investing membranes. The nurse cells-oocyte complex is representative of

the first 6 stages of oogenesis. Within the vitellarium, sectioned egg chambers are drawn so as to show the

morphology of the nuclei of the oocyte and six of the fifteen nurse cells.

Attached to the surface of the germarium and to all developing egg chamber, there is

an acellular membrane, with 50 µm thick, called the tunica propria (Fig.II_1B). The germarium

and vitellarium reside within a tubular multinucleated epithelial sheath: the peritoneal sheath.

This sheath is a contractile structure containing numerous smooth muscle fibrils and abundant

trachaeoles. Apically, the tubular epithelial sheath tapers to a point where its lumen disappears

and, its inner surface becomes confluent with the tunica propria, surrounding the anterior tip

of the germarium. Anterior to this region, the epithelial sheath transforms into a solid

cytoplasmic cylinder which attaches the ovarioles to the inner apex of the peritoneal sheath.

Each ovarioles is a linear string of gradual maturing follicle. In the germarium, the eggs

chambers are made up in the anterior tip of each ovarioles, maturing progressively through

successive stages, as they move towards posterior part of this autonomous unit of the ovary,

24

Page 49: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

to form a polarized oocyte surrounded by a patterned epithelium of somatic follicle cells

(Zhang and Kalderon, 2000; Lin and Spradling, 1993).

II_2- Drosophila germarium offers the unique opportunity of to study three

types of stem cells and their niches

In the germarium, three types of stem cells was identified.

The apical portion of the germarium (region 1) contains 2-3 germline stem cells (GSC)

identified by clonal analysis (Wieschaus and Szabad, 1979) and laser ablation studies (Lin and

Spradling, 1993). They are marked by the presence of the spectrosome. The terminal filament

cells (TF) and the cap cells (CC), which are both none dividing somatic cells, support the GSC

niche (Fig.II_2). The CC directly contacts with GSC by adherent junctions enriched in DE-

cadherin and Armadillo (β-catenin). CC signal for stem cells (SC) maintenance, regulation of

proliferation and differentiation of the SC progeny. GSC divides asymmetrically every 20 hours

to produce a SC and a daughter cystoblast. The differentiating cystoblast undergoes four

rounds of mitosis, with incomplete cytokinesis, to generate a 16-cell cyst (Spradling, 2005).

One of the 16 cyst cells becomes the oocyte while the other 15 cells differentiate as nurse cells

(reviewed in King, 1970).

A recent study, has implicated a newly identified escort stem cells (ESC) as a part of

GSC niche (Decotto and Spradling, 2005). ESC was identified as the most anterior inner

germarial sheath (IGS) cells that directly contacts CC and surround GSC with their cellular

processes (Fig.II_2). These groups of four to six IGS cells are also capable of dividing and

generating differentiated IGS cells. Their progeny are renamed as escort cells (EC). It has

been proposed that these ESCs divide synchronously with GSC to generate differentiated EC,

which tightly wrap early germline cells and move posteriorly with the germline cells. Once

germline cysts pass through region 2a of the germarium, they lose their connections to EC and

become surrounded by the follicle cells (Decotto and Spradling, 2005; Kirilly and Xie, 2007).

In the region 2a/2b, two follicle stem cells (FSC) divides, in average every 10 hours, to

generate 16 cells that initially cover each cyst by an epithelial cell-like follicle cells and then

bud off him from the germarium to form individual egg chambers separated by 5-7stalk cells

(Spradling, 2005). The follicle cells go through approximately five rounds of mitotic division

before leaving the germarium and than more three to four rounds of mitosis (until stage 6),

before switching to the endocycle since stage S7 until stage S9, in which they become

polyploid by cycling through S phase without the intervention of cell division (Calvi et al.,

1998, Nystul and Spradling, 2010).

Previously, Clonal analysis has revealed that, the follicle cells are already divided into two

populations in the germarium. One lineage gives rise to the stalk cells and the polar cells,

which are a pair of follicle cells at each end off the egg chamber that connects to the stalk.

25

Page 50: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

Once the egg chamber is formed, both cells types are never divided (Tworoger et al., 1999;

Lopez-Schier and Johnston, 2001). The rest of the follicle cells derive from a separate lineage

and form the epithelium around the cyst (epithelial follicle cells or main-body).

Figure II_2- Drosophila germarium cross section showing the locations of germ line stem cells

(GSC), follicle stem cells (FSC), and their niches (adapted from Lie and Xie, 2005).

Two or three GSC (red cells, left) are situated in their niche, composed of cap cells (CC, green cells, left) and terminal

filament cells (TF, light blue cells, left tip), whereas their differentiated progeny, including cystoblasts (CB) and

differentiated cysts (yellow cells, middle), are surrounded by inner germarial sheath (IGS) cells recently renamed by

escort cells EC, purple cells and green cells, bottom and top). As the CB divides it becomes a 16-cell developing cyst

(DC) and it passes through region 2a, after which it will be surrounded by follicle cells (FC, orange cells on right),

generated by the follicle stem cells (FSC, red cells, bottom and top). Two or three FSC directly contact the posterior

group of inner sheath cells (green cells, bottom and top) forming their niche. The three regions of the germarium are

indicated below the diagram.

Nystul and Spradling, (2010) by examining clones induced at low frequency, shown

recently that FC type specification is independent of cell lineage. Instead, follicle cells are

multipotent prior to polar or stalk specification. This fits well with recent studies showing that

many additional cells in the germarium can be induced to take on a polar cell fate by strong

Notch signalling, while high levels of JAK-STAT signalling can induce more stalk cells (Assa-

Kunik et al. 2007). In addition, they map the specification of the first anterior and posterior

polar cells, when cysts are associated with 8-16 FC (in mid to late region 2b), what is in

agreement with previous studies (Margolis and Spradling, 1995; Besse and Pret, 2003) which

found that polar cells were first specified at the 14-cell stage.

26

Page 51: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

The two distinct populations of stem cells, FSC and GSC, work in a coordinated fashion

to efficiently produce egg chambers and share some molecular mechanisms to control self-

renewal and proliferation. First, like the GSC, the FSC divides to generate a self-renewing FSC

(which remains in the niche) and a differentiating daughter cell (which one moves away from

the niche for further proliferation and differentiation). Second, like the GSC, the FSC is also

anchored to niche cells (the posterior EC) through DE-cadherin-mediated cell adhesion (Song

and Xie, 2002).

II_3- Origin of GSC, TF and CC

The pole cells, which are equivalent to primordial germ cells of vertebrate embryos,

constitute the first distinct cell lineage established in the Drosophila embryo. Pole cells (a

group of cells set aside at the start of embryogenesis) are the precursor of the GSC and

initially are located at the posterior pole of the blastoderm embryo. At the beginning of

gastrulation, they move dorsally and are carried inside the embryo by posterior midgut

invagination. Next, pole cells migrate across the midgut to contact the embryonic mesoderm

and subsequently divide into two groups, with interact with the gonadal mesoderm to form the

primitive gonads (Starz-Gaiano and Lehmann, 2000). Each female embryonic gonad hosts 12

pole cells on average. During embryogenesis and larval stage, these cells proliferate to make

80-110 pole cells per larval ovary. At the end of the larval development, pole cells stop their

proliferation and the first sign of oogenic differentiation is observed (King, 1970; Lin and

Spradling, 1997). At the larval-pupal transition, pole cells become refractory to hybrid

dysgenesis, which suggests that larval and pupal germline cells possess different properties

(Bhat and Schedl, 1997).

The somatic cells of the gonad also proliferate during the larval stages, remaining

relatively undifferentiated until the end of the larval phase of the development, when they

ultimately subdivide the gonad into 15-20 ovarioles. At this stage, TF and a group of cells, that

at least on morphological grounds, look like CC, form the anterior structure of the germarium

(Chen et al., 2001; Godt and Laski, 1995). In close contact with these presumptive CC are two

or three germline-derived cells behaving like stem cells in mature ovaries: they possess a

spherical fusome located apically, near the boundary with the CC and they divide, so that one

of the daughter cells always remains in contact with the CC (Gonzales-Reyes, 2003).

Considering that germ cells become refractory to hybrid dysgenesis at the larval-pupal

transition, and that this coincides with the differentiation of the TF and presumably CC, it is

tempting to speculate that it is the apposition of CC and pole cells that push these last ones

into acquiring a stem cell fate. In this model, pole cells proliferate throughout larval stages to

make the pool of germline cells present in third instar larvae. Once the TF and CC of the gonad

are differentiated and organised into a niche, they induce the germline cells in contact with the

27

Page 52: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

CC to adopt a stem cell fate. The newly formed GSC will then initiate asymmetric divisions to

sustain oogenesis (Gonzales-Reyes, 2003).

II_4- Ovary stem cells and their molecular mechanisms of regulation

II_4.1- Genes and Pathways that control GSC self-renewal, proliferation and

differentiation

In the last years, it was identified a number of genes with critical functions in GSC self-

renewal, proliferation and differentiation. Some of these genes encode intrinsic factors,

including nuclear factors and translational regulators that function inside the GSC, while the

others produce proteins that function to generate signals and regulators of signal production

within the niche.

In the apical tip of the germarium, two or three GSC can be easily identified by their

localisation (directed anchored to posterior side of the CC), size (the biggest cell at the tip of

the germarium) and anterior localisation of a round spectrossome. The spectrossome and its

counterpart in the mitotic cyst (the branched fusome) are germ cell-specific organelles that are

rich in membrane skeletal protein such as α-Spectrin and Hu li tai shao (Hts). Germ cells,

including GSC, also express the Vasa protein (Lin et al., 1994; Cuevas et al., 1996).

Instead of asymmetrically deposited of intracellular factor, observed for neuronal stem

cell-like neuroblasts in embryo and larval brain, the asymmetric division of GSC is

accomplished by differential signalling resulting from an asymmetric organisation of the GSC

niche (Song et al., 2002; Jan and Jan, 2001). Normally, the GSC is anchored to CC by

adherent junctions formed between the two cell types, which are enriched in DE-cadherin and

Armadillo (more details in II_4.1.3.3). As the GSC divides to generate two daughters cells, one

remains in contact with the CC and the other one is placed one cell away from the CC. The

anterior daughter continues to receive the signals from GSC niche and self-renew as a stem

cell, while the posterior daughter differentiates into a cystoblast due to differential signalling

from their microenvironment. Following loss of one neighbouring GSC, the two stem cells

daughter’s stay in the niche, due to available niche space, and becomes GSC (symmetric

division in this particular case; Xie and Spradling, 2000). If they move away from their niche

due to the loss of adherens junctions, both daughters can differentiated into cystoblasts

resulting in the loss of GSC (Song et al., 2002).

Significant progress has been made in the past few years in gaining a better

understanding of how the niche controls GSC function by identifying signalling pathways and

genes that regulate these pathways (Fig.II_3).

28

Page 53: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

II_4.1.1- BMP, Piwi and Yb from the niche to regulate the maintenance and

division of the GSC

The GSC asymmetric cell division can be partially attributed to the extrinsics factors

emanating from the niche. Regulatory molecules: bone morphogenetic protein (BMPs) and Piwi

are produced from CC to modulate ovarian GSC maintenance and division via intracellular

signal pathway (Fig. II_3).

The pathway best studied for control GSC function is the BMP pathway, which is both

necessary and sufficient for GSC self-renewal and proliferation (Xie and Spradling, 1998; Song

et al., 2004). Two BMP ligands, Decapentaplegic (Dpp) and Glassbottomed boat (Gbb) are

expressed in CC and TF (Xie and Spradling, 1998; Song et al., 2004). They serve as a short

range signal to activate BMP signalling in GSC through type I [Thickveins (Tkv) and Sax] and

TypeII (Punt) receptors. When Dpp reaches a GSC, it interacts with its receptors. Within this

ligand-receptor complex, the constitutively actived receptor Punt phosphorylates Tkv, which in

turn phosphorylates the receptor SMAD (R-SMAD): mother against dpp (MAD). Phosphorylated

MAD (p-MAD) subsequently interacts with the common-mediator-SMAD (co-SMAD, also know

as SMAD4, with only a single member in D. melanogaster encode by the Medea gene). This

heteromeric complex (p-MAD and Medea) is translocated into the nucleus for: (I) to participate

in the activation of a BMP response gene, Daughters against dpp (Dad), primarily in the GSC

(Chen and McKearin, 2003; Kai and Spradling, 2003; Casanneuva and Ferguson, 2004; Song

et al., 2004; Affolter and Basler, 2007), and (II) to controls GSC self-renewal by directly

repressing the expression of differentiation-promoting genes such as bag of marbles (bam)

(Chen and McKearin, 2003; Song et al., 2004). Bam acts with its partner, benign gonial cell

neoplasm (BGCN; which is a large DExH box protein), to initiate differentiation of the GSC

daughters that leave the niche by an unknown mechanism (Fuller and Spradling 2007).

One of the two proteins domains of the SMAD proteins, the so called SMAD-homology

domain I, behaves as a DNA-binding domain that recognises a GC-rich motif, such sequence

elements were identified in Dpp-responsive genes (Affolter and Basler, 2007).

Mutations in dpp, gbb or downstream components lead to the GSC loss by premature

differentiation and slower division rates (Xie and Spradling, 1998; Xie and Spradling, 2000;

Song et al., 2004). Overexpression of dpp, but not gbb, completely blocks cystoblast

differentiation, resulting in proliferation of GSC-like tumors throughout the germarium (Xie and

Spradling, 1998; Song et al., 2004). These studies indicated that BMP signalling is necessary

and sufficient for GSC self-renewal. Since Gbb and Dpp use common signal transduction, it is

still to be determinate why overexpression of dpp, but not gbb, is sufficient to block germline

cell differentiation (Haerry et al., 1998; Khalsa et al, 1998). BMP signalling also have an

important role in controlling the division of primordial germ cells (PGSC) in the developing

female gonad and the recruitment of GSC to their niche (Zhu and Xie, 2003). Surprisingly,

partially differentiated 2-, 4- and 8-cells cysts are capable to revert back to GSC, when the

29

Page 54: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

niche signal dpp is provided (Kai and Spradling, 2004). How Dpp signalling can completely turn

off the active differentiation program in cystoblasts is not clear. Understanding this

phenomenon would help us to know how GSC self-renewal and differentiation is regulated, and

it may allow the regeneration of stem cells from differentiated cells, in future regenerative

medicine, if such cell fate reversal also exists in mammalian stem cells. How the short range

effects of BMP signal are controlled and whether or not there is other repressed target gene for

beyond bam; they are some major question about BMP pathway in GSC without answer for

now.

BMP and Janus kinase–signal transducers and activators of transcription (JAK-STAT)

signalling are indispensable for GSC self-renewal in Drosophila testis (Kigger et al., 2001;

Tulina and Matunis, 2001; Shivdasani and Ingham, 2002; Kawase et al., 2004). Germ cell

regulation by somatic BMP signals is probably an ancient mechanism (Gilboa and Lehmann,

2005; Li and Xie, 2005); however, bam and bgcn orthologs are unknown outside Drosophila.

The roles of Bam, Bgcn, and BMP signalling are all substantially different in males, where Bam

and Bgcn function to end rather than initiate mitotic cyst divisions. BMP signal reception is still

required to maintain GSC and repress bam transcription, but gbb appears to have a stronger

effect on GSC maintenance than dpp in testis, which is consistent with their relative expression

levels (Shivdasani and Ingham, 2002; Kawase et al., 2004; Fuller and Spradling, 2007). gbb is

expressed in cyst cells and the hub, and bam is repressed in gonialblasts as well as in GSC.

BAM misexpression can be toxic to male GSC, rather than just promoting differentiation, as it

does in females (Schulz et al., 2004). The primary task of controlling GSC self-renewal in

males is performed by a different signal, the cytokine-like ligand Unpaired (UPD) (Kigger et al.,

2001; Tulina and Matunis, 2001). UPD, expressed by hub cells, activates the JAK-STAT

pathway in GSC to specify stem cell self-renewal. When a male GSC divides, one of the

daughter retains contact with the hub maintaining the stem cell identity, while the other

daughter displaced away experiences a weaker signal and initiates differentiation (Decotto and

Spradling, 2005; Li and Xie, 2005). Therefore, JAK-STAT signalling plays an instructive role in

GSC self-renewal in the testis, similar to the Dpp/BMP signalling pathway in the ovary. JAK-

STAT pathway is also able to revert back differentiated germ cells into GSC (Brawley and

Matunis, 2004). In contrast, female GSC have no autonomous requirement for JAK-STAT

signalling (Decotto and Spradling, 2005; Li and Xie, 2005).

Like BMP, piwi and fs(I)Yb (Yb) are involved in GSC maintenance. Piwi and Yb are required in

somatic cells, such as TF and CC. Loss-of-function mutations in piwi or Yb cause rapid loss of

GSC, while Yb and piwi overexpression increases GSC-like or cystoblast-like germ cells (Lin

and Spradling, 1997; Cox et al., 1998; King and Lin, 1999; Cox et al., 2000; King et al.,

2001). Yb encodes a novel intracellular protein that regulates piwi and hedgehog (hh)

expression (King and Lin, 1999; King et al., 2001). piwi, is the founding member of the piwi

family genes coding for proteins containing conserved PAZ and Piwi domains (which bind to

30

Page 55: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

RNA), and it is involved in the regulation of microRNA (miRNA) production (Cox et al., 1998;

Cox Figure II_3- Diagrams of the identified signalling pathways and intrinsic factors that regulate GSC

maintenance and differentiation in the Drosophila ovary (in Kirilly and Xie, 2007).

Terminal Filament cells (TF)/ cap cells (CC) express Yb, Piwi and BMPs (Dpp and Gbb), which control germline stem

cell (GSC) self-renewal and/ or division in a non-cell-autonomous fashion, while the insulin pathway controls GSC

division. Also, gap junctions formed by Zpg and adherens junctions formed by E-cadherin encoded by shg controls

GSC maintenance and anchorage, respectively. The translational regulators (Pum, Nos and Pelo), the cell cycle

regulator CycB, the chromatin remodeling factor Iswi are required intrinsically in the GSC to control its self-renewal,

while the GSC-expressed Piwi and miRNA pathway components (Dcr-1 and Loqs) are required intrinsically for

controlling GSC division. In the cystoblast (CB), Bgcn, Bam, Sxl and Otu control CB differentiation.

31

Page 56: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

RNA), and it is involved in the regulation of microRNA (miRNA) production (Cox et al., 1998;

Cox et al., 2000; Grivna et al., 2006a; Grivna et al., 2006b; Kotaja et al., 2006; Megosh et al.,

2006).

Piwi-mediated pathway is absolutely essential for GSC self-renewal, while Hh signalling

play a non essential role in controlling GSC self-renewal by having redundant function with the

piwi-mediated pathway (King et al., 2001).

Recently, two studies indicated that Piwi also controls bam repression in GSC. Like it

was reported for dpp and gbb mutants, the specific removal of piwi function from GSC niche,

results in the derepression of bam in GSC. Remains unclear if piwi pathway could regulate BMP

production, secretion or activation or, for other hand, it could control the production of a new

signal that is also essential for controlling GSC self-renewal and bam repression independently

from BMP signalling (Kirilly and Xie, 2007).

II_4.1.2- External factors: Nutrition

Several external factors influence the rate of egg production such as nutrition,

humidity, availability of males and of egg laying sites (Ashburner, 1989; Bownes and Blair,

1986; King, 1970). In particular, the rate of proliferation of germline and follicle stem cells

(SC), as well as of their mitotically dividing progeny, responds to nutritional input (Drummond-

Barbosa and Spradling, 2001). In females kept on a protein-rich diet, the follicle cell

proliferation rate is 3 to 4-fold higher compared to the one of females feet with a protein-poor

diet. Follow switching from a rich to poor food source, or vice versa, the adjustment of

proliferation rates occurs within 18–24 h. This rapid and reversible change of proliferation

rates in response to nutritional input requires the insulin pathway (Fig.II_3), based on the

analysis of a female sterile mutation of the insulin receptor substrate (IRS)-like gene chico,

which is a major insulin pathway mediator (Drummond-Barbosa and Spradling, 2001). The

physiology of insulin action during the proliferative response to nutrition is poorly understood,

although female sterility results from mutations of several components of the insulin pathway

(Bohni et al., 1999; Chen et al., 1996; Montagne et al., 1999).

Insulin is not produced by cells in the germarium, instead by fat cells in the brain.

Insulin is a well-know signal for regulating sugar metabolism and is modulated by nutritional

conditions. Two recent studies demonstrated that, Drosophila α-endosulfine (dendos) gene and

the neuronal-derived Drosophila insulin-like peptides (DILPs) directly regulate GSC divisions’

rates, demonstrating that signals mediator of the ovarian response to nutritional input can

modify stem cells activity in a niche-independent manner (Drummond-Barbosa and Spradling,

2004; LaFever and Drummond-Barbosa, 2005). The GSC defective in insulin divides much

slower than wild-type ones, but insulin signals pathway only affects stem cells division and not

self-renewal in Drosophila ovary.

32

Page 57: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

II_4.1.3- Intrinsic factors in GSC maintenance and differentiation

II_4.1.3.1- Translational factor: Pumilio (Pum) , Nanos (Nos), Vasa (Vas) and

Pelota (Pelo)

Two classes of intrinsic factors control GSC: self-renewing factors or differentiation

factors (Fig.II_3).

Translational regulation by the intrinsic factors plays a critical role in GSC self-renewal.

The translational repressors Pumilio (Pum) and Nanos (Nos), are required to maintain GSC in

adult ovary as well as to prevent precocious differentiation of PGC in the developing female

gonad, by repressing the translation of mRNA that encode for differentiation factors (Lin and

Spradling, 1997; Forbes and Lehmann, 1998; Gilboa and Lehmann, 2004; Wang and Lin,

2004). They are RNA biding proteins, which form proteins complexes that repress the

translation of mRNA in Drosophila embryos (Baker et al., 1992). Mutations in both (pum and

nos), cause precocious differentiation of PGC as well as premature GSC loss (Gilboa and

Lehmann, 2004; Wang and Lin, 2004). In addition, the germ cell-specific factor, vasa (vas),

encoding a Drosophila homologue of eukaryotic initiation factor 4A, is also required for ovarian

GSC self-renewal. vas mutants germaria contain few degenerate or growth-arrested germ cells

(Styhler et al., 1998). Recently, another gene was identified for its essential role in controlling

GSC self-renewal and proliferation: pelota (pelo). pelo encodes a translation release factor-like

protein and, pelo mutants completely lose GSC in the first week after eclosion and also slow

down GSC division (Xi et al., 2005).

The identification of the mRNA target for Pum, Nos, Vas and Pelo will help to

understand how GSC self-renewal is controlled.

II_4.1.3.2- MicroRNA (miRNA) and repeat-associated small interfering RNA

(rasiRNA) is important for GSC proliferation and maintenance

One of the key characteristics of stem cells (SC) is their capacity to divide for long

periods of time in an environment where most of the cells are quiescent. Therefore, a critical

question in stem cell biology is how stem cells escape from cell division stop signals.

Recently, it was reported the necessity of the microRNA (miRNA) pathway for proper

control of GSC division in Drosophila melanogaster (Fig.II_3; Rochat et al., 1994; Zhang et al.,

2003; Hatfield et al., 2005; Clarke and Fuller, 2006). miRNAs and short interfering RNAs

(siRNAs), processed by the type III double-stranded RNase Dicer, function in an RNA-based

mechanism of gene silencing (Rochat et al., 1994; Zhang et al., 2003; Hatfield et al., 2005;

Clarke and Fuller, 2006). Most characterised miRNAs repress gene expression by blocking the

33

Page 58: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

translation of complementary messenger RNAs into protein. Experimental evidence has

suggested that small RNAs regulate stem cell character in plants and animals (Bjerknes and

Cheng, 1999; Hatfield et al., 2005; Xie et al., 2005). Moreover, some miRNAs are differentially

expressed in stem cells, suggesting a specialised role in stem cell regulation (Lin, 2002;

Hatfield et al., 2005).

MicroRNAs (miRNAs) are 21 to 23 nucleotide single stranded RNAs that are encoded in

the chromosomal DNA and repress cognate mRNA targets (Forstemann et al., 2005; Clarke

and Fuller, 2006). They are transcribed as long hairpin-containing precursors by RNA

polymerase II and processed in the nucleus by the multidomain RNase III endonuclease

Drosha (Rochat et al., 1994; Bjerknes and Cheng, 1999; Lin, 2002; Zhang et al., 2003;

Forstemann et al., 2005; Xie et al., 2005). Drosha converts pri-miRNA transcripts to precursor

miRNA (pre-miRNA); in the cytoplasm, a second RNase III endonuclease, Dicer, converts pre-

miRNA into mature miRNA (Xie and Spradling, 1998; King and Lin, 1999; King et al., 2001;

Song et al., 2004; Forstemann et al., 2005). In Drosophila, two Dicer paralogs define parallel

pathways for small RNA biogenesis. Dicer-1 (Dcr-1) liberates miRNA from pre-miRNA, whereas

Dicer-2 (Dcr-2) excises small interfering RNA (siRNA) from long double-stranded RNA (dsRNA)

(Cox et al., 1998; Cox et al., 2000; Forstemann et al., 2005).

Dcr-1 mutant GSC exhibit normal identity but are defective in cell cycle control, due to

delay in the G1 to S transition, which is dependent on the cyclin dependent kinase inhibitor

Decapo, suggesting that miRNAs are required for SC to bypass the normal G1/S checkpoint

(Hatfield et al., 2005).

The dsRBD protein Loquacious (Loqs, also known as R3D1), a paralog of R2D2, it was

identified as the partner of Dcr-1. Mutation of loqs disrupt normal pre-miRNA processing, in

adittion, loss of Loqs function in the ovary disrupts GSC maintenance, rendering loqs mutant

females sterile (Jiang et al., 2005; Forstemann et al., 2005).

It is intriguing, that two interacting protein partners (Dcr-1 and Logs), that are

essential for miRNA production, have distinct role in the control of the GSC function. One

explanation is that the dcr-1 mutations, used in the study, are not null alleles and retain

enough function to control GSC self-renewal but not enough to GSC division. Alternatively,

Loqs could signal for controlling self-renewal in the niche and for GSC division in SC, similar to

piwi (Kirilly and Xie 2007). In the future, it will be important to distinguish between these two

hypotheses and to clarify miRNAs functions produced by niche and GSC.

Piwi is known to be involved in generating 29-31 nucleotides repeat-associated small

interfering RNA (rasiRNA) for silencing retrotransposons and repeat sequences (66). In piwi

mutant GSC divides much slower than wild-type but have normal self-renewal capacity,

indicating that rasiRNA pathway is also required to control GSC division (Cox et al., 2000).

II_4.1.3.3- Junctional proteins: DE-cadherin, Armadillo (Arm) and Zero

population growth (Zpg)

34

Page 59: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

Junctional proteins are also important for GSC anchorage in the niche and for the

communication between the niche and GSC (Fig. II_3). Germline cells come into contact with

the specialised somatic cells that form the tip of the germarium during late larval development.

The juxtaposition of germline cells with future CC seems to initiate the establishment of

adherent junctions that are maintained throughout pupal development and adult life.

Cadherin-mediated adhesion, probably in concert with other factors, is necessary to recruit SC

to their developmental microenvironment. In addition, DE-cadherin accumulates at the

anterior side of GSC forming adherens junction with CC in the adult ovary, which supports a

role for this adhesion system in anchoring GSC to their niche. Mutations in Armadillo (Arm,

β−catenin) and shotgun (E-cadherin), the two key components of adherens junctions, cause

rapid GSC loss (Song et al., 2002). However, a lack of DE-cadherin or Armadillo does not seem

to compromise the identity or behaviour of SC, since the mutant SC persist for at least three

weeks after removal of DE-cadherin or Armadillo. As long as these cells remain in the

appropriate environment, even in the absence of DE-cadherin or Armadillo proteins, they

retain the self-renewal capacity and asymmetric division typical of SC. (Song et al., 2002). The

main function of adherens junctions is to keep GSC in close proximity to niche cells to receive

maximal BMP signalling for self-renewal. In Drosophila testis, adherent junctions also function

as a focal point for regulating spindle orientation of SC division, assuring the asymmetric cell

division (Yamashita et al., 2003). Cadherin also accumulates between blood SC and their

niche, suggesting that cadherin-mediated cell adhesion may be a general mechanism for

anchoring SC in their niche (Zhang et al., 2003).

In addition, gap junctions are also formed between GSC and CC. Zero population

growth (Zpg), a component of gap junctions (inexin-4) in the ovary and testis, is required to

maintain GSC and to promote germ cell differentiation (Tazuke et al., 2002; Gilboa et al.,

2003). Loss of zpg function results in partial GSC loss due to cell death and accumulation of a

few single cells. Because zpg is a gap junction components, it helps in the transport of small

molecules from supporting somatic cells to control the survival of GSC and the differentiation

of their progeny. zpg may function in parallel with bam to regulate germ cell differentiation.

II_4.1.3.4- Cell cycle regulators: Cyclin B (CycB) and Imitation switch (ISWI)

Cyclin B (Cyc B) is a G2 cyclin that acts in complexes with cyclin dependent kinase 1

(Cdk1), and it is specifically required in germ cells for promoting division of PGC and GSC (Fig.

II_3). Cyc B mutation doesn’t have obvious effect on somatic development but, severely

delays PGC proliferation and affects GSC maintenance. Curiously, the overexpression of Cyclin

A (Cyc A) and Cyclin B3 (Cyc B3), which are others G2 cyclins partners of Cdk1 (see sections

III_2.1.2 and III_2.1.3 of this chapter), can not rescue the cyc B mutant defects. However,

35

Page 60: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

remain unclear how this general cell cycle regulator specifically affect PGC and GSC self-

renewal and proliferation (Wang and Lin, 2005).

ATP-dependent chromatin remodelling factors are required to control stem cell self-

renewal. Imitation switch (ISWI) changes the conformation of the chromatin, making the DNA

accessible to transcription factors for gene activation or repression (Tsukiyama et al., 1995;

Corona et al., 1999). In Drosophila ovary, at least in part, ISWI controls GSC self-renewal

through the regulation of the responses of SC to BMP niche signals. Mutants GSC for iswi are

defective to repress bam expression and are lost rapidly (Xi and Xie, 2005).

II_4.1.3.5- Differentiation promoting factors: Bag of marbles (Bam), Benign

germ cell neoplasm (Bgcn), Orb, Sex lethal (Sxl) and ovarian tumor (Otu)

Several differentiation-promoting factors are involved in cystoblast differentiation in the

ovary; these include Bag of marbles (Bam), Benign germ cell neoplasm (Bgcn), ovarian tumor

(Otu), Orb and Sex lethal (Sxl) (Fig. II_3; Boop et al., 1993; Christerson and McKearin, 1994;

Lantz et al., 1994; McKearin and Ohlstein 1995; Lavoie et al., 1999; Ohlstein et al., 2000).

Bam and Bgcn are the best studied (McKearin and and Spradling, 1990; Gateff et al., 1996;

Lavoie et al., 1999; Ohlstein et al., 2000). Mutations in both, bam and bgcn germline cells,

abolish cystoblast differentiation leading to a cystoblast like germ cell tumor phenotype (Gateff

et al., 1996; McKearin and and Spradling, 1990; Ohlstein et al., 2000). However, bam

overexpression (but not bgcn in GSC) causes their premature differentiation and consequently

germ cell depletion (Ohlstein and McKearin, 1997). Bam acts as a differentiation-promoting

factor in cystoblasts. One of the bam function is to downregulate Dpp signaling downstream of

Dpp receptor activation. This Bam role is functionally redundant with the degradation of the

target phosphorilated Mad (a BMP specific SMAD) by action of the ubiquitin protein ligase

Smurf (Casaneuva and Ferguson, 2004).

Mutations in Orb, Sxl and Out, cause an accumulation of GSC-like or cystoblast-like

cells mixed with early differentiated cysts, suggesting that they regulate, but are not

absolutely required for cystoblast differentiation and early cyst division (Boop et al., 1993;

Christerson and McKearin, 1994; Lantz et al., 1994). Orb encodes a protein involved in the

regulation of mRNA polyadenylation (Christerson and McKearin, 1994). Sxl is a RNA biding

protein, which is known to be involved in the regulation of the mRNA splicing and translational

regulation (Christerson and McKearin, 1994; Lantz et al., 1994). Otu is also a putative mRNA

binding protein but, its function at the level of the mRNA stability, splicing or translation, is not

clear. It remains unclear whether overexpression of otu would be sufficient to cause GSC

differentiation, like bam (Kirilly and Xie, 2007).

Self-renewal and differentiation are two antagonists process for stem cell and must be

correctly balanced in a normal situation. We expected that the two mechanisms neutralise

36

Page 61: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

each other, in order to assure the normal switch between SC maintenance and differentiation

of their descendent. In fact, BMP and Piwi signal from the niche are required to repress bam

expression in GSC since, defects in BMP or Piwi signalling result in the upregulation of bam in

GSC (Szakmary et al., 2005). In other hand, Bam seems to repress BMP signal in GSC.

Removal of bam function, completely suppress the loss of GSC cause by defective BMP and

Piwi pathway. In addition, the loss of GSC phenotype, caused by bam overexpression, could be

partially suppressed by elevated BMP expression (Casaneuva and Fergunson, 2004; Kai and

Spradling, 2004). As BMP and Piwi are redundant to repress bam, this last one, also has a

redundant function with Smurf in the downregulation of the Dpp signalling (Casaneuva and

Fergunson, 2004).

However bam, doesn’t seems to be the only differentiating factor to induced cystoblast

differentiation. In pum and pelo GSC mutants, Bam is not upregulated indicating that, Pum

and Pelo normally repress the (same or different) Bam-independent pathway (Szakmary et al.,

2005; Xi et al., 2005; Chen and McKearin, 2005). Consistently, bam pum and bam pelo double

mutants are able to differentiate in germ cell cyst, while bam mutations alone arrested cells as

pre-cystoblasts (Chen and McKearin, 2005; Szakmary et al., 2005; Xi et al., 2005).

In resume, these signals could be integrated and balanced by the following model. GSC

are retained as SC because Pum:Nos complexes repress translation of a pool of mRNAs that

induce cystoblasts differentiation.

Dpp/BMP signal from the niche is received by GSC through its receptors Punt and TKV,

and it is transduced by pMad to silence bam transcription in these cells. The silencing of bam

transcription blocks the formation of Bam:Bgcn complexes and allows maintaining active the

Pum:Nos translational repression, thus maintaining the SC fate. Piwi in niche cells has an

essential and cooperative function to repress bam expression. Piwi and Dpp signalling

pathways converge at some unknown point upstream of bam, in either niche cells or GSC.

After a GSC division, the strength of Dpp signalling falls to levels that cannot longer efficiently

silence bam transcription in the cell displaced to the posterior and away from CC. This could be

due to declining Dpp levels or diminished piwi-dependent signals, which leads to the reduction

of p-Mad levels. As bam transcription increases, Bam:Bgcn complexes antagonise Pum:Nos

action and cause expression of cystoblasts-promoting mRNAs, initiating the events of

cystoblasts differentiation. Therefore, Pum/Nos can be considered as a switch between self-

renewal and differentiation, whereas niche signalling through Bam/Bgcn regulates this switch

at a single cell level (Chen and McKearin, 2005; Szakmary et al., 2005).

However, it is important continuing the search of molecular links between self-renewal

and differentiation pathway and learn more about biochemical functions of these factors, to

clarify how self-renewing factors and differentiation-promoting factors interact with each other

to control the balance between the antagonistic self-renewal and differentiation processes in

GSC.

37

Page 62: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

II_4.2- Genes and Pathways that control FSC self-renewal, proliferation and

differentiation

II_4.2.1- Molecular mechanisms shared by GSC and FSC for self-renewal and

proliferation

The continuous production of egg chambers in the Drosophila female depends on the

coordinated work of two types of stem cells: GSC and FSC, which are responsible for producing

differentiated germ cells and somatic follicle cells, respectively. Thus, these two types of SC

share some molecular mechanisms controlling self-renewal and proliferation.

Just like GSC, FSC are also anchored to their neighbouring supporting cells (posterior

escort cells, EC), through DE-cadherin-mediated cell adhesion (Fig.II_4; Song and Xie, 2002).

Such anchorage is important for maintaining FSC identity. DE-cadherin and Armadillo (Arm,

Drosophila β-catenin) accumulate in the junctions between FSC and posterior EC. Removal of

DE-cadherin from FSC results in SC loss in the adult ovary. Furthermore, the cadherin-

mediated adhesion is also important for maintaining FSC in their niches before adulthood

(Song and Xie, 2002).

As observed for GSC, when FSC divides one daughter (that retains stem cell identity)

remains in its position and the other daughter moves posteriorly to proliferate and then

generates differentiated follicle cells and stalk cells.

BMP signalling is also necessary and sufficient for promoting self-renewal and

proliferation of FSC in the Drosophila ovary (Fig.II_4). BMP signalling (by Gbb) is required in

FSC to directly control their maintenance and division, but it is dispensable for proliferation of

their differentiated progeny (Kirilly et al., 2005).

Finally, like the GSC that requires ISWI (see section II_4.1.3.4 of this chapter) for its

self-renewal, the FSC requires Domino (Dom; Xi and Xie, 2005). Dom is a member of

SWI2/SNF2 family of DNA-dependent ATPases, which function as a transcriptional repressor by

interfering with the chromatin structure (Ruhf et l., 2001). FSC mutants for dom, are lost

rapidly due to precocious differentiation, but not apoptosis (Fig.II_4; Xi and Xie, 2005).

II_4.2.2- Hedgehog (Hh) and Wingless (Wg), two critical signals for FSC

maintenance and proliferation

Despite the similarities in mechanisms shared by GSC and FSC, there are significant

differences between these two SC type. The organization of the FSC niche is quite different

from that of the GSC niche. The GSC niche is composed by the neighbouring somatic cells (TF,

CC and ESC).

38

Page 63: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

Figure II_4- Diagrams of the identified signalling pathways and intrinsic factors that regulate

follicle stem cells (FSC) and escort stem cells (ESC) maintenance and differentiation in the Drosophila

ovary (adapted from Kirilly and Xie, 2007).

Wg and Hh produced by terminal filament cells (TF)/ cap cells (CC) and Gbb produced by posterior escort cells (EC)

control FSC maintenance, while the Jak-Stat pathway controls ESC maintenance. E-cadherin-mediated cell adhesion is

required for anchoring FSC, while the chromatin remodeling factor, Dom, is required for controlling FSC maintenance.

39

Page 64: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

from that of the GSC niche. The GSC niche is composed by the neighbouring somatic cells (TF,

CC and ESC). In contrast, beyond the posterior EC, located adjacently to the FSC, the TF and

CC, located a few cells away, are also integrant part of the FSC niche.

II_4.2.2.1- Wingless (Wg) signalling is essential for FSC maintenance and

proliferation

Wingless (Wg) is essential for FSC, but is dispensable for GSC (Fig.II_4). Wg is involved

in regulation of many different cellular processes during Drosophila development. Wg signal is

transduced by Frizzled receptors, Frizzled (Fz) and Frizzled 2 (Fz2), resulting in the

phosphorilation of the cytoplasmic protein Dishevelled (Dsh), inhibition of Shaggy (Sgg) and

Axin (Axn), stabilization of Arm and activation of target genes expression (reviewed by Peifer

and Polakis, 2000).

In Drosophila ovary, Wg protein is expressed in TF and CC and functions as a long-

range signal to directly act on the FSC controlling its self-renewal. Disruption of Wg signalling

in FSC abolishes SC self-renewal (Song and Xie, 2003). Hyperactive Wg signalling, resulting

from removal of negative regulators, such as Axin and sgg, causes over-proliferation and

improper differentiation of follicle cells, and intriguingly also destabilizes FSC (Song and Xie,

2003; Kirilly et al., 2005).

II_4.2.2.2- Hedgehog (Hh) signalling is essential for FSC maintenance and

proliferation

Hh signalling is essential for self-renewal and proliferation of the FSC, while it plays a

non-essential role in maintaining the GSC by having redundant function with the piwi–

mediated pathway (Fig.II_4; King et al., 2001). Hh, is produced by TF and CC, and also

functions as a long-range signal to control FSC self-renewal and proliferation and for budding

of egg chambers (Forbes et al., 1996; King et al., 2001; Zhang and Kalderon, 2001).

Hh signalling contributes to pattern formation and regulates proliferation in a number of

settings in Drosophila and other animals (Ingham, 1998). The Hh signalling pathway has been

well characterised, and the same pathway is employed in many different tissues (Murone et

al., 1999). Upon binding of Hh to its receptor, Patched (Ptc), this last one derepresses a

second 7-transmembrane domain protein, Smoothened (Smo) and ultimately leading to the

activation of Cubitus interruptus (Ci) (Fig.II_5). Ci is a transcription factor that transmits the

Hh signal from the cytoplasm to the nucleus, where it is primarily responsible for the activation

and repression of Hh target genes (Aza- Blanc and Kornberg, 1999; Ingham, 1998). Ci exists

in at least two forms. In the absence of the Hh signal, Ci is proteolytically cleaved to a 75 kDa

repressor form. In the presence of Hh, this cleavage is prevented, and Ci functions instead as

40

Page 65: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

a transcriptional activator. Consistent with this model, effects of Hh signalling are observed

only in the presence of Ci (Methot and Basler, 2001). The regulation of Ci is complex and

involves the function of positive, as well as negative regulators. The negative regulators

required for repression of this pathway include the transmembrane protein Ptc (Hooper and

Scott, 1989; Nakano et al., 1989), protein kinase A (PKA) (Lepage et al., 1995; Li et al., 1995;

Pan and Rubin, 1995), and a kinesin-related protein, Costal2 (COS2; COS – FlyBase; Robbins

et al., 1997; Sisson et al., 1997). Loss of either ptc, cos2 or Pka stimulates intracellular Hh

signalling, even in the absence of Hh.

Figure II_5- The Hedgehog pathway based on what is known in Drosophila (in Wicking et al., 1999).

In the absence of hedgehog (hh), the patched protein (ptc) inhibits signalling by smoothened (smo). Full-length

Cubitus interruptus (Ci) forms a complex with Fused (Fu), Costal 2 (Cos-2) and Supressor of fused [Su(fu)], via which

it associates with microtubules. This association leads to targeting of Ci to the proteasome where it is cleaved to

release the transcriptional repressing form Ci75. Upon binding of hedgehog to ptc, the inhibitory effect on Smo is

suppressed leading to the dissociation of the Fu–Cos-2–Ci complex from microtubules and blocking the cleavage of

Ci155. This results in the release and stabilization of Ci, which translocate to the nucleus, initiates Hedgehog target

gene expression. In vertebrates the role of ci is achieved by complex interactions involving three Gli genes (Gli1, Gli2

and Gli3).

(Hh) hedgehog, (Ptc) patched, (Smo) smoothened, (Fu) fused, [Su(Fu)] suppressor of fused, (cos-2) costal-2, (Ci)

Cubitus interruptus.

41

Page 66: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

In Drosophila ovary, hyperactivation of hh signalling causes the overproduction of

somatic follicle cells, increases the number of FSC (Forbes et al., 1996a; Forbes et al., 1996b),

mis-position the oocyte within egg chambers and generates ectopic polar cells (Forbes et al.,

1996a; Forbes et al., 1996b Liu and Montell, 1999; zhang & kalderon). The mechanism by

which overexpression of Hh signalling promotes ectopic polar cell fate is not known. Ectopic Hh

signalling is able to transform other follicle cells into ectopic polar cells through the

suppression of eye absent (eya) expression (Bai and Montell, 2002). Disruption of the Hh

signalling cascade results in rapid FSC loss (King et al., 2001; Zhang and Kalderon, 2001).

This gives rise to compound egg chambers that contain multiple germline cysts in a single

follicle epithelium. It as therefore been postulated, that Hh signalling regulates follicle cells

proliferation at an early stage in the germarium and may also regulated oocyte positioning,

perhaps via effects on follicle cell-type determination.

The differentiated somatic follicle cell types have similar characteristics to epithelial

cells in mammals, such as basal-lateral and basal-apical polarities. Interestingly, in mammals,

Wnt and Shh has been implicated in the regulation of epithelial stem cell/precursor, cell

maintenance and proliferation in the intestine and airway (He et al., 2004; Korinek et al.,

1998; Watkins et al., 2003).In addition, in mice and humans, hyperactivation of the shh

pathway, either by mutations or overexpression, causes over-proliferation of skin cells,

resulting in the formation of skin tumors (Johnson et al., 1996; Fan et al., 1997; Oro et al.,

1997). These studies, along with those from Drosophila, suggest that the molecular

mechanisms regulating epithelial stem cells are likely conserved from Drosophila to humans.

Thus, FSC in the Drosophila ovary could represent an effective system to study epithelial stem

cell self-renewal, proliferation, and differentiation (Song and Xie, 2002).

Interestingly, in germarium, where all germline cells were removed, EC enter in

apoptosis and are lost. In this situation, FSC moves to anterior and occup the empty niche

continuing their SC prolifereation due to the presence of Hh, Wg and BMP signal in adjacent TF

and CC (Margolis and Spradling, 1995; Xie and Spradling, 2000; Kai and Spradling, 2003).

Some studies in the last few years have been improving our current understanding and

knowledge about regulation of FSC self-renewal, maintenance and proliferation, although this

is only the beginning and many questions persist. How BMP, Hh and Wg are integrating in FSC

to regulate expression of target genes that are important for FSC self-renewal and

proliferation? Although dom is ubiquitous present in the germarium, it is only required in FSC

for their self-renewal. Can dom helps to regulate the expression of target genes of the BMP,

Hh and Wg signalling pathways (Xi and Xie, 2005)?

The answers to these and other questions will provide novel insight into how SC self-

renewal and differentiation are controlled in general.

42

Page 67: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

II_4.3- Genes and Pathways controlling the maintenance of ESC and EC

Recently, a new type of somatic stem cell was identified in the Drosophila germarium. A

the escort cells are maintained by 4-6 anteriorly located escort stem cells (ESC) that

extensively contact GSC within the niche. ESC strongly resembles the cyst progenitor cells of

the testis in morphology, location and behaviour (Decotto and Spradling, 2005).

It has been proposed, that these ESC divides synchronously with GSC to generate

differentiated escort cells, which tightly wrap early germline cells and move posteriorly with

the germ cell, until they are lost by apoptosis and replaced by follicle cells in the 2a/2b

boundary(Decotto and Spradling, 2005).

In Drosophila, JAK/STAT signalling initiates when the ligand unpaired (upd; or its close

relatives) binds to the domeless receptor and activates hopscotch (the Janus kinase; JAK). In

turn, the activated JAK kinase phosphorylates the Stat92E transcriptional activator factor that

translocates into the nucleus (reviewed in Rawlings et al., 2004). Mutations in stat cause the

degeneration of the germarium probably due to loss of escort cells, while overexpression of

upd dramatically expands the escort cells population. Therefore, JAK/STAT signalling controls

the maintenance and the proliferation of ESC (Fig.II_4; Decotto and Spradling, 2005). In

germarium, where escort cells are mutants for stat, GSC are also lost indicating that ESC

participates of the GSC niche (Decotto and Spradling, 2005).

There are 40-50 EC in the germarium (Xie and Spradling, 2000; Decotto and Spradling,

2005). Like escort cells, also IGS cells have protuberances that contact germline cysts.

Germline cyst fail to differentiated in mutants where the production of these protuberances are

affected, like in mutation in stem cell tumor (stet), which is a rhomboid-like membrane

protease that activates Egfr ligands in the signalling cell. In stet mutant ovaries, the cellular

processes of IGS cells are severely reduced, and spectrosome-containing single germ cells

accumulate, suggesting that stet is required for cystoblast differentiation. Supporting a role of

EGF signalling in EC, the activated MAP kinase accumulates in wild-type EC, but is reduced in

stet mutant EC (Schultz et al., 2002).

In the future, it will be important to demonstrate whether EGFR signalling is required

for the production or maintenance of the cellular processes of the escort cells, and also clarifies

how escort control germ cell differentiation.

II_5- Drosophila ovary as a model system in stem cells studies.

The extreme conservation of the relation between SC and their niche led the

investigators to confidently use Drosophila as a model system to study SC regulation. In

addition, the molecular mechanisms regulators of the epithelial SC are likely conserved from

Drosophila to humans (Song and Xie, 2002). Thus, Drosophila ovary offers several unique

advantages to study molecular and genetic networks that control SC regulation.

43

Page 68: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Oogenesis

First, the Drosophila ovary offers the unique opportunities for simultaneously study

three types of SC and respective niches: GSC, FSC and ESC (Decotto and Spradling, 2005;

Kirilly and Xie, 2007).

Second, Drosophila ovary is a relatively simple and well-characterized structure; in

addition, the linear arrangement between SC and their progeny simplifies qualitative and

quantitative analyses (Lin, 2002; Gilboa and Lehmann, 2004; Xie et al., 2005). All the three

SC types are present in small number (2-3 GSC, 2-3 FSC and 4-6 ESC) and they are located in

a simple tubular structure, also known as the germarium, in which SC and their surrounding

cells are well defined and easily distinguishable from one to another, specially GSC based on

their morphology and size (Lin, 2002; Xie et al., 2005).

Third, powerful genetic and molecular tools are available to manipulate easily and

precise gene function in SC and their niche (Xie et al., 2005).

Fourth, more than 50% of the Drosophila genes mutations are available. In addition,

microarray and proteomic reagents are available due to the finished Drosophila genome, which

dramatically enhance our ability to discover genes that encode intrinsic factors and niche

signals, important for controlling self-renewal, proliferation and differentiation of SC.

Finally, comparative studies on the SC in the Drosophila ovary and in the testis, help to

find general SC regulatory mechanisms, because the Drosophila testis also contains easily

identified SC and niche cells (Wong et al., 2005). Therefore, the Drosophila ovary represents

one of the best systems for studying basic SC biology.

III- Overview of Drosophila Cell Cycle

My PhD work started with the challenge of clarifying why some tsh mutants cause late

larval lethality with neuroblasts arrested in a metaphase-like state. To understand better the

results presented in chapter IV, we will make a brief overview about the phases of the cell

cycle, specially the progression during mitosis and the regulation of their critical steps.

III_1- The Cell Cycle

The cell cycle is an ordered and sequential process by which cells duplicate, grow, and

prepare themselves to divide again (Fig. III_1).

The cell cycle was divided in two steps, based in microscopes studies: the phase M,

and the interphase. Many morphologic changes are observed in the cells and culminate with

the cell division during M phase. In somatic cells the period of division is called mitosis or M-

phase, while in germline cells the cell cycle ends in meiosis (the process that produces

44

Page 69: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

II-Overview of Drosophila Cell Cycle

gametes with haploid nuclei). The interphase is the period between two M phase, in which the

cell improves their size and mass. Studies in cellular biology and biochemistry led to the

classification of the interphase in three distinct steps: G1, S phase and G2. The DNA

replication, important to assure the distribution of equal genetic material (DNA) to the two

daughter cells, occurs during S (synthesis) phase. G1 and G2 are gaps where cells prepare to

S phase and M phase, respectively (Fig. III_1).

Figure III_1- Cell cycle phases.

After a cell division (M-phase) a new cell cycle starts at G1 phase. During G1 cell gets prepared to duplicate its DNA

contents in S phase (synthesis). In G2, cells prepare the machinery needed to divide in the next M-phase. Some cells

(like fibroblasts) leave the cell cycle after mitosis and just before S-phase, being able to remain in this G0 stage for

long periods. Key control points (checkpoints signalised by the red star), ensure the correct order of these events.

The cell cycle has been the subject of intense research. Rudolf Virchow set forth the

first cell theory, in which he proposed that the cell was the basic unit of all living organisms.

We now know that this theory is true; cells make up all living things, whether they are plants,

animals, or microorganisms. In the late 1970s and 1980s, the molecular biology together with

the already achieved development in biochemistry and genetics allowed the cell biologists to

better understand the working of a cell at the molecular level.

III_2- M-phase

M-phase can be divided in Mitosis and Cytokinesis (Nigg, 2001). The word mitosis came

from the Greek mitos that means thread. It was originally used by Walter Flemming in the

early 1880s and it was based on the shape of the chromosomes during cell division (Flemming,

1965).

45

Page 70: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

The main purpose of mitosis is to segregate sister chromatids into two daughters cells,

ensuring that each daughter cell will receive a complete set of chromosomes. Other organelles,

such as the centrosomes are also segregate at this time. Cytokinesis is the process that

promotes the physical separation of the two daughter cells and occurs just after last mitotic

phase, the telophase.

Mitosis has been divided in five distinct phases, in accord with the changes observed in

the structure and behaviour of the spindle and the chromosomes (Fig. III_2):

Prophase: at the beginning of mitosis, cells start to condense interphase chromatin

into well defined mitotic chromosomes. At the same time, the already duplicated centrosomes

move apart, defining the poles of the future mitotic spindle and the nuclear envelope breaks

down (NEBD). Centrosomes nucleate highly dynamic microtubules (MT)

Prometaphase: the highly dynamics microtubules are captured by kinetochores

(proteinaceous structures that are associated with the centromere DNA of the mitotic

chromosomes). The attachment of microtubules with kinetochores will become stable if the

chromosomes are bipolar attached (attached to both poles) otherwise, the monopolar

attachment (the attachment to only one pole) is unstable.

Metaphase: Bipolar attached chromosomes will congress to the equatorial region of

the cell to form the metaphase plate. Chromosomes will oscilate in the metaphase plate, a

dance that finishes when all are under tension.

Anaphase: Loss of sister chromatids cohesion triggers the anaphase onset and the

sister chromatids are pulled apart to opposite poles (anaphase A). The poles also move apart

leading to the elongation of the spindle and cell (anaphase B).

Telophase: After reaching the poles, chromatin decondenses and nuclear envelop

reforms. The formation of an actomyosin contractile ring in the equatorial region of the cells

leads to the invagination of the membrane, originating the cleavage furrow that separates the

two daughters cells.

Cytokinesis begins with the formation of a MT-based midbody near the original spindle

equator.

M-phase has to be tightly regulate to ensure that one event just take place after the

end of the previous one in a concerned fashion. Like other cellular process, M-phase is mainly

regulated by two distinct post-translational mechanisms: protein phosphorylation/

dephosphorylation and proteolysis. In fact, mitotic kinases are the major regulators of cell

division, where cyclin dependent Kinase 1 (Cdk1), a member of cyclin dependent kinase

family, is the most prominent.

46

Page 71: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

Figure III_2- Diagram of mitosis in animal cells (in <eapbiofield.wikispaces.com>).

During interphase the DNA is doubled in preparation for cell division. According with structure and behaviour of the

chromosomes and spindle, Mitosis has been divided in five consecutive phases: Profase, Promethaphase, Methapase,

Anaphase (with two sub-phase Aand B) and Telophase. In Prophase, the nuclear envelope breaks down and a spindle

forms between two centrioles. In prometaphase the chromosomes attach to the spindle fibers and then will undergo

movements until all chromosomes to be aligned in the equator of the cell (the metaphase plate). In anaphase A the

sister chromatids start to be separated and then, the poles also move apart leading to the elongation of the cell

(anaphase B). In telophase, the chromosomes reached the poles and the chromatin decondense and nuclear envelop

reforms. After the end of the mitosis, the cytoplasmic content of the mother cells is separated by cytokinesis and the

two daughters cells are formed.

47

Page 72: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

III_2.1- Mitosis Promoting Factor (MPF) and Mitosis entry

Progression through mitosis is governed by activation and inactivation of M-phase

promoting factor (MPF), a complex comprising the kinase CdK1 and a cyclin B regulatory

subunit (Fig. III_3).

Figure III_3-The regulation of the MPF activity by phosporilation (adaptated from Ohi and Gould,

1999).

Activatction of Cdc2-Cyclin B by Cdc25-mediated Thr14/ Tyr15 desphosphorylation of Cdc2 is promoted by activation

of Cdc25 by Polo. The Plkk is involved in the activation of Polo. The cdc2-Cyclin B complex is inactivated by the

phosphorylation of Thr14 and Tyr15 by the kinases Myt1 and Wee1, respectively.

The first evidence for the existence of a factor capable of inducing mitosis was

described in the 1970s. Some embryologists observed that the transfer of the cytoplasm from

a mature oocyte to a recipient immature oocyte could induce oocyte maturation even without

hormonal stimulation or protein synthesis (Mausiand Market, 1971; Smith and Ecker, 1971;

Kishimoto and Kanatani, 1976). This unidentified cytoplasmic component, responsible for

meiotic maturation, was called Maturation Promoting Factor (MPF). Around 1980, MPF was

renamed M-Promoting Factor, after their activity being also identified in mitotic cells (see

Khishimoto et al., 1988 for review).

MPF purifications revealed that it consisted of two subunits, one was identified as a

protein kinase with homology to Cdc2 (Labbe et al., 1988). The other subunit was found to be

a mitotic cyclin, the cyclin B protein (Lohka et al., 1988). This protein is called cyclin because

48

Page 73: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

embryos synthesized and destroyed it in a cyclic and synchronized way with cell cycle division

(Evans et al., 1983). At the ends of 1980s it was showed that the MPF is a universal regulator

of the M-phase onset (Nurse, 1990).

III_2.1.1- CdK1 family

The Cdc2/Cdc28 proteins are members of the conserved family of ser/thr protein

kinases; know as Cyclin-dependent kinases (Cdk). These kinases are inactive as monomers

and need to bind to a Cyclin partner in order to become active.

Cdk1 is involved in many stages of the cell cycle, including mitosis (for review see

Porter and Donoghue, 2003). Cdk1 can bind cyclin A or cyclin B, depending on the stage of the

cell cycle. Cdk1 binding to cyclin A controls the entry and progression through the G2 phase,

whereas Cdk1-cyclin B1 complex regulates the G2-M transition. Once bounded and activated in

the nucleus, Cdk1/ cyclin B1 phosphorylates multiple targets that initiate mitotic entrance,

regulates its progression, and controls mitotic exit, at which point cyclin B1 degradation is

necessary (Schmit and Ahmad, 2007).

Cdk1 (Cdc2 in Drosophila) protein level is stable throughout the cell cycle; therefore,

Cdk1 activity is controlled directly by phosphorylation and indirectly through regulation of its

cyclin binding partners. Inactivation of Cdk1 is maintained by phosphorylation of Thr14 and

Tyr15 by the kinases Myt1 and Wee1, respectively (Porter and Donoghue, 2003). These

residues reside in the ATP-binding site of Cdk1; thus, phosphorylation inhibits ATP binding

(Porter and Donoghue, 2003). Myt1 and Wee1 are active during the S and G2 phases of the

cell cycle (Porter and Donoghue, 2003). At the mitotic transition, Myt1 is phosphorylated by

Plk1 (only one member in Drosophila: Polo) and Cdk1 in vitro, resulting in lowered kinase

activity, indicating a possible feedback loop (Porter and Donoghue, 2003). Wee1 is

phosphorylated by the kinases Chk1 and Cds1, resulting in kinase inactivation, the 14-3-3

binding decreased and protein stability (Porter and Donoghue, 2003).

Activation of Cdk1 occurs through the combination of three required steps. First,

phosphorylation of Thr161 opens up the catalytic region of Cdk1. Phosphorylation of Thr161 is

done by Cdk-activating kinase (CAK), which occurs late in G2 and is not removed until the

cyclin B1 degradation in late mitosis (Fesquet et al., 1993). Second, nuclear localization of

Cdk1/cyclin B1 is promoted by Plk1 phosphorylation of Ser147 on Cdk1 (Porter and Donoghue,

2003). The final activation step of Cdk1 involves dephosphorylation of Thr14 and Tyr15 by

members of the Cdc25 phosphatase family, Cdc25A, Cdc25B, and Cdc25C (only one member

in Drosophila: String; Porter and Donoghue, 2003).

Cdc25A seems to be involved in the G1-S phase transition, whereas Cdc25B and

Cdc25C are involved in dephosphorylation and activation of Cdk1 in G2, with Cdc25C being the

primary phosphatase at the G2-M transition. Activation of Cdc25C is achieved by

49

Page 74: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

phosphorylation of Ser198 by Plk1 (Roshak et al., 2000). Ser198 lies within the nuclear export

signal of Cdc25C, and phosphorylation of this residue promotes localization of Cdc25C to the

nucleus. Once Cdc25C is activated and localised to the nucleus, it becomes

hyperphosphorylated by Cdk1/cyclin B1, creating a positive feedback loop, increasing Cdc25C

and Cdk1/ cyclin B1 activities. Inactivation of Cdc25C is achieved through phosphorylation of

Ser216, creating a binding site for 14-3-3 protein, sequestering Cdc25C in the cytoplasm, and

preventing Cdc25C and Cdk1 interaction. Phosphorylation of Ser216 has been attributed to

Chk1, Chk2, C-TAK1, and Plk3 (Porter and Donoghue, 2003; Ouyang et al., 1999).

Deregulation or mutations of Cdk1 itself have not been reported in any cancers.

However, inhibition of Cdk1 has been showed to induce cell cycle arrest and apoptosis. Cdk

inhibitors may impart their effects through either inhibiting the catalytic subunit, preventing

downstream phosphorylation, or through inhibiting the ability of Cdk1 to control transcription

through phosphorylation of RNA polymerase II. Many of the Cdks, including Cdk1,

phosphorylate the COOH-terminal domain of RNA polymerase II, inhibiting mRNA production

during mitosis (Oelgeschlager, 2002). Tumor cells seem to be particularly sensitive to RNA

polymerase II inhibition, indicating that the proapoptotic response to Cdk inhibition may be

due to both blocking its kinase activity on traditional targets and altering its affects on

transcription (Koumenis and Giaccia, 1997).

The basic biochemistry of the regulation of the Cdk-Cyclin B activation is conserved

from yeasts to humans.

In all known eukaryotes, the successive activation and inactivation of different Cdk-

Cyclin complex control the correct cell cycle progression (Pines, 1999). In lower eukaryotes,

only one or two cdc2/ cdc28-like genes products are found, while in mammals nine Cdks

(cdk1-9) have been already identified (Johnson and Walker, 1999). In budding yeast, the main

Cdk involved in cell cycle control is Cdc28, which is activated by nine different cell cycle

specific Cyclins (Levine et al.,1999). Three CLN or G1 Cyclins are involved in cell cycle events

that occur during G1 or at the start point (the point where a cell is committed to enter S-

phase). Six other cyclins know as Clb are required for initiation of DNA replication during S-

phase and subsequent mitosis (Nasmyth, 1996). In mammals, seventeen cyclins have been

identified: A, B1, B2, B3, C, D1, D2, D3, E, F, G1, G2, H, I, K, T1 and T2. All cyclin contain a

common region of homology, known as cyclin box, which is the domain used to bind Cdks

(Hunt, 1991).

III_2.1.2- Cyclin A

Cdk-Cyclin B is the major activator/regulator of mitosis, although another Cdk complex

is also present, at least during the firs stage of the mitosis, the Cdk-Cyclin A complex. Cdk1-

Cyclin A is formed in late G1 and is required for S-phase and G2/M transition. Little information

50

Page 75: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

is knowed about the importance of CdK1-Cyclin A complex in initiating S-phase. It has been

proposed that this complex is required for maintaining DNA synthesis once S-phase has been

initiated by another complex, the Cdk1-Cyclin E (Resnitzky et al., 1995). If the role of Cdk1-

Cyclin A is not well understood in S-phase, the activity of this complex during G2 and M-phase

is worse known. Drosophila cyclin A mutants can develop until cycle 14 of embryogenesis due

to the presence of maternal mRNA, however, after the exhaustation of maternal contribution,

cells no longer enter in mitosis, which suggests that Cyclin A is required for the initiation of the

M-phase (Lehner and O’Farrel, 1989). During early embryonic Drosophila cycles, Cyclin A is

mainly nuclear and the reduction of Cyclin A amount results in a increase in the normal

duration of the nuclear cycles (Stiffler et al., 1999). Further, the injection of anti-Cyclin A

antibodies in G2 tissues cultured cells, stops the cell cycle progression and cells are arrested

prior to mitosis onset, indicating that Cyclin A is required to initiate the mitosis (Pagano et al.,

1992). In addition, microinjections of exogenous Cdk1-Cyclin A in G2 vertebrate’s cells induce

a premature entry in mitosis and eliminate G2 phase. However, if the injections are made

during S-phase no effect on the progression to the next mitosis is observed (Furuno et al.,

1999). These authors suggested that Cdk1-Cyclin A complexes are required for cells to

complete prophase, since inhibition of this complex in mid prophase causes cell return to G2

phase. Cyclin A is degraded ahead of Cyclin B and it is degraded by proteolysis at the end of

pro-metaphase suggesting, that Cyclin A destruction is required for the subsequent alignment

of the chromosomes at the metaphase plate and for anaphase (Elzen and Pines, 2001; Geley

et al., 2001).

III_2.1.3- Cyclin B

Cyclin B is the mitotic cyclin with the clearest cell cycle role. Together with Cdk1, form a

primary mitotic Cdk which phosphorylates a large number of mitotic substrates (Nigg, 1993).

Among these substrates were identified nuclear lamins, cytoskeleton proteins, such as actin

biding proteins, and microtubules-associated proteins (MAPs). Therefore, it is easy to

understand that this complex has to be correctly activated in order to prevent drastic

consequences for cell.

In vertebrates cells, the cellular localisation of the Cdc2-Cyclin B complexes is

determined by the Cyclin B variant in which Cdc2 is bound. Cdc2-Cyclin B1 complexes co-

localise with cytoplasmic microtubules during interface and abruptly translocate to the nucleus

upon entry into mitosis (Heald et al., 1993; Jackman et al., 1995; Ookata et al., 1992; Pines

and Hunter, 1991). Cdc2-Cyclin B2 complexes co-localise with Golgi apparatus (Jackman et al.,

1995), and Cyclin B3, which share properties with both A and B-types cyclins, is constitutively

nuclear during cell cycle. In Drosophila, the localisation patterns of Cyclin B and B3 are similar

51

Page 76: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

to those of vertebrates Cyclin B1 and B3, respectively (Jacobs et al., 1998; Lehner and

O’Farrell, 1990).

The contribution of each class of MPF to the mitotic progression is not understood at

this point. Analyses of mices that are nullizygous for either Cyclin B1 or B2 have showed that

Cyclin B1, but not Cyclin B2, is essential for viability and fertility (Brandeis et al., 1998). This

indicates that Cdc2-Cyclin B2 is dispensable for mitotic progression, or alternatively, Cdc2-

Cyclin B1 is capable of compensating the Cdc2-Cyclin B2 loss. Cdc2-Cyclin B2 probably is

required for mitotic fragmentation of the Golgi apparatus (Lowe et al., 1998). It will be

interesting to determine the consequence of deleting Cyclin B3 in mice and investigate the

potential functional redundancies with Cyclin B1 and B2. In Drosophila, genetic studies have

demonstrated that, surprisingly, neither Cyclin B nor B3 are essential for cell division, however

removal of both Cyclins results in delayed entry into mitosis and formation of aberrant and

functionally compromised spindle (Jacobs et al., 1998). The Drosophila cyclin B3 is required for

female fertility, but not for male fertility (Jacobs et al., 1998).

Cdc2-Cyclin B1 activity is regulated by Wee1 and Cdc25C: Wee1 is nuclear until the

onset of mitosis while, Cdc25C is imported into the nucleus at G2/M transition (Heald et al.,

1993). Perhaps, Myt1 and Cdc25B function to coordinate cytoplasmic mitotic events, which are

mediated through Cdc2-Cyclin B1 (and possibly Cdc2-Cyclin B3). The division of labour

between the various MPF complexes may not be this straightforward; however, a cytoplasmic

accumulation of Cdc25B has been implicated in triggering centrosomal microtubule nucleation

in HeLa cells (Gabrielli et al., 1996).

Investigations about Cyclin B1 in Xenopus and HeLa cells have demonstrated a more

dynamic localisation; Cyclin B1 shuttles continuously between the cytoplasm and nucleus

during interphase (Hagting et al., 1998; Toyoshima et al., 1998; Yang et al., 1998). Import of

Cdc2-cyclin B1 into the nucleus occurs through binding of cyclin B1 to importin-β; Cdc2 is

dispensable for cyclin B1 nuclear import (Moore et al., 1999). Phosphorylation of four serine

residues located within the cyclin B, has been implicated in the nuclear retention of Cdc2-cyclin

B1 during the onset of mitosis (Li et al., 1997). Remains to be identified the kinase which

phosphorylated cyclin B1 and regulates its nuclear retention. Prior to the G2/M transition,

cdc2-cyclin B1 is maintained in the cytoplasm by the activity of the nuclear export factor CRMI

(also know as a exportin 1; Pines and Hunter, 1994).

In S. cerevisae, six B-type cyclins were identified (clb1-6; for review see Nasmyth,

1993). Clb1 and Clb2 are a closely related pair of B-type cyclins and are expressed in late S-

phase (Surana et al., 1991). Clb3 and Clb4 are expressed in early S-phase, and as Clb1 and

Clb2, are also expressed until late mitosis (Dahmann and Futcher, 1995). Clb5 and Clb6 are

the less similar to metazoan B-type cyclins (Mendenhall and Hodge, 1998).

The activation of the protein kinase by cyclin partner normally induces the entry into a

new cell cycle stage. Very often, the exit of a particular stage also requires the inactivation of

52

Page 77: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

the Cdk-Cyclin complexes. In mitosis, the Cdk-cyclin B complexes are normally inactivated by

proteolysis of the cyclin B. This destruction is thought to control progression during mitosis.

Immunofluorescence studies on tissue culture cells have showed that cyclin B

degradation starts during anaphase (Girard et al., 1995). Live analysis of cell proteins (Clute

and Pines, 1999) showed that in HeLa cells, cycB 1-GFP fusion proteins start to be degraded

30min before the metaphase/ anaphase transition, as soon as the last chromosomes is

attached to the spindle MTs. Cyclin B localisation was detected at the spindle poles during

prophase and prometaphase, but not in metaphase. Cyclin B was also seen associated with

chromosomes during prometaphase but again not in metaphase. These authors proposed a

pattern for Cyclin B degradation: Cyclin B starts to be degraded first from the spindle poles

and then from the chromosomes, just after the attachment of the last chromosome on the

metaphase plate.

A Cyclin B-GFP fusion protein was also studied in living Drosophila embryos (Huang and

Raf 1999). These authors showed that Cyclin B in interphase is placed at centrosomes (the

organelles that organise microtubules) and during mitosis, it is located at the mitotic spindle in

prometaphase and at the microtubules that overlap in the spindle mid-zone during metaphase.

A spatial regulation of Cyclin B destruction was also reported. Cyclin B disappears at the end of

metaphase, in a wave that started at the spindle poles and spreads to the spindle equator.

There is almost no Cyclin B-GFP at the moment that chromosomes enter in anaphase.

These two independent studies suggest that a substantial fraction of Cyclin B is being

degraded while the cell is still in metaphase, which is in contradiction with previous

immunostained studies that, reported an abrupt degradation of the protein at the

metaphase/anaphase transition. Moreover, the disappearance of Cyclin B seems to be

regulated spatially in both Drosophila and vertebrates cells.

In Addition, in Drosophila embryos it was showed that centrosomes (the organelles that

organise microtubules) have a role in the destruction of cyclin B (Wakefield et al., 2000). Using

a mutation called cfo (centrosome fall off), which produces embryos where the centrosomes

detach from formed spindle (centrosomeless spindles); Cyc B degradation was only detected at

the centrosome level and not on the spindle. These observations lead the authors to suggest

that cyclin B degradation starts at the poles and a connection between centrosomes and

spindle is required for the normal destruction of the cyclin B associated with the spindle.

III_3- The bipolar spindle and the kinetocore-microtubule attachment until

the formation of the metaphase plate

After nuclear envelope breakdown (NEBD), at prometaphase, each sister chromatid is

exposed and thus capable of capturing the spindle microtubules.

53

Page 78: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

Microtubules are metastable polymers of α- and β-tubulin that switch between phases

of growth and shrinkage, a phenomenon known as ‘dynamic instability’ (Mitchison and

Kirschner, 1984). Microtubule plus ends attach to the chromosome through a macromolecular

protein complex known as the kinetochore (see section III_3.1 of this chapter; reviewed in

Maiato et al., 2004). The minus ends of spindle microtubules are crosslinked to each other and

to interdigitated microtubules which is stretched from pole to pole, creating the spindle poles

(reviewed in Zimmerman and Doxsey, 2000). Anchorage of kinetochore-bound microtubules at

spindle poles, allows the force applied to a chromosome to be translated into directional

movement (Gordon et al., 2001).

Spindle poles and centrosomes anchor microtubules in distinct manners. Spindle poles

provide anchorage through crosslinking of microtubules by proteins such as NuMA, LGN, and

Dynein (Gaglio et al., 1996; Merdes et al., 1996; Heald et al., 1997; Du et al., 2001). In the

absence of centrosomes, spindle poles are sufficient to anchor microtubules allowing for high-

fidelity chromosome segregation.

Centrosomes nucleate and anchor microtubules.The γ-tubulin ring complex is directly

responsible for the centrosomes nucleating activity (Zheng et al., 1995) and also caps the

minus ends of microtubules (Wiese and Zheng, 2000; Moritz et al., 2000). Appendages on the

mother centriole within the centrosome, and a growing list of proteins, have been implicated in

the anchorage of microtubule minus ends at the centrosome, although the mechanism of this

activity is still unclear (reviewed in Bornens, 2002).

The kinetochore that has successfully captured a MT laterally is usually subjected to

rapid poleward motion. It is believed that, the poleward movement enables the kinetochore to

run into more MTs emanated from the approaching pole, to facilitate the stable, or “end-on”,

MT attachment (Rieder et al., 1998; Cleveland et al., 2003). Chromosomes with such

monooriented kinetochores, then experience oscillations toward poleward and away from their

attaching pole until their unattached kinetochores capture microtubules from the opposite pole.

For monooriented kinetochores, their poleward movement is attributed to the poleward

pulling force at the attached kinetochore, whereas their away pole movement is likely a

composite effect of both the pushing force at the kinetochore and the polar ejection force

acting on chromosome arms through non attached kinetochore- microtubules (Cleveland et al.,

2003; Kapoor and Compton, 2002). These antagonizing forces appear to act alternatively to

enable the oscillation, during which kinetochore- microtubules (k-fibers) remain attached but,

assemble and disassemble accordingly (Cleveland et al., 2003; Kapoor and Compton, 2002).

Full chromosome alignment is a prerequisite for equal segregation of sister chromatids into

daughter cells. The spatial location of each chromosome must differ from cell to cell after

nuclear envelope breakdown. Therefore, it is amazing that usually metaphase is accomplished

within 20 min (Alberts et al., 1994). How cells achieve chromosome alignment so efficiently is

still not fully understood.

54

Page 79: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

The bi-oriented chromosomes move towards the metaphase plate and finally align

there, waiting for inactivation of the spindle checkpoint (for more detail see section III_5.2 of

this chapter) to allow anaphase onset mediated by the Anaphase promoter complex or

clyclosome (APC/C for more detail see section III_4 of this chapter; Cleveland et al., 2003;

Kapoor and Compton, 2002; Rieder et al., 1998).

III_3.1- The Kinetochore

Kinetochores of animal cells can be subdivided into two regions. The inner kinetochore

normally forms on highly repetitive DNA sequences and, assembles into a specialised form of

chromatin that persists throughout the cell cycle. The outer kinetochore is a proteinaceous

structure with many dynamic components, that assembles and functions only during mitosis.

Kinetochore functions include: (1) attachment of the chromosomes to the dynamic plus-ends

of spindle microtubules, (2) mediation of mitotic chromosomes movements, and (3) the

control of the metaphase/anaphase transition by inhibiting sisters chromatids separation until

completion of a proper bipolar attachment.

The kinetochore is the physical location where the checkpoint signal is produced (detail

in section III_5.2 of this chapter). They also contain motor proteins that are involved in their

attachment and mobility.

The histone variant CENP-A and additional constitutive proteins mark the inner plate of

mammalian kinetochores, which is located at the periphery of centromeric chromatin

(Cleveland et al., 2003; DeLuca and Salmon, 2004; Meraldi et al., 2006). Before mitosis, other

evolutionarily conserved proteins, including Spc105 (KNL-1 in Caenorhabditis elegans), the

Mtw1 (MIS12 in humans), Ndc80 (HEC1 in humans) and minichromosome maintenance

protein-21 (MCM21) complexes, are recruited to the kinetochore outer plate to form

attachment sites for bundles of spindle microtubules, which are known as the kinetochore

microtubules (Cheeseman et al., 2006; Cleveland et al., 2003; DeLuca and Salmon, 2004;

DeLuca et al., 2006; Meraldi et al., 2006). Several other proteins, including the SAC and motor

proteins, bind within a region of the kinetochore that is known as the fibrous corona, and is

found at the periphery of the outer plate. The temporal order and specific requirements for the

recruitment of kinetochore and SAC proteins are complex (for review see Chan et al., 2005;

Liu et al., 2006; Johnson et al., 2004; Maiato et al., 2004; Taylor et al., 2004; Vigneron et al

2004; Wojcik et al., 2001).

The attachment of microtubules to multiple points within the spindle is a critical, yet

poorly understood, process. Two types of kinetochore-microtubule attachments have been

described: one dependent of Cenp-E and Dynein, and the other involving the Ndc80 complex.

The microtubule motors Cenp-E and Dynein initially form lateral attachments with

microtubule polymers and use their motor activities to move chromosomes (Wood et al., 1997;

55

Page 80: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

Hyman and Mitchison, 1991). These lateral or ‘‘side-on’’ attachments mature into ‘‘end-on’’

attachments consisting of bundles of spindle microtubules, termed kinetochore fibers (K-

fibers), embedded in the kinetochore’s outer plate. Mature K-fibers remain dynamic,

continuously adding tubulin subunits at their plus ends, but are resistant to depolymerization

treatments such as low doses of nocodazole and cold temperatures (reviewed in Maiato et al.,

2004).

Dynein is widely involved in cellular functions requiring MT-based motility, for instances,

mitosis, cell migration, and cargo transport (Alberts et al., 1994; Gupta et al., 2002; Karki and

Holzbaur, 1999). A pool of dynein is located to the kinetochore in M phase through dynactin

(Echeverri et al., 1996; King et al., 2000). The multiprotein complex, dynactin, increases the

processivity of dynein and anchors dynein to certain cargos or target sites. Dynein is recruited

to kinetochores by dynactin that binds the ZW10/Rod complex (Karess, 2005; Starr et

al.,1998). ZW10 may thus serve as an ideal target for regional elimination of dynein/dynactin.

Kinetochore dynein is generally believed to play a role in congression by driving the poleward

kinetochore movement (Cleveland et al., 2003; Maiato et al., 2004; Rieder et al., 1998).

The hetero-tetrameric Ndc80 complex residing in the kinetochore outer plate is required

for mature microtubule attachment and is a second type of kinetochore-microtubules

attachment (He et al., 2001; Wigge and Kilmartin, 2001; Deluca et al., 2002; McCleland et al.,

2003, 2004). In higher eukaryotes, the Mis12 and Zwint complexes also localise to the outer

plate and are also involved in kinetochore-microtubule attachment (Cheeseman et al., 2004;

Emanuele et al., 2005; Kline et al., 2006). Outer kinetochore proteins are thought to interact

with microtubule-associated proteins (MAPs) to facilitate mature microtubule binding.

III_4- The anaphase promoting complex/ Cyclosome (APC/C)

The anaphase promoting complex (APC/C) was first described as an ubiquitin ligase,

essential to the proteolytic degradation of the mitotic cyclins (King et al., 1995). Ubiquitin-

mediated proteolysis ensures the specific and rapid protein degradation in such a way that cell

cycle events become irreversible.

Ubiquitination of proteins is mediated by three types of enzymatic activity: an ubiquitin-

activating enzyme (E1), an ubiquitin-conjugated enzyme (E2) and an ubiquitin-ligase enzyme

(E3). The attachment of ubiquitin molecules to a target proteins are catalysed by the Ubiquitn

ligase. This will signal the target proteins for degradation by the 26S proteosome (Hershko,

1999). The 26S proteosome has a highly complex structure, it is composed by a 20S catalytic

core and a 19S regulatory cap and, it is present in the cytoplasm and nuclei of higher

eukaryotes (Wilkinson, 1999). The APC/C is an E3 ligase composed for eight to thirteen sub-

unities highly conserved (Peter, 1999). The APC/C is required for the degradation of mitotic

proteins such as mitotic cyclins A and B and securins (King et al., 1996; Morgan, 1999).

56

Page 81: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

III_4.1- The APC/C activators and substrate recognition

The specificity of the APC/C substrate is in part regulated by the activator associated to

the APC in a cell cycle dependent manner. APC/C bind to one of the two highly conserved WD

40 repeat proteins: Cdc20 (Fizzy/Slp1/p55Cdc) and cdh1 (hct1/Fizzy-related/Srw1/ste9). Cdc20

activates APC/C during early-mid mitosis (including mitotic cyclins and securins degradation)

and Cdh1 directs APC/C activity at final steps of mitosis and early G1 (Fang. et al., 1999).

In Cyclin B and securins, it was identified a sequence of nine amino acids (RXXLXXXXN)

called destruction box (D-box) which are essential for APC/C-mediated degradation (Glotzer et

al., 1991; King et al., 1996). Some models have been proposed to explain the contribution of

the Cdc20 to the D-box dependent recognition by the APC/C. A second motif, known as KEN

box, was identified in Cdc20, which is a target of APC/Ccdh1 that does not possess a D-box

motif (Pfleger and Kirschner, 2000). In Xenopus extracts, it was shown that APC/CCdc20 has a

strong affinity to D-boxes substrate, while APC/CCdh1 binds preferentially to a sequence of three

residues of amino acids known as KEN-boxes substrates (Pfleger et al., 2001a). PlK1 and ARK-

2 (kinase member of Aurora kinase family) are also APC/CCdh1 substrates during anaphase and

G1. Nevertheless, the efficient degradation of some proteins requires both motifs (Burton and

Solomon 2001).

III_4.2- APC/C regulation

The anaphase-promoting complex/ cyclosome (APC/C) is under complex control via

phosphorylation (Rudner and Murray, 2000) and by binding one of two WD 40 repeat co-

factors: Cdc20 (Fizzy/Slp1/p55Cdc) and Cdh1 (hct1/Fizzy-related/Srw1/ste9), which functions

as rate-limiting and cell cycle stage-specific activators of the APC/C (Morgan, 1999).

During the entry into mitosis, APC/C components are phosphorylated by Plk1 (Plk in

Drosophila). This phosphorylation acivates the APC/C complex and improves their affinity to

the Cdc20. At the same time, the phosphorylation of the Cdh1 components reduces its affinity

to the APC/C. At this moment, the APC/CCdc20 is able to signal to destruction proteins with D-

box motif, like Cyclin B and Securins (Pds1 in S. cerevisae and Cut2p in S. pombe). Cdc20 is

negatively regulated by the binding to the checkpoint proteins Mad2 and BubR1 (for details see

section III_5.2 of this chapter).

During late anaphase, the Cdc14 phosphatase removes the inhibitory phosphate from

the co-factor Cdh1. The desphosphorylation of Cdh1 promotes the replacement of Cdc20 by

Cdh1, and now the APC/CCdh1 targets to destruction Ken-box motif substrates, like Cdc20 (not

binding to the APC/C), PlK1 and AIRK-2 (kinase member of Aurora kinase family). The

APC/CCdh1 complex is maintained until G1.

In Drosophila, both the Cdc20 and the Cdh1 homolog (Fizzy and Fizzy related,

respectively), promote the degradation of cyclin B in a temporal and spatial way during

57

Page 82: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

mitosis. Early in mitosis, Fzy/Cdc20 (concentrated at kinetochores and centrosomes) initiates

the degradation of the spindle-associated cyclin B by the centrosomes and spreads to the

spindle equator. Slightly later in mitosis, Fzr/Cdh1 (concentrated at centrosomes throughout

the cell cycle) degrades the remaining cytoplasmic cyclin B (Huang and Raff, 1999, 2002; Raff

et al., 2002). In syncytial embryos, only Fzy/Cdc20 is present, and only the spindle-associated

cyclin B is degraded at the end of mitosis.

Cyclin A is also targeted for destruction by Fzy (Cdc20)–APC/C complexes, but it is not

concentrated on spindles and it seems unlike that Fzy/Cdc20 also catalyses the destruction of

cyclin A only on the spindle (Pines and Hunter, 1991; Dawson et al., 1995). Therefore, it was

proposed that, there must exist separate pools of Fzy/ Cdc20 responsible for the degradation

of cyclins A (in cytoplasm) and B (on the spindle). This hypothesis has important implications

in the target control by the spindle checkpoint. Only the pool of Fzy/Cdc20 that passes through

the kinetochore is inhibited from activating the APC/C by the spindle checkpoint system, and

only this pool of Fzy/Cdc20 is competent to catalyse the destruction of cyclin B. Thus, the

spindle checkpoint in Drosophila inhibits only the cyclin B degradation, whereas the destruction

of cyclin A is not (Whitfield et al., 1990; denElzen and Pines, 2001; Geley et al., 2001).

III_5- The regulation of the cell cycle: the checkpoints

Two major events, S-phase and mitosis are required for all cell cycles (except

endoreplication, where cells duplicate their DNA without mitosis); they ensure that both

daughter cells receive a full complement of chromosomes. For example, cells cannot divide

their DNA content in mitosis before proper duplication in S-phase. If these events are not

properly completed in the correct sequence, then the newly divided cells will not receive the

full set of genes required for all cellular activities; daughter cells then either die or sustain

genetic damage. During the cell cycle, control step are necessary to bring an orderly

progression through S-phase and mitosis and to couple these two processes with cell growth.

The concept of checkpoint arose from the observation that different mutants for cdc (cell

division cycle) arrest in different phases of the cell cycle, preventing the subsequential events

to occur (Weinert and Hartwell, 1988).

Nowadays, checkpoints are regulatory pathways at critical transition points, where

progression to the next cell-cycle step can be delayed or arrested by negative signals until the

correct conditions arise.

Two independent well-established checkpoints ensure the maintenance of the genetic

stability during the cell cycle: the “DNA damage checkpoint” that arrest cell in G2/M transition

in response to DNA damage or DNA replication errors, and “Spindle assembly checkpoint” that

prevents anaphase onset until all chromosomes are bipolarly attached. The components of

58

Page 83: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

these checkpoint pathways are conserved in all eukaryotes, enhancing the regulatory

machinery of the cell cycle.

III_5.1- DNA damage checkpoint

In response to DNA damage, such as that caused by ionizing radiation, the most

prominent arrest is in the G2 phase (Fig. III_4; Iliakis et al., 2003).However, in cells that have

a functional p53 pathway, both a G1 and a G2 arrest can take place in response to DNA

damage (Crosby e al., 2007; Wahl and Carr, 2001). It has been established that the G2

checkpoint initiation is primarily regulated through the control of Cdk1 (Cdc2) activity, which is

regulated at multiple levels (Fig. III_4), including periodic association with the B-type Cyclins,

reversible phosphorylation, and intracellular compartmentalisation (Atherton et al., 1993).

Cyclin B1, the prototypical cyclin B (B2 and B3 are less abundant), reaches maximum levels in

G2, when it enters into the nucleus to form a complex with Cdk1 in a phosphorylation-

dependent manner (Hagting et al., 1998). The complex is activated by phosphorylation of

Cdk1 on threonine 161 (T161) by Cdk-activating kinase (CAK), but is kept inactive by its

phosphorylation on threonine 14 (T14) and tyrosine 15 (Y15) by the Myt1 and Wee1 kinases,

respectively (explained in section III_2.1.1 of this chapter; Parker and Piwnica-Worms, 1992).

Following DNA damage, Chk1 and Chk2 kinases are phosphorylated and thus activated

by ATM (ataxia telangiectasia-mutated) and ATR (A-T- and rad3-related) kinases, related

members of the phosphatidylinositol 3-kinase (PI3K) family of proteins (Kastan and Bartek,

2004; Ray et al., 2007). The activacted Chk1 and Chk2 kinases phosphorylate the Cdc25C

protein phosphatase at serine 216 (Blasina et al., 1999; Furnari et al., 1999). Once

phosphorylated, Cdc25C can then to be bound by 14-3-3s and to be sequestered in the

cytoplasm (Blasina et al., 1999). Cdc25C, only when it is in the nucleus, dephosphorylates the

Thr14 and Tyr15 residues and thus activates the Cdk1/cyclin B1 complex (Peng et al., 1997;

Sanchez et al., 1997; Solomon et al., 1990). Additional phosphatases, Wip1 (PPM1D)

(Shreeram et al., 2006) and PP2A, (Margolis et al., 2006) may also regulate the checkpoint by

dephosphorylating other factors that contribute to the activation of Cdk1. Cdk1/cyclin B can

phosphorylate Cdc25C, further activating it and initiating a positive feedback loop (Izumi et

al.,1992). A nuclear export signal (NES) in Cyclin B1 facilitates rapid export of the Cdk1/cyclin

B1 complex from the nucleus (Pines and Hunter, 1994). Phosphorylation of the NES blocks the

nuclear export by interfering with the binding of the nuclear export receptor.

In response to incomplete DNA replication, another kinase is activated instead (Elledge,

1996). Chk1 and Cds1 kinases modulate the activity of Cdk1 by activating Wee1/Myt1 kinases

or inhibiting Cdc25 phosphatase. Both kinases can phosphorylate Cdc25 in vitro, thereby

inhibiting their activity (Furnari et al., 1997).

59

Page 84: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

Figure III_4- Schematic representation of the G2 checkpoint (in Plesca et al., 2007).

Cdk1 is the key control point for G2 checkpoint initiation, which is dependent on both positive and negative regulators.

To be active, Cdk1 requires binding of its catalytic partner, cyclin B1 (also B2 and B3 in some circumstances), which is

regulated transcriptionally and by its nuclear localization. Cdk1 is regulated by its phosphorylation, as determined by a

balance of kinase and phosphatase activities, as well as at the level of transcription. G2 progression to mitosis is

triggered by the Cdc25 mediated dephosphorylation of the Cyclin B/Cdk1 complex. Cyclin B/Cdk1 is activated by

phosphorylation of Thr161 by CAK (Cyc H/Cdk7 complex) and the dephosphorylation of Thr14 and Tyr15 by Cdc25C.

The complex is kept in an inactive state due to the phosphorylation of Thr14 and Tyr15 by the Myt1 and Wee1 kinases

that can in turn be regulated by Plk1. The stable maintenance of the G2 arrest is determined by the activity of E2F,

Rb, or p53 family of transcription factors through their transcriptional targets, which include many of the components

described here. p53 regulates the stability of the checkpoint through levels of its mitosis target genes cdk1, cyclin B1,

but also p21 (CDKN1A) and 14-3-3s. The activity of Cdc25 is also regulated by Chk1 or Chk2-mediated

phosphorylation, leading to its inactivation through binding to and sequestration by 14-3-3s. ATM/ATR kinases

transduce the DNA damage signal to the effector kinases Chk1 and Chk2. Chk1 and Plk1, by regulating Wee1, also

play a role in the recovery from the arrest.

More recent studies suggest that Plk is also an important target of DNA damage

checkpoint. In human cells, DNA damage inhibits the activation of Plk and CdK1-Cyclin B in an

ATM-dependent manner. Moreover, G2/M arrest induced by DNA damage could be overcome

by constitutive expression of a Plk1 active form (Smits et al., 2000). In addition, a decrease in

Plk1 was observed in an ATM-dependent response to radiomimetic drugs and in an ATR-

dependent response to ultraviolet treatment and to topoisomerase-II inhibitors (Van Vugt et

al., 2001; Deming et al., 2002). Furthermore, Plk1 was identified as a putative substrate of the

Chk2 kinase (Seo et al., 2003). All theses studies indicated Plk1 is also a target of the DNA

damage checkpoint cascade.

60

Page 85: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

III_5.2- Metaphase or Spindle assembly checkpoint (SAC)

We will make a more detailed description of this control step, once identified mitotic

mutants (see chapter II part IV of this manuscript) revealed problems at the metaphase-

anaphase transition/regulation.

The spindle assembly (or metaphase) checkpoint is a quality control mechanism that

prevents chromosome segregation errors. The spindle assembly checkpoint detects spindle

assembly defects and, loss or impairment of functional connections between kinetochores and

spindle microtubules during mitosis, disseminating inhibitory signals that delays anaphase

onset (Fig. III_5). This signal is presumably amplified and ultimately acts to arrest the APC/C

(reviewed by Hoyt, 2001; Shah and Cleveland, 2000; Amon, 1999).

In 1991, two independent screens identified various genes mutation, of which bypassed

the ability of wild-type S. cerevisiae cells to arrest in mitosis in the presence of spindle poisons

(Hoyt et al., 1991; Li and Murray, 1991). The genes identified in these screens included the

Mad (mitotic-arrest deficient) genes mad1, mad2 and mad3 (bubR1 in humans), and the Bub

(budding uninhibited by benzimidazole) gene bub1 (Hoyt et al., 1991; Li and Murray, 1991).

Another bub gene, bubB3, was identified as an extra-copy suppressor of the bub1-1 mutant1.

These genes are conserved in all eukaryotes, and they are collectively involved in a spindle-

assembly checkpoint (SAC) pathway that is actived in prometaphase and which prevents the

precocious separation of sister chromatids (Musacchio and Hardwick, 2002; Taylor et al.,

2004).

The target of the complex intracellular mechanisms of the spindle assembly checkpoint

is the protein Cdc20, a co-factor of the ubiquitin ligase APC/C (Hwang et al.1998; Kim et al.,

1998). Specifically, the SAC negatively regulates the ability of Cdc20 to activate the APC/C-

mediated polyubiquitynation of two key substrates, cyclin B and securin, thereby preventing

their destruction by the 26S proteasome. Securin is a stoichiometric inhibitor of a protease

known as separase. Separase is required to cleave the cohesin complex that holds sister

chromatids together, and cohesin cleavage is required to execute anaphase (Peters, 2007).

The inhibition of the APC/C causes high cyclin levels, sustained cyclin-dependent kinase 1

(cdk1) activity, and prolonged mitotic arrest. The mitotic arrest allows time for the correction

of the chromosome connections to the spindle. Only when all chromosomes have become bi-

orientated between separated spindle poles on the metaphase plate, the checkpoint is silenced

relieving the mitotic arrest and allowing anaphase to proceed (Bhalla, 2003; Jordan and

Wilson, 2004; Kops et al., 2005).

61

Page 86: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

Figure III_5- Relationship of the SAC with the cell-cycle machinery (in Musacchio and Salmon, 2007).

Mitosis is traditionally subdivided into five consecutive and morphologically distinct phases: prophase, prometaphase,

metaphase, anaphase and telophase. To enter mitosis, the cell requires the activity of the master mitotic kinase,

cyclin-dependent kinase-1 (CDK1), which depends strictly on the binding of cyclin B to CDK1 (Peters, 2006). Separase

is a protease, the activity of which is required to remove sister-chromatid cohesion at the metaphase-to anaphase

transition (cohesin is indicated in yellow on the expanded view of the chromosome). Prior to anaphase, separase is

kept inactive by the binding of a protein known as securin (SEC). Unattached kinetochores (red hemi-circles)

contribute to the creation of the mitotic checkpoint complex (MCC), which inhibits the ability of CDC20 to activate the

anaphase-promoting complex/cyclosome (APC/C). The attachment of all sister-kinetochore pairs to kinetochore

microtubules, and their bi-orientation — which produces congression to the spindle equator — negatively regulates the

spindle-assembly checkpoint (SAC) signal. This releases CDC20, which can now activate the APC/C. This results in the

polyubiquitylation of anaphase substrates such as cyclin B and securin, and their subsequent proteolytic destruction by

the proteasome. The degradation of SEC results in the activation of separase, which targets the cohesin ring that is

holding the sister chromatids together, thus causing the loss of sister-chromatid cohesion and the separation of sister

chromatids. The degradation of cyclin B at this stage also inactivates the master mitotic kinase CDK1–cyclin B,

initiating cytokinesis and the mitotic-exit programme.

62

Page 87: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

III_5.2.1- Models for spindle assembly checkpoint sense the defects

Two models are proposed to explain how spindle assembly checkpoint detects a single

spindle defects. One model proposes that checkpoint machinery is sensitive to the tension

exerted in kinetochore by the opposite spindle microtubules forces. The other explains that

metaphase checkpoint monitors the availability of kinetochores unattached.

Figure III_6- The attachment process (in Musacchio and Salmon, 2007).

(a) Unattached kinetochores generate a ‘wait’ signal and recruit the spindle assembly checkpoint (SAC) proteins. The

levels of mitotic-arrest deficient homologue-2 (MAD2) are high at unattached kinetochores (left) and moderately high

at attached kinetochores in a monotelic pair (right). Under these conditions, the Aurora-B kinase concentrates at

centromeres and is believed to be activated by the lack of tension between the sister chromatids. Bi-orientation

depletes MAD2 (and budding uninhibited by benzimidazole (BUBR1) from kinetochores and promotes the acquisition of

tension in the centromere area, which is visualized as an increase in the inter-kinetochore distance between sister

chromatids. When all chromosomes have achieved this situation, the SAC signal is extinguished and anaphase ensues

thanks to the activation of separase, which removes sister-chromatid cohesion by proteolysing cohesin. At entry into

anaphase, Aurora-B is also depleted from the centromere region.

(b) Correct and incorrect attachments can occur during mitosis, and correction mechanisms exist to prevent incorrect

chromosome inheritance, which would occur if improper attachment persisted until anaphase. Monotelic attachment is

a normal condition during prometaphase before bi-orientation. In syntelic attachment, the both sisters in a pair

connect to the same pole. A mechanism that corrects this improper attachment depends on the Aurora-B/Ipl1 kinase.

Merotelic attachment occurs quite frequently and is also corrected by the Aurora-B kinase. The SAC might be able to

sense syntelic attachment, but it is unable to detect merotelic attachment.

63

Page 88: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

The first arguments in favour of tension model come from experiments of

micromanipulation performed in the spermatocytes of praying mantids, which have three

sexual chromosomes (two X chromosomes and one Y chromosome; Gorbsky and Ricketts,

1993; Li and Nicklas, 1995; Li and Nicklas, 1997; Nicklas et al., 1995). In this system, only

one chromosome attached to a single spindle pole is sufficient to inhibit the progression to

anaphase. When mono-attach X-chromosome is pulled with a microneedle, the tension is re-

established. In this case, the inhibition of the progression of mitosis is cancelled and the cells

progress normally to anaphase.

However, tension don’t explain everything, the laser ablation of the last unattached

kinetochore silence the spindle checkpoint and allows the progression to anaphase, despite the

lack of tension in the monooriented sister chromatid (Rieder et al., 1995).

III_5.2.1.1- Microtubule attachment model

The SAC monitors the attachment of kinetochores to microtubule plus-ends during an

unperturbed mitosis. Ideally, SAC activity at sister kinetochores should remain active until bi-

orientation, the only condition that ensures accurate segregation at anaphase (Fig. III_6a).

Cyclin B and securin start to be degraded after the last chromosome has been aligned, and

they become largely depleted prior to anaphase (Clute and Pines, 1999; Hagting, A. et al.,

2002). Early during this process, the addition of spindle poisons stops proteolysis immediately

and blocks anaphase onset in a SAC-dependent manner (Clute and Pines, 1999). Interference

with kinetochore assembly impairment of microtubule motors (for example, dynein or CENP-E)

and interference with microtubule dynamics also activate the SAC (Rieder and Maiato, 2004).

Certain SAC proteins are immediately removed after the attachment of microtubule plus-ends

and formation of kinetochore microtubules. For example, Mad2 localises to unattached

kinetochores during prometaphase or in cells treated with nocodazole, which induce MT

depolymerization and prevent formation of MT–kinetochore end-on attachments (Chen et al.,

1996; Waters et al., 1998). Conversely, the amount of Mad2 becomes highly reduced at

metaphase kinetochores (50–100-fold comparatively with unattached prometaphase

kinetochores). Therefore, kinetochore localization of Mad2 decreases substantially as

kinetochores fill more of their attachment sites with kinetochore microtubules (occupancy).

Because Mad1 and Mad2 accumulate at unattached kinetochores and are removed on

attachment, their kinetochore localisation is interpreted to signify lack of attachment (Howell

et al., 2001; Skoufias et al., 2001; Waters et al., 1998). In metazoans, Mad2 depletion from

kinetochores depends not only on MT attachment, but also on cytoplasmic dynein motility

along MT. The inhibition of the dynein at metaphase kinetochores results in the return to

~25% of the level of Mad2 at unattached kinetochores, without a loss in kinetochore-

microtubule number (Howell et al., 2001; Wojcik et al., 2001). This indicates that,

64

Page 89: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

kinetochore-MT formation promotes Mad1 and Mad2 dissociation by providing high local

concentrations of MT, in order to promote the ATP-dependent motility of dynein along

microtubules (Howell et al., 2001; Wojcik et al., 2001).

III_5.2.1.2- Tension model

In addition to microtubule–kinetochore attachment, tension is important for SAC

inactivation (Nicklas et al., 1995; Nicklas, 1997). Stretching of centromeric chromatin on bi-

orientation, increases kinetochore-to-kinetochore distance and, consequently, kinetochore

tension. MT–kinetochore attachment is normally destabilised at low kinetochore tension and

stabilised by high tension between bi-orientated sisters kinetochores (Nicklas, 1997; Nicklas et

al., 2001). Therefore, tension might provide a fundamental criterion to discriminate against

incorrect attachments. For example, if both sister kinetochores attach to microtubules from the

same pole (syntelic attachment; Fig. III_6b), not enough tension is generated and MT–

kinetochore attachment is destabilised to correct the problem. This destabilisation depends on

Aurora-B/Ipl1 kinase (Hauf et al., 2003; Lampson et al., 2004; Pinsky and Biggins, 2005;

Tanaka et al., 2002). Aurora-B/Ipl1 is also critical for correcting merotelic attachments, which

are not sensed by the SAC. Merotelic attachment occurs when one sister kinetochore becomes

attached to MT from opposite poles. Bi-orientation of chromosomes with merotelic

kinetochores produces sufficient occupancy and tension to turn off SAC activity (Fig. III_6b).

As a result, merotelic kinetochores, if left uncorrected, can produce lagging chromatids and

potential chromosome mis-segregation in anaphase (Cimini and Degrassi, 2005). Tension and

centromere stretch also seem to have a significant role in turning off SAC activity by inhibiting

the association rate of SAC proteins at kinetochores. For example, SAC activity is turned on by

adding taxol or low doses of vinblastine to metaphase HeLa cells to stop kinetochore-MT

dynamics and reduce tension. (Taxol decreases inter-kinetochore tension in bi-orientated

sisters, while stabilising the number of metaphasic kinetochore-MT.) In cells undergoing these

treatments, SAC activation correlates with a significant increase in the concentration of BubR1

and Bub1 at kinetochores, as well with the phosphorylation of an unknown phosphoepitope by

Plk1 kinase that is detected by the antibody 3F3/2 (Gorbsky and Ricketts, 1993; Nicklas,

1997; Waters et al., 1998). This type of experiment does not prove that lack of tension alone

is sufficient to activate the SAC, once that, metaphase cells treated with taxol were shown to

contain one or a few Mad2-positive kinetochores (possibly owing to low occupancy), which

could be sufficient to trigger a SAC arrest (Waters et al., 1998). However, 3F3/2

phosphorylation and re-recruitment of BubR1 and Bub1, but not Mad2 to all kinetochores,

imply that the reduction in tension changes the kinetochore in a significant way, independently

of any change in occupancy. Therefore, irrespective of whether the checkpoint is indeed

regulated by tension, independently of occupancy, kinetochore localisation of checkpoint

65

Page 90: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

components is differentially regulated by occupancy and tension. Even so, it must be

remembered that, distinguishing the relative contributions of tension and attachment, when

manipulating spindles is difficult, since interfering with the creation of tension probably affects

attachment (Pinsky and Biggins, 2005; Zhou et al., 2002).

We remain unaware of any SAC or SAC-related protein for which activity is directly

regulated by tension in an adequately reconstituted system. SAC activity during the successive

phases of bi-orientation, which macroscopically define the attainment of attachment and

tension, seems to result in the generation of the same cell-cycle inhibitor, the MCC18.

Consistently, Mad2 is required to sustain the SAC, even after its substantial depletion from

kinetochores (Howell et al. 2001; Shannon et al., 2002). Similarly, although kinetochore Mad2

remains low in taxol-treated cells, Mad2 is critically required to maintain the taxol-dependent

arrest (Howell et al. 2001; Shannon et al., 2002; Skoufias et al., 2001; Waters et al., 1998).

The separation at the molecular level between attachment and tension will be clarified through

a better understanding of the physical connections at the kinetochore between the MT-binding

machinery and the SAC.

III_5.2.2- Inhibition of APC/C by the spindle assembly checkpoint

Although the nature of the direct molecular interactions between SAC proteins and

kinetochores and, the interdependencies of SAC proteins have not been determined

(Musacchio and Hardwick, 2002), the basic plan of the signalling cascade is established (Fig.

III_7).

APC/CCdc20 is a key molecular target of the spindle checkpoint (Bharadwaj and Yu,

2004). Biochemical analysis revealed that (Mad)1-3 and (Bub)1-3, the components of the

spindle checkpoint (Bharadwaj and Yu, 2004) use multiple strategies to inhibit APC/CCdc20 upon

checkpoint activation. Stoichiometric binding of either Mad2 or BubR1 (the vertebrate ortholog

of yeast Mad3) to Cdc20, inhibits APC/CCdc20 in vitro (Fang et al., 1998a; Tang et al., 2001a).

Mad2, Mad3/BubR1, Bub3, and Cdc20 can form a single mitotic checkpoint complex in vivo

(Sudakin et al., 2001). In addition, Bub1 phosphorylates Cdc20 inhibiting catalytically

APC/CCdc20 (Tang et al., 2004). Finally in yeast, Mad2 and Mad3 binding to Cdc20, triggers the

auto-ubiquitination of Cdc20 and reduces the protein level of Cdc20 (Pan and Chen, 2004),

although it remains to be determined whether this mechanism is conserved in other

organisms.

Fluorescence recovery after photobleaching (FRAP) experiments revealed that, the

kinetochore-bound pools of BubR1, Mad2, and Cdc20 exchange rapidly with their cytosolic

pools (Howell et al., 2004; Shah et al., 2004). Furthermore, structural and biochemical

analysis of Mad1 and Mad2 suggests that the kinetochore-bound Mad1 promotes a

conformational change of Mad2, which is required for its efficient binding to Cdc20 (reviewed in

66

Page 91: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

Yu, 2006). Finally, in Xenopus egg extracts, phosphorylation of Cdc20 by MAPK and Cdk1 is

required for efficient Mad2 binding and, for the spindle checkpoint (Chung and Chen, 2003;

D’Angiolella et al., 2003). These findings support an attractive model, in which the unattached

kinetochore facilitates the formation of APC/C-inhibitory checkpoint complexes containing

BubR1, Bub3, Mad2, and Cdc20: by increasing the concentrations of these proteins locally, by

triggering a conformational change of Mad2 and, by posttranslational modifications on Cdc20.

In addition, Bub1 bound to chromosomes exhibiting higher autophosphorylation activity as

compared to cytosolic Bub1 (Chen, 2004). This suggests that unattached kinetochores might

also stimulate the kinase activity of Bub1 toward Cdc20 facilitating inhibition of APC/CCdc20. It

remains to be determined, whether phosphorylation of Cdc20 by Bub1 also enhances its

binding to Mad2 or BubR1, similar to phosphorylation of Cdc20 by MAPK and Cdk1 (Chung and

Chen, 2003; D’Angiolella et al., 2003).

In an alternative model, it was proposed that the mitotic checkpoint complex forms

constitutively during the cell cycle, but it is only capable of associating with the APC/C core

modified during mitosis by unattached kinetochores and spindle checkpoint signalling (Sudakin

et al., 2001). This model is inconsistent with the fact that, the concentrations of mitotic

checkpoint complex are much higher in mitosis than during interphase in several organisms

(Chen, 2002; Millband and Hardwick, 2002). Furthermore, no such spindle checkpoint-

dependent modifications of APC/C, which allow mitotic checkpoint complex binding in mitosis,

have been discovered so far.

It has been suggested that Mad2 binding to Cdc20 prevents the release of substrates

(presumably ubiquitinated substrates, i.e., products of the ubiquitination reaction), thereby

inhibiting APC/CCdc20 (Pfleger et al., 2001b). It is unclear, how binding of BubR1 to Cdc20 or,

Bub1-mediated phosphorylation of Cdc20, inhibits APC/CCdc20. Both Mad2 and BubR1 can be

coimmunoprecipitated with core subunits of APC/C, such as Cdc27 in nocodazole-arrested

mitotic human cells (Fang et al., 1998a;Sudakin et al., 2001), suggesting that binding of Mad2

or BubR1 to APC/C does not dissociate Cdc20 from APC/C. Likewise, phosphorylated Cdc20

remains bound to APC/C in mitosis. Thus, Mad2, BubR1, and Bub1 inhibit APC/CCdc20 through

the interference with the function of Cdc20 at a step that is subsequent to Cdc20 binding to

APC/C, possibly by substrate recruitment and/or product release.

Though counterintuitive, this mode of APC/CCdc20 inhibition by the spindle checkpoint is

in fact advantageous for two reasons. (1) While the spindle checkpoint inhibits the ubiquitin

ligase activity of APC/C toward securin and cyclin B, it does not block degradation of certain

APC/CCdc20 substrates, such as cyclin A and Nek2A (Geley et al., 2001; Hayes et al., 2006). (2)

The association of Mad2- and BubR1-bound Cdc20 with APC/C provides an opportunity for

Cdc20 autoubiquitination by APC/C. In one hand, Cdc20 autoubiquitination, at least in yeast,

reduce the protein levels of Cdc20, facilitating inhibition of APC/CCdc20 by the spindle checkpoint

67

Page 92: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

(Pan and Chen, 2004). In other hand, Cdc20 autoubiquitination dissociates Mad2 from Cdc20

to inactivate the spindle checkpoint (Reddy et al., 2007; Stegmeier et al., 2007).

III_5.2.2.1- Recruitment of spindle checkpoint proteins to unattached/

untensed kinetochores

Regardless of the competing models, it is generally agreed upon that ultimately,

checkpoint signal has to originate from the kinetochores of misaligned chromatids and is then

distributed and amplified throughout the cell to exert inhibition of APC/C (Fig. III_7).

A signalling network that recruits spindle checkpoint proteins to unattached

kinetochores has been established, by examining their interdependency at kinetochores in

mammalian cells and in Xenopus egg extracts.

The chromosome passenger complex (CPC), containing Aurora B, INCENP, survivin and

Dasra/Borealin, recruits the Bub1-Bub3 complex and Mps1 to the kinetochores (Johnson et al.,

2004; Vigneron et al., 2004). The CPC and Bub1 collaborate to recruit Plk1 to kinetochores

through Bub1-Plk1 and INCENP-Plk1 interactions (Goto et al., 2006; Qi et al., 2006). Plk1 then

facilitates the kinetochore recruitment of Cdc20 and the BubR1-Bub3 and Mad1-Mad2

complexes (Ahonen et al., 2005; Wong and Fang, 2005). Plk1 phosphorylates and targets

Plk1-interacting checkpoint helicase (PICH, which is a member of the SNF2 family of DNA-

binding ATPases) to kinetochores during prometaphase and in anaphase to stretched

centromeric chromatin that contains concatenated DNA (Baumann et al., 2007). PICH is

specifically required for the kinetochore localisation of Mad2, but not of Mad1, suggesting that

PICH maintains the Mad1-Mad2 interaction at the kinetochores in response to the lack of

tension across the kinetochores (Baumann et al., 2007). Interestingly, depletion of Nek2A also

causes delocalisation of Mad2 from kinetochores without affecting the kinetochore localisation

of Mad1 (Lou et al., 2004). Recently, Draviam and collegues identified the protein kinase TAO1

as a novel spindle checkpoint protein, that was required for the kinetochore localisation of

Mad2, but not other spindle checkpoint proteins examined, including Mad1 (Draviam et al.,

2007).

Therefore, upon checkpoint activation, an elaborate signaling network targets the Mad

and Bub proteins to unattached/untensed kinetochores, which then promotes the formation of

APC/C-inhibitory complexes.

Recently, Cdc20 autoubiquitination was suggested as an important mechanism for

regulating the Mad2-Cdc20 interaction and for checkpoint inactivation. New antagonistic

proteins UbcH10 and USP44 was recently identified and implicated in Cdc20 autoubiquitination.

The UbcH10 (UBC) was implicated in the checkpoint inactivation (Reddy et al., 2007). It

was showed that UBC promotes Ccd20 autoubiquitination, disrupting Mad2-Cdc20 interaction

and activating APC/C (Reddy et al., 2007). Consistent with earlier studies that established a

68

Page 93: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

role of p31 comet (a conformation-specific Mad2-binding protein) in checkpoint inactivation

(Xia et al., 2004), p31 comet and UBC showed a synergistic effect on checkpoint inactivation

through Cdc20 autoubiquitination (Reddy et al., 2007).

Figure III_7- A Model for the Regulation of APC/CCdc20 by the Spindle Checkpoint (in Yu, 2007).

Upon checkpoint activation, an elaborate signaling network targets the spindle checkpoint proteins to unattached

kinetochores. The chromosome passenger complex (CPC) containing Aurora B and INCENP recruits Bub1-Bub3.

Phosphorylation of INCENP and Bub1 by Cdk1 creates docking sites for the polo-box domain of Plk1, targeting Plk1 to

kinetochores. Plk1 then facilitates the kinetochore localization of PICH, Mad1-Mad2, BubR1-Bub3, and Cdc20. The

close proximity of Cdc20 and the checkpoint proteins at kinetochores and a Mad1-triggered conformational change of

Mad2 facilitate the formation of the mitotic checkpoint complex (MCC) containing BubR1, Bub3, Mad2, and Cdc20,

which associates and inhibits APC/C. Phosphorylation of Cdc20 by Bub1 also inhibits APC/CCdc20. The concentration of

Mad2-Cdc20-containing checkpoint complexes is controlled by two opposing dynamic processes: kinetochore-

facilitated formation and p31comet- and UbcH10-dependent disassembly through Cdc20 autoubiquitination. Protectin

deubiquitinates Cdc20 and maintains the Mad2-Cdc20 interaction, contributing to checkpoint activation. When the

kinetochores are captured by spindle microtubules and are under tension, the concentrations of checkpoint proteins at

kinetochores and possibly their activities in certain cases decrease. p31comet binds to Mad1-bound Mad2, preventing

the generation of the active conformer of Mad2. These mechanisms reduce the rate of formation of Mad2-Cdc20.

Meanwhile, p31comet binds to Cdc20-bound Mad2 and, along with UbcH10, stimulates the autoubiquitination of Cdc20

and the disassembly of Mad2-Cdc20. This leads to checkpoint inactivation and activation of APC/CCdc20, which

indirectly activates APC/CCdh1. APC/CCdh1 then targets several spindle checkpoint proteins for degradation, including

Plk1,Aurora B, Mps1, and Bub1, and prevents the reestablishment of the spindle checkpoint following mitotic exit.

69

Page 94: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

III-Overview of Drosophila Cell Cycle

The USP44 protein, an ubiquitin isopeptidase, was identified as a novel spindle

checkpoint protein (Stegmeier et al., 2007). It was shown that USP44 has a role in overlap the

mitotic arrest induced by taxol treatment, and is also required for the proper timing of mitotic

events during the normal cell cycle, a function shared with Mad2 and BubR1 (Meraldi et al.,

2004). In addition, USP44 antagonised the ability of UbcH10 and p31 comet to induce the

disruption of the Mad2-Cdc20 interaction and APC/C activation in vitro. Importantly, the

isopeptidase activity of USP44 is required for spindle checkpoint signaling in cells and appears

to be activated by phosphorylation during mitosis. Activated recombinant USP44 directly

removed ubiquitin from ubiquitinated Cdc20. The USP44 protein was renamed protectin to

reflect its role in reversing Cdc20 autoubiquitination and consequently protecting the Mad2-

Cdc20-containing checkpoint complexes from disassembly (Stegmeier et al., 2007).

Collectively, these two studies demonstrated that cellular concentrations of Mad2-Cdc20

and the activation status of the spindle checkpoint depend on the delicate balance between

two opposing dynamic processes, providing a fast on-or-off switch for the spindle checkpoint

(Reddy et al., 2007; Stegmeier et al., 2007). It is unclear whether UbcH10- and p31comet-

dependent Cdc20 autoubiquitination and the activity of protectin are directly regulated by the

spindle checkpoint.

70

Page 95: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

CHAPTER II

RESULTS

Page 96: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

72

Page 97: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_Part I:

Page 98: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

74

Page 99: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

New teashirt alleles obtained by P-element excision mutagenesis

Silvia Pimentel1,2, Isabel Salazar2, Christine Vola2, Rui Gomes 1 and Laurent Fasano2

1 Departamento de Biologia Vegetal, Faculdade de Ciências da Universidade de Lisboa,

Edifício C2-4º, Campo Grande, 1749-016 Lisboa, Portugal

2 Institut de Biologie du Développement de Marseille Luminy (IBDML), UMR 6216,

CNRS-Université de la Méditerranée, Campus de Luminy, Case 907, 13288 Marseille Cedex 09,

France.

Page 100: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

76

Page 101: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

Abstract

The hypomorphic allele tshA3-2-66 corresponds to a P{lacW} insertion 1652 base pairs

(bp) upstream the translation start of the teashirt (tsh) gene. New mutations had been

generated by mobilization of the P-lacW transposon, which is marked with a mini-white+ gene.

The endogenous white locus was the w1118 allele. Precise and imprecise excisions (E{P}) of

the tshA3-2-66 transposon were induced in the presence of the Sb P[ry+ 2-3] source of

transposase and male selected in F2 on the basis of the loss of the eye color marker.

Out of 760 F2 males screened, we obtained 145 E{P}/CyO P-element-excised

revertants lines. These revertant were tested with the original P{lacW}A3-3-66 mutant, 20 were

embryonic lethal, 101 died during larval stages and 2 didn’t survive the pupal stage. One line

that gave fertile adults might correspond to a precise P-element excision event. We observed

female escaper sterile in 6 of the F2 lines, probably resulting from imprecise P-element

excision events. This female failed to lay eggs and presented atrophied ovaries with a strong

arrest in early oogenesis. The P-element excision from tsh locus were confirmed. We identified

6 lines that was homozygous lethal (∆P30, ∆P99, ∆P104, ∆P123, ∆P126 and ∆P128) and gave

a severe female sterile phenotype when in Trans with tshA3-2-66, probably resulting from

imprecise P-element excision events. This female show a modifications in the eye patterned

white expression observed in the tshA3-2-66 adult eyes. Absence of re-integration of the P-

element was tested by PCR.

Key words: Drosophila, P-element excision, P-lacW transposon, teashirt

77

Page 102: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

Materials and methods

Drosophila strains

All genetic markers and balancers are described by Lindsley and Zimm (1992) and

FlyBase (http://flybase.bio.indiana.edu) except as noted.

The hypomorphic allele tshA3-2-66 (P{lacW}A3-3-66; Bloomington stock 10842) have a

P{lacW} insertion at 1652 base pairs (bp) upstream the translation start of the teashirt (tsh)

gene and was identified after the screening of the Bloomington P-element insertions at the

38A6-40B1 cytogenetic region (Salazar and Gomes, 1999). The lethal P{lacW} insertion allele

tshA3-2-66 fail to complement the deficiencies Df(2L)TW161, Df(2L)305 and tsh8 allele .

P-element excision

For mobilisation of the P{lacW}A3-3-66 insert, w/w;T(2;3)CyOTM6,Tb/ P{lacW}A3-3-66;+

females were crossed to males carrying P{ry+, ∆2-3} on a Stubble (Sb) marked third

chromosome (Robertson et al., 1988) of genotype w/Y; T(2;3)CyOTM6,Tb/S Sp Bl bwD; ∆2-

3,Sb (Fig.1). All the strains used in the reversion scheme, carried X-chromosomes with the

eye-marker white1118 (w1118), and thus individuals carrying the P{lacW} chromosome (F2 w/Y;

T(2;3)CyOTM6Tb/P{lacW}A3-3-6) had coloured eyes due to the presence of the extra wild-type

copy w+ on the second chromosome, while lines having lost the P{lacW} (F2 w/Y;

T(2;3)CyOTM6Tb/∆P{lacW}A3-3-6) showed white eyes due to the white background on the X-

chromosome. The F1 males of genotype w/Y;P{lacW}A3-3-66/S Sp Bl bwD;+/∆2-3Sb were

crossed to females of genotype w/w;T(2;3)CyOTM6Tb/P{lacW}A3-3-66;+. 145 white-eyed males

(w/Y; T(2;3)CyOTM6Tb/∆P{lacW}A3-3-6) were found amongst 760 F2 males examined (Fig.1;

Table1). The 145 putative P-element excision chromosomes were then independently re-

balanced over T(2;3)CyOTM6Tb to establish P-element excision lines and test for viability and

fertility. For simplify, the F2 males were numbered in ascending order according to the order

that they have been gotten. The progeny of each F2 male correspond to a reversion line. Thus,

we analysed 760 lines of which 145 correspond to excision lines (∆P).

Staining of neuroblast with acetic orcein

For orcein acetic staining, the third instar larval brain were dissected in 0.7% NaCl and

fixed for 30 seconds in a 45% acetic acid and then transferred individually to be labelled in a

4% orcein in 45% acetic acid, for more 30 seconds. The brains were then washed in 45%

acetic acid and mounted in a drop of 4% orcein between slide and coverslip. The preparation

was squashed using filter paper and observed in the Zeiss Axioplan II microscope. Images

were acquired with a SPOT-2 camera (Diagnostics Instruments, Inc.) and processed with

Adobe Photoshop 6.0 (Adobe System).

78

Page 103: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

Figure 1- Diagram of the flies matting

during P-element excision mutagenesis.

tshA3-2-66 flies carry the P{lacW} chromosome, while

∆PtshA3-2-66 represents the loss of the P{lacW}

chromosome. ∆2-3 was used as the source of the

transposase. In F2, the male were individually

matted (each male was matted with two virgin

female).

Staining of ovaries

The ovaries were dissected in PBS and then fixed in 3,7% formaldehyde in PBS during

10 minutes (adapted from Ashburner, 1989). The ovaries were washed with PBST (PBS with

0,1% Triton X-100) during 1 hour and then the DNA was stained with 1µg/ml of Hoechst

33258 during 20 minutes. After 1 hour of washes with PBST, the ovaries were mounted in

Vectashield medium (Vectors). The preparations were observed in fluorescence using a Zeiss

Axioplan II microscope. Images were acquired with a SPOT-2 camera (Diagnostics

Instruments, Inc.) and processed with Adobe Photoshop 6.0 (Adobe System).

Wing Flies preparation

Wings from adult flies were dissected in 100% etanol and then transferred to xylene. In

a slide, the wings were placed in the top of a drop of Eukitt mounting medium (Electron

Microscopy Sciences). Finally, in the top of this was placed a coverslip and some weight during

the drying of EuKitt. The preparations were observed in the LEICA MZ FL III.

5’P and 3’P PCR amplification and contiguous regions of the PlacWA3-2-66

For tshA3-2-66 and in ∆P126, 5' and 3' P-element ends (5'P and 3'P) and the contiguous

regions of the P{lacW}A3-3-66 were PCR amplified. The 5’P was amplified with the primers:

ATTCAGTGCACGTTTGCTTG (5P_Fwd); TTTGGGAGTTTTCACCAAGG (5P_Rev);

AACGACGCATTTCGTACTCC (5P_Rev1); GCCTCTTCGCTATTACGCC (5P_Rev2);

TGGGCGCATCGTAACCGT (5P_Rev3) and GCAGGCTTCTGCTTCAATCAG (PlacW_1). The primers

5P_Rev2 and 5P_Rev3 are localise downstream the 5’P end of the P{lacW}A3-3-66 and in pair

79

Page 104: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

with the 5P_Fwd amplified part of the 5’P end region and the contiguous downstream region.

The 3’P end was amplified with the primers: AAGGATTTCCTTTGCCCAGT (3P_Fwd) and

TCCGTGGGGTTTGAATTAAC (3P_Rev).

Absence of re-integration of the P-element was confirmed by PCR.

Genomic DNA was isolated from L3 larvae homozygous for tsh126 allele after treatment

with a phenol/chloroform/isoamyl alcohol mixture to remove protein contaminants and

precipitation with 100% ethanol. The DNA was re-suspended in water for further

experimentation.

DNA sequencing of the ∆P99, ∆P104, ∆P123 and ∆P126 lines

Genomic DNA was isolated from L3 larvae homozygous for the ∆P99, ∆P104, ∆P123 and

∆P126 revertant lines after treatment with a phenol/chloroform/isoamyl alcohol mixture to

remove proteins contaminants and precipitation with 100% ethanol. The DNA was re-

suspended in water for further experimentation.

We sequenced and analysed 3913bp upstream of the translation start of tsh in the

∆P99, ∆P104, ∆P123 and ∆P126 lines for comparison. This region was amplified by PCR and

fragments were purified and sent for sequencing (GenomExpress). The following PCR primers

were used: 5’CACAAACATTGACACCTTGCC3’; 5’GACTACGCCGAACCGCCAA3’;

5’CCTTCTTTTGTCCTATTCCG3’;5’GCCAAATCTGAGCCTTAGAC3’;5’CCCAGCACCTCTTCTTTAGG3’;

5’CGTCACCCTTCTTTTGTCC3’;5’GCAGCCAAAAAGAAAAGTGG3’;5’TCGTCGTCACTACTCGAACG3’;5

’GTATGGTTAAATATTGTATATGCAA3’;5’CAAAATTTAGTTGCGAGGAGT3’;5’CGTTGTTGTTGTTTGTC

AGC3’ and 5’TTAGTTGGATTTTGGAACTTAAC. Sequences were compared and BLAST with the

tsh sequence reported in flybase (http://flybase.bio.indiana.edu) and the genomic sequence

that surround the insertion site of other transposable elements inserted in the same region

(P{RS3}CB-5359-3; P{XP}cl00773; P{lacW}l(2)k16406; P{XP}cl05049).

Results and Discussion

1-Excision Rate of the PlacW excision mutagenesis

The hypomorphic allele tshA3-2-66 (Bloomington stock 10842) was identified as a tsh

allele (tshA3-2-66) while we screened for P-element insertions at the 38A6-40B1 interval that fail

to complement the deficiencies Df(2L)TW161 and Df(2L)305, the tshNG1 and the tsh8 alleles

(Salazar and Gomes, 1999). We found that tshA3-2-66/tsh8 heterozygous were lethal, the tshA3-2-

66 homozygous dye at pupal stage and tshA3-2-66/Df(2L)TW161 also dye at pupal stage with very

few escapers.

After mobilizing the P element in tshA3-2-66, we isolated 145 F2 males lines with white

eyes. The screening for excision or modification of the transposon was based on the loss of the

80

Page 105: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

white+ eye colour marker. F2 males that had lost the P{lacW} and concomitantly had lost the

extra wild-type copy w+ on the second chromosome, shown in consequence white eyes due to

the white background on the X-chromosome. The 145 F2 males were obtained out of 760 F2

screened males (Table 1). This results points out the excision rate around 19% for that

mutagenesis.

Table1- Counting of F2 male that lost P-element based in the colour of the eyes.

Eyes colour F2 males coloured eyes 615 white eyes 145 Total 760

2-Classificatation based in the lethality of the 145 F2 P-excision lines in Trans

with the tshA3-2-66 allele

For better classified the F2 white eyes males, we grouped in class according with their

lethality in Trans with the tshA3-2-66 allele. Several broad categories could be defined on the

basis of their lethality. We obtained lines lethal at different developmental stages and lines

that give viable adults escapers in Trans with the tshA3-2-66 allele (Table 2). The majority of P-

jump out lines (69,7%) were lethal in larvae stage and should corresponds to internal

deletions within the P-element (P*). Probably, these P internal deletions include, at least, the

partial elimination of the white+ eye colour marker.

Table 2- Classification of the P-element excision line based in their lethality in Trans-heterozygous with tshA3-

2-66.

Viability F2 males Dead 11

Sterile 4

Embryo stage 20

Larvae stage 101

Leth

al

Pupae stage 2

Escapers 7

Total 145

In order to obtain alleles with abnormal mitotic phenotype, we randomly picked eight

L3 lethal lines and we stained the DNA of their homozygous neuroblasts with acetic orcein (Fig.

2). No abnormal mitotic figures were observed indicating that apparently none of these

81

Page 106: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

analysed lines represent a new mitotic tsh allele. In addition, the no detection of new mitotic

tsh allele suggest that the mitotic defects observed in tshA3-2-66 are due to the presence of the

P{lacW A3-2-66} in the predicted region of the tsh promoter.

Figure 2- No mitotic anomalies were detected in neuroblasts from

8 P-element excision lines L3 lethal.

L3 neuroblasts from (A, A’) Wild-type; (B, B’) homozygous ∆P6; (C, C’)

homozygous ∆P97; (D, D’) homozygous ∆P118; (E, E’) homozygous ∆P124; (F, F’)

homozygous ∆P131; (G, G’) homozygous ∆P132; (H, H’) homozygous ∆P137 and

(I, I’) homozygous ∆P139 stained with acetic orcein. All these ∆P lines are L3

lethal in Trans with tshA3-2-66 but, no mitotic anomalies were detected. The scale

bar represents 10µm.

Figure 3- No mitotic anomalies were detected in neuroblasts from

P-element excision lines lethal during pupae stage.

L3 neuroblasts from (A, A’) Wild-type; (B, B’) homozygous ∆P48 and (C, C’)

homozygous ∆P127 stained with acetic orcein. Both ∆P lines are pupae lethal,

when in Trans with tshA3-2-66. No mitotic anomalies were detected. The scale bar

represents 10µm.

The L3 neuroblasts of the two P-element excisions lines lethal at the pupal stage (∆P48

and ∆P127 lines) were also stained with acetic orcein (Fig. 3). No abnormal mitotic figures

were observed for this class of lines, similar to the results obtained for the randomly studies L3

lethal lines.

82

Page 107: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

3- Preliminary oogenesis characterization of the ∆P48/tshA3-2-66 and the

∆P127/tshA3-2-66 ovaries

To eliminate the possible contribution of the genetic background of the chromosome 3

to the lethality at the pupal stage, the ∆P48 and the ∆P127 lines (balanced over T(2:3) CyO

TM6 Tb e) were re-balanced over CyL. From re-balanced lines, we obtained a few ∆P48/tshA3-2-

66 and ∆P127/tshA3-2-66 escapers, showing that the change in the genetic background allowed

the development of some trans-heterozygous flies until adult stage. The preliminary analyses

of the ∆P48 and the ∆P127 ovaries showed strong oogenesis defects (Fig.4). The ∆P48/tshA3-2-

66 and the ∆P127/tshA3-2-66 ovaries resemble a big ovarian tumor. The ovaries seem an amount

of cells, where it was difficult to delimitate the germarium, none cellular types (germline and

somatic cells) were distinguished and none individualised egg chamber was identified, not even

S1 stage in the germarium. This suggests that oogenesis is severely affected in these lines. In

wild-type, pupae ovaries already presented a functional germarium and 2 or 3 egg chambers

perfectly formed in the vitelarium. The phenotype displayed by the ovaries from ∆P48 and

∆P127 revertant lines, indicates a premature oogenesis arrest, perhaps during ovaries

formation in pupa stage.

Figure 4- Adult ∆P48/tshA3-2-66 and ∆P127/tshA3-2-66 ovaries showed a very strong and early arrest

in oogenesis.

Ovaries from (A) wild-type, (B) ∆P48/tshA3-2-66 and (C) ∆127P/tshA3-2-66 in which, the DNA was stained with

Hoechst 33258. No individualised egg chambers were observed in lines ∆P48 and ∆P127 indicating that oogenesis

arrests before S1 stage in theses germarium. The scale bar represents 50µm and anterior is to the right and posterior

is to the left.

Mutations in fused, female sterile and narrow genes are known to produce ovarian

tumours but not as generalized as in ∆48/tshA3-2-66 and ∆127/tshA3-2-66 females, where all the

ovaries display a tumor-like phenotype.

83

Page 108: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

4- Preliminary characterization of the ∆P28, ∆P30, ∆P99, ∆P104, ∆P123,

∆P126 and ∆P128 lines

4.1- Based in viability and male/ female fertility

The seven F2 male showing viable escapers (in Trans with the tshA3-2-66) were classified

according to their fertility (Table 3). The ∆P28/tshA3-2-66 male and female were fertile, and

probably corresponds to a precise excision of P{lacW}. The other six lines showed female

sterile, apparently due to imprecise excisions of P{lacW}. In addition the ∆P30/tshA3-2-66 and

∆P99/tshA3-2-66 males were also sterile. In all the seven lines homozygous are lethal.

Table 3- Fertility of the seven P-element excision lines with viable escapers, in Trans with the tshA3-2-66.

Lines Female Male ∆P28 Fertile Fertile

∆P30 Sterile Sterile

∆P99 Sterile Sterile

∆P104 Sterile Fertile

∆P123 Sterile Fertile

∆P126 Sterile Fertile

∆P128 Sterile Fertile

We tested the viability and fertility of the trans-heterozygous of the ∆P30, ∆P99,

∆P104, ∆P123, ∆P126 and ∆P128 lines combined between them (table4). Curiously, trans-

heterozygous females resultant from the combination of the ∆P99 with the other ∆P104,

∆P123, ∆P126 and ∆P128 lines were viable and sterile, while combinations between the last

other four lines, produce trans-heterozygous lethal before achieved the adult stage. (1) This

behaviour of the trans-heterozygous ∆P/∆P (table 4) and (2) the proximity of the timing when

the F2 male were obtained (illustrated by the proximity between the number of these lines)

make us suspect that ∆P30, ∆P99 revertant lines correspond to an event while, ∆P104, ∆P123,

∆P126 and ∆P128 revertants lines correspond to other independent event. In addition, the

preliminary ovaries phenotype characterization, no revealed significant differences between

ovaries from the 6 lines (30, 99, 104, 123, 126 and 128; data not show): all these female are

sterile: fail to lagg eggs and present atrophied ovaries (Fig.6E), where oogenesis is arrested

before S3 stage (for details see chapter II part II of this manuscript).

All the seven lines produce homozygous lethal. The existence of ∆P28/tshA3-2-66 viable

and fertile, suggested that the lethality observed in the homozygous of the revertant lines is

due to other mutation in chromosome 2, which occurred during P excision mutagenesis and

was not present in the initial chromosome 2 of the tshA3-2-66 allele.

84

Page 109: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

Table 4- Fertility of the progeny (female) resultant from the combination between the seven P-element

excision lines with viable escapers.

∆P30 ∆P99 ∆P104 ∆P123 ∆P126 ∆P128

∆P28 nd nd nd nd nd nd

∆P30 L nd nd nd nd nd

∆P99 nd L L L L nd

∆P104 nd VS L L L nd

∆P123 nd VS L L L L

∆P126 nd VS L L L L

∆P128 nd nd nd L L L

F= fertile; L= lethal; nd= not determined; S= sterile and V= viable

4.2- Confirmation of the P{lacW} excision by PCR (tested for ∆P126)

In order to confirm the P{lacW} excision from the ∆P126, we designed primers insight

the P{lacW}: the forward prime was designed in 5’P region and the three reversed primers

(5P-Rev2, 5P-Rev3 and PlacW_1) were designed downstream at 639bp (119bp downstream

5’P),at 811bp (290bp downstream 5’P) and at 1498bp (978bp downstream 5’P), respectively.

These regions were amplified in tshA3-2-66 but not in ∆P126, as expected, apparently indicating

the absent of the PlacW from the ∆P126 genome (Fig.5B).

Since, sometimes during the excision of a P element, this one could undergo some

rearrangements and then re-integrates the genome, we tested the presence of 5P and 3P

regions in ∆P126 genome, for note, these regions have to be intact to allow the P-element re-

integration.

A fragments of 217 bp within the 5’P and another of 211 bp within 3’P regions of the

P{lacW} was amplified by PCR. Unexpectedly, we obtained DNA amplification from the tshA3-2-

66 and the ∆P126 DNA with the pair primers designed into the 5’P and 3’P region of the

P{lacW} (Fig.5A, C and D, respectively). These fragments was then sequence and no

modification was detected comparatively to the reported in flybase database

(http://flybase.bio.indiana.edu; data not show). This suggests that, the ∆P126 allele conserved

in their genome intacts 5’P and 3’P sequences. This could be a result (1) of the duplication of

P-element extremities during an ancient P-element excision. (2) In other hand, ∆P126 could

have a residual P-element. This residual P-element could be the PlacW which, after excision

from tsh locus went through a rearrangement and, it was re-integrated in another region of

the ∆P126 genome. (3) Other hypothesis can be an insertion of other P-element (beyond the

PlacW) already present in tshA3-2-66 genome when the P-element excision mutagenesis was

performed. (4) Finally, since we only amplified 217 bp of the 5P regin and 211 bp from de 3P

85

Page 110: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

region, the 5P and 3P regions in ∆P126 could have a mutation in the non sequenced region,

indicating that this is a defective P-element. Only in this last case, we could exclude completely

the possibility of a PlacW re-insertion during P-excision mutagenesis.

Some of these assays were performed with DNA from other ∆P lines with the same

results. However, we choose to present here only the results obtained with ∆P126 DNA (the

justification for this choice is explained below in the item 5.2 of this part).

Figure 5- Amplified and sequenced regions of the PlacW in tshA3-2-66 and ∆P126.

(A and B) PlacW fragments amplified and migrated in 0,7% agarose gel at 100 volts. (M) 1Kb Ladder DNA marker;

water was used as a control.

(A) ∆P126 and tshA3-2-66 DNA amplification of 512 bp fragment of the 5 Prime region using the 5P-Fwd and the 5P-Rev1

primers pair.

(B) In the left of the marker (M), ∆P126 and tshA3-2-66 DNA amplification of the 639bp fragment (521 bp from 5P region

and 118bp of the contiguous downstream region) using the 5P-Fwd and the 5P-Rev2 primers pair. In the right of the

marker (M), ∆P126 and tshA3-2-66 DNA amplification of the 811bp fragment (521 bp from 5P region and 290 bp of the

contiguous downstream region) using the 5P-Fwd and the 5P-Rev3 primers pair. No amplification was obtained for

∆P126 DNA using the combination of the primer that amplified 5P region and the contiguous downstream PlacWA3-2-66

DNA.

(C and D) 5’P ends and 3’P ends fragments amplified by PCR and migrated in 1,5% agarose gel at 100 volts. (M) 100

bp Ladder DNA marker; for control was used water.

(C) ∆P126 and tshA3-2-66 DNA amplification of 217 bp of the 5 Prime region using the 5P-Fwd and the 5P-Rev primers

pair;

(D) ∆P126 and tshA3-2-66 DNA amplification of 211 bp of the 3’P ends region using the 3P-Fwd and the 3P-Rev primers

pair. Both regions were amplified with ∆P126 and tshA3-2-66 DNA.

86

Page 111: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

4.3-Sequence of the upstream region contiguous to the tsh mRNA and allelism

tests (tested in ∆P99, ∆P104, ∆P123 and ∆P126)

We sequenced and analysed 3913bp upstream of the translation start of tsh in ∆P99,

∆P104, ∆P123 and ∆P126 lines for comparison with the tsh sequence published in FlyBase

(http://flybase.bio.indiana.edu) and the genomic sequence that surrounds the insertion site of

other transposable elements inserted in the same region (P{RS3}CB-5359-3; P{XP}cl00773;

P{lacW}l(2)k16406; P{XP}cl05049). We were unable to identify any mutation. Instead, we

identified a 6 nucleotide sequence (AACTGC) located 1297bp upstream of the translation start

site, which is absent from the current release of the Drosophila melanogaster genome but

present in all sequences that we analyzed (Supplementary Fig. 1).

In the absence of a mutation identified, we cross the ∆P126 with some reported tsh

alleles in order to found a combination that give a sterile female phenotype, resemble the

trans-heterozygous ∆P126/tshA3-2-66 (table 5). To simplify, we only presented the allelism test

performed with the ∆P126 line, but the results was the same for the other ∆P lines.

Unfortunately, behind the tshA3-2-66, all other combinations with ∆P126 give viable and fertile

adult, disabling us to demonstrate that ∆P126 is a new tsh allele.

Table 5- List of the tsh alleles crossed with ∆P126 and tshA3-2-66 to test for the viability and fertility of the

adult female.

Alelle Characterization Viability/Fertility in Trans with:

References

Bloomington stock

symbol P-element insertion ∆P126 tshA3-2-66

167 Df(2L)TW161 nd L

Df(2L)305 VF L induced by B. Wakimoto

tsh 8 --- VF L Fasano et al., 1991

tsh 2757 R7 --- VF L Andrew et al., 1994

tsh NG1 --- VF L Salazar and Gomes, 1999

10842 tshA3-2-66 1652 bp upstream ATG VS L

1018 P{A92}tsh[i71] VF VF

1296 P{WA[R]}tsh[4-3] VF VF

3040 P{GAWb}tsh[md621] VF VF

11178 P{lacw} l(2)K16406[K16406] 1317 bp upstream ATG VF nd

11370 P{PZ}tsh[04319] 720 bp upstream ATG VF nd

F= fertile; L= lethal; nd= not determined; S= sterile and V= viable

Our sequence data and allelism results, doesn’t allow us to confirm that ∆P99, ∆P104,

∆P123 or ∆P126 are a new tsh allele, however, in the Trans with tshA3-2-66, we observed other

morphologic defects that could be co-related with a diminution of Tsh.

87

Page 112: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

5- Other morphologic defects observed in ∆P30/tshA3-2-66, ∆P99/tshA3-2-66,

∆P104/tshA3-2-66, ∆P123/tshA3-2-66, ∆P126/tshA3-2-66 and ∆P128/tshA3-2-66 flies co-

related with a diminution of Tsh expression

5.1-Wings defects

All Trans-heterozygous viable female for ∆PlacW/tshA3-2-66 behind the oogenesis defects

show difficulties in to move their wings. In addition, their wings were smaller than wild-type,

and sometimes it was possible to observe ectopic veins (Fig.6). This wing phenotype could be

related with some tsh deregulation. For note, one of the reported tsh loss of function

mutations cause an abnormal wing posture phenotype called aeroplane characterized by a

wings paralysis among other defects (Soanes and Bell, 1999 ; Soanes et al., 2001; Soanes et

Bell, 2001).

Figure 6- Morphologic aspects of

DP126/ tshA3-2-66 flies.

(A) Wild-type female Oregon-R with red eyes

(arrowhead) and ∆P/ tshA3-2-66 female with orange

eyes (arrow) due to the presence of the white

background on the X-chromosome and a extra wild-

type copy w+ on the second chromosome (tshA3-2-66

allele). (B) Drosophila eyes from W;tshA3-2-66/CyO

(left side) and W;∆P126/tshA3-2-66∆126 (right side)

adults. ∆P126/tshA3-2-66 have white eyes, despite the

presence of one copy of the P{lacW}A3-3-66

responsible for the orange eyes phenotype of the

W;tshA3-2-66/CyO female.

(C) Wing from wild-type female and (D) wing from

∆P126/tshA3-2-66 female. The dotted in (C) represents

the contours of the wild-type wing that doesn’t

coincide with the contours of the ∆P126/ tshA3-2-66

wing. ∆P126/ tshA3-2-66 wings are a little smaller and

showed sometimes ectopic veins (arrow in panel D).

(E) wild-type ovaries and (F) ∆P126/ tshA3-2-66

ovaries from female with the same age and grow at

the same temperature. The ∆P126/tshA3-2-66 ovaries

are atrophied and smaller than wild-type.

88

Page 113: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART I

5.2-Reduction in eyes colour patterning

In the sterile female line, white was expressed as previously reported for other P{lacW}

inserted in the tsh locus (Fig. 6B; Sun et al., 1995; Bhojwani et al., 1995; FL unpublished

data). The tshA3-2-66 adults show the anterior half of the eye coloured, reporting the tsh

expression eye domain, in virtue of the presence of the mini-white in the promoter of the tsh

gene. The carefully observation of the ∆P/tshA3-2-66 adults eyes revealed a slight reduction in

the coloured eye domain, suggesting that tsh expression is reduced in the eyes of these

mutants (Fig. 6A).

During the analyses of the tsh alellism, and in order to eliminated the possible genetic

interaction between tsh gene and the CyO (Curly of oyster) balancer, the tshA3-2-66 and the

∆P126 chromosomes were balanced with the Cy L (curly lobe) balancer. From the cross

between w;tshA3-2-66/CyL female and the w; ∆P126 /CyL male we obtained 2/3 of the w; ∆P126/

tshA3-2-66 adults with white eyes, indicating that ∆P126 modifies the patterned white expression

observed in the tshA3-2-66 adult eyes (Fig. 6B). As the expression patterns of white may reflect

the activity of regulatory elements belonging to the tsh gene, we hypothesised that the ∆P126

chromosome could bare a mutation in the tsh locus even if we were incapable of identify it

(Sun et al., 1995; Bhojwani et al., 1995; FL unpublished data).

In conclusion, we fail to clearly prove that we isolated a new tsh allele (∆P126), but we

can argue that ∆P126/ tshA3-2-66 display a phenotype co-related with a diminution of Tsh

suggesting that Tsh expression is affected in this revertent line. These observations, prompted

us to investigate whether the w; ∆P126/ tshA3-2-66 female sterile phenotype could be associated

to an altered expression of tsh in the ovaries (for details see chapter II part II of this

manuscript). Subsequently, the ∆P126 line will be referred as ∆126.

For note, the mutagenesis data display in part II of this chapter included the results

described above in this mutagenesis chapter and the results obtained during a independent

mutagenesis performed by Salazar (Salazar, 2000) in order to ensure that the lethality of the

tshA3-2-66 allele was directly attributable to the P{lacW}A3-3-66 insertion (for resume see

supplementary table 1).

89

Page 114: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Supplementary Data

Supplementary table 1- PlacW excision results obtained in two independents P-excision mutagenesis from

the tshA-3-2-66 allele by Salazar (2000, refered in chapter IV of the results of this manuscript) and by Pimentel

(presented in this chapter).

Salazar data Pimentel data Total

Males F2 analysed 1622 760 2382

Revertant line with escapers 10 7 17

Escapers viable 6 1 7

Escapers with sterile female 4 6 10

Page 115: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_Part II:

Page 116: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

92

Page 117: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Regulation of cell proliferation and patterning in Drosophila

oogenesis by Hedgehog signalling depends on teashirt

Silvia Pimentel1,2, Isabel Salazar1, Christine Vola2, Rui Gomes 1 and Laurent Fasano2†

1 Universidade de Lisboa, Faculdade de Ciências, Departamento de Biologia Vegetal,

Campo Grande, 1749-016 Lisboa, Portugal 2 Institut de Biologie du Développement de Marseille Luminy (IBDML), UMR 6216, CNRS-

Université de la Méditerranée, Campus de Luminy, Case 907, 13288 Marseille Cedex 09,

France.

†Authors for correspondence: (emails: <[email protected]>)

Page 118: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal
Page 119: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

Abstract

Interaction between intrinsic factors with extrinsic signals plays a central role to control

stem cell self-renewal, proliferation and differentiation. In Drosophila ovaries, Hedgehog (Hh)

signalling is essential for self-renewal and proliferation of follicle stem cells (FSC). Here, we

show that Teashirt (Tsh) is co-expressed with Hh at the tip of Drosophila germarium, in

terminal filament cells (TFs) and in cap cells (CC). The latter are a subset of hh-expressing

cells located adjacently to germline stem cells (GSC) and are described as a niche for GSC and

FSC. We demonstrated that overexpression of tsh mimics the phenotype associated with hh

overexpression. Phenotypic characterisation of ovaries into which tsh expression was perturbed

revealed an early oogenesis arrest (stage S2/S3 of oogenesis) and a perturbed expression of

hh and engrailed (en). In these mutant ovaries, ectopically provided Tsh partially rescued the

defects and restored hh and en expression. Here we explore for the first time the expression

and the function of tsh during oogenesis. Our data suggest that in adult ovaries tsh expression

might be required to regulate hh expression, contributing for the regulation of FSC proliferation

and the specification of somatic cell identity.

Key words: Drosophila, oogenesis, follicle stem cells, follicle cells, teashirt, hedgehog

95

Page 120: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

Introduction

Drosophila oogenesis represents an excellent model system for studying how

proliferation and patterning of both germline and somatic cell lineages are controlled by

signalling pathways. It produces ovarian follicle continually throughout adult life and it involves

a high degree of coordination between the proliferation and differentiation programs of two

lineages cells, soma and germ line (Narbonne-Reveau et al., 2006).

Each of the two ovaries in adult fly typically contains 16 ovarioles. Eggs chambers are

made up within the germarium, a distinct anterior region of every ovariole. Egg chambers

leave the germarium and mature as they move posteriorward within the ovariole. They

develop through successive stages to form a polarized oocyte, surrounded by a patterned

epithelium of somatic follicle cells (Zhang and Kalderon, 2000; Lin and Spradling, 1993).

In the germarium, three types of stem cells have been identified: the germline stem

cells (GSC), the follicle stem cells (FSC) and the escort stem cells (ESCs) (Fig. 1A) (Decotto

and Spradling, 2005; Lin and Spradling, 1993; Margolis and Spradling 1995; Wieschaus and

Szabad, 1979). The apical portion of the germarium (region 1) contains 2-3 GSC identified by

clonal analysis (Wieschaus and Szabad, 1979) and laser ablation studies (Lin and Spradling,

1993). At the anterior end of the germarium, the terminal filament cells (TFs) and the cap cells

(CC), which are both none dividing somatic cells, support the GSC niche. The CC directly

contact with GSC by adherent junctions enriched in DE-cadherin and Armadillo (β-catenin)

(Song et al., 2002). CC signals for stem cell (SC) maintenance, regulation of proliferation and

differentiation of the SC progeny (Xie and Spradling, 1998; Song et al. 2004; Cox et al.,

2000). GSC divide asymmetrically every 20 hours to produce a SC and a daughter cystoblast.

The differentiating cystoblast undergoes four rounds of mitosis with incomplete cytokinesis, to

generate a 16-cell cyst (kai et al., 2005). One of the 16 cyst cells becomes the oocyte while

the other 15 cells differentiate as nurse cells (reviewed in King, 1970). The escort stem cells

(ESCs) were identified as the most anterior cells until that time known by inner germarial

sheath (IGS) cells that directly contacts CC and surrounds GSC with their cellular processes

(Decotto and Spradling, 2005). Their progeny, the squamous escort cells (ES) are responsible

to enclose the cystoblasts in a thin covering and help him to move posterior (Fig. 1A). At the

junction between region 2a and region 2b, cysts are forced into single file, lose their escort cell

covering, which by turn enter in apoptosis, and begin to acquire a follicular layer.

In the region 2a/2b, two somatic follicle stem cells (FSC) divide, in average every 10

hours, to give rise to all polar cells, stalk cells and main body cells needed to form the

epithelial cell-like follicle cells (FC) that cover up each cyst and mold them into a “lens shape”

characteristic of region 2b. Under the influence of continued somatic cell growth, cysts and

their surrounding cells round up, enter in region 3 (also known as stage 1), and bud off from

the germarium to form individual egg chambers connected to their neighbours by 5-7 stalk

96

Page 121: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

cells (Kai et al., 2005; Nystul and Spradling, 2010). Before leaving the germarium, FC undergo

approximately five rounds of mitotic division. Three to four mitotic cycles later, in stage 6,

division cease following three endocycles (from stage 7-10) to generate a follicle’s monolayer

of around 650 polyploid FC comprising multiple subtypes (Nystul and Spradling, 2010)

Previously, clonal analysis has revealed that the FC are already divided into two

populations in the germarium (Besse and Pret, 2003). One lineage gives rise to the stalk cells

and the polar cells, which are a pair of follicle cells at each end of the egg chamber that

connects to the stalk (Tworoger et al., 1999; Lopez-Schier and Johnston, 2001). The rest of

the follicle cells derive from a separate lineage and form the epithelium around the cyst; they

are called epithelial follicle cells or main-body. Nystul and Spradling (2010) by examining

clones induced at low frequency, shown recently that FC types specification is independent of

cell lineage. Instead, FC are multipotent prior to polar or stalk specification. This fits well with

recent studies showing that many additional cells in the germarium can be induced to take on

a polar cell fate by strong Notch signalling, while high levels of JAK-STAT signalling can induce

more stalk cells (Assa-Kunik et al. 2007). In addition, they map the specification of the first

anterior and posterior polar cells, when cysts are associated with 8-16 FC (in mid to late region

2b), what is in agreement with previous studies (Margolis and Spradling, 1995; Besse and

Pret, 2003) which found that polar cells were first specified at the 14-cell stage.

The two distinct populations of stem cells, FSC and GSC, work in a coordinated fashion

to efficiently produce egg chambers and share some molecular mechanisms to control self-

renewal and proliferation (Song and Xie, 2002). Despite these similarities, it is worth noting

that the organization of the FSC niche is different from that of the GSC niche. The FSC niche

includes not only neighbouring posterior EC but also TF cells and CC located over a few cells

away. As a consequence, proliferation and self-renewal of GSC and FSC depend on different

combination of niche signals, but both depend on Hedgehog (Hh) signalling (for a review see

Kirilly and Xie, 2007).

In adult Drosophila ovaries, Hh is detected in TF cells and CC (Fig. 1C). Hh functions as

a long-range signal to control FSC self-renewal, proliferation and budding of egg chambers

(Forbes et al., 1996a; Forbes et al., 1996b; King et al., 2001; Zhang and Kalderon, 2001). Hh

signalling is essential for self-renewal and proliferation of the FSC, while it plays a non-

essential role in maintaining the GSC, by having redundant function with the piwi–mediated

pathway (King et al., 2001). Hh acts by binding to its receptor, Patched (Ptc), a

transmembrane protein that normally acts to restrict the activity of Smoothened (Smo), a

seven-transmenbrane domain protein (Hooper and Scott, 2005). Upon binding of Hh to Ptc,

Smo becomes active, ultimately leading to the activation of the transcription factor, Cubitus

interruptus (Ci), which in turn regulates the expression of Hh target genes (Ingham, 1998;

Aza-Blanc and Kornberg, 1999). Disruption of the Hh signalling results in rapid FSC loss and

gives rise to compound egg chambers that contain multiple germline cysts in a single follicle

97

Page 122: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

epithelium (King et al., 2001; Zhang and Kalderon, 2001). Hyperactivation of Hh signalling

increases the number of FSC, causes the overproduction of somatic FC that accumulates

between egg chambers, induces mis-positioning of oocyte within egg chambers and generates

ectopic polar cells (Forbes et al., 1996a; Forbes et al., 1996b; Liu and Montell, 1999; Zhang

and kalderon, 2000). It is therefore postulated that Hh signalling in the germarium regulates

somatic FC proliferation and contributes to oocyte positioning.

The Teashirt (tsh) gene was first identified in Drosophila as a region-specific homeotic

gene specifying embryonic trunk identity (Fasano et al 1991; Röder et al 1992; Andrew et al

1994; Taghli-Lamallem et al 2007). The zinc-finger protein Teashirt (Tsh) was also

characterized as one of the factors necessary for Hh target gene regulation and biochemical

approaches suggested that Tsh and Ci interact directly in protein complexes (Gallet et al.,

2000; Angelats et al., 2002). Based on sequence homology, mouse tsh-like genes (Tshz1-3)

were identified and, their specific expression patterns raised the possibility that they may play

developmental roles in mammals (Caubit et al. 2000 and 2005). Indeed, each substitutes for

fly tsh in genetic rescue experiments (Manfroid et al 2004). Recently, we reported that Tshz3

plays a key role for ureteral smooth muscle differentiation downstream of Sonic Hedgehog

(Caubit et al., 2008).

In this study, we characterize for the first time the expression of tsh during oogenesis

and our data suggest that the expression of tsh might be necessary for controlling hh

expression in ovary and, as a consequence, for controlling the follicle stem cells proliferation.

Materials and Methods

Genetics

The enhancer trap tsh1 (Fasano et al., 1991) was used to perform X-Gal staining.

The lethal P{lacW} insertion allele tshA3-2-66 (P{lacW}A3-3-66) was identified after the

screening of the Bloomington P-element insertions at the 38A6-40B1 cytogenetic region. The

P{lacW}A3-3-66 insertion was mapped 1652 base pairs (bp) upstream the translation start of the

teashirt (tsh) gene. Northern blot analysis revealed that tshA2-3-66 causes a significant reduction

on the tsh transcript levels (data not shown). We found that tshA3-2-66/tsh8 heterozygotes were

lethal, tshA3-2-66/tshA3-2-66 all dye at pupal stage and tshA3-2-66/Df(2L)TW161 also dye at pupal

stage with very few escapers.

We performed a P-element excision mutagenesis from the tshA3-2-66 allele. The

endogenous white locus was the w1118 allele. Precise and imprecise excisions (E{P}) of the

transposon were induced in the presence of the Sb P[ry+ 2-3] source of transposase

(Robertson, et al., 1988). E{P}/CyO P-element-excised revertants can be distinguished by

their white eye color and were tested with the original P{lacW}A3-3-66mutant lines. Analysis of

98

Page 123: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

2382 males allowed us to identify 17 lines that gave escapers when in Trans with tshA3-2-66: out

of 17 revertants, seven were fully viable and fertile and the other ten were also viable but

female sterile (two were male sterile).

Absence of re-integration of the P-element was confirmed by PCR with specific primers

designed for the 5’P of the P{lacW}; tshA3-2-66 was used as a positive control for the PCR

conditions.

The revertant line ∆126 and the tshA3-2-66 allele were balanced over T(2;3)CyOTM6B,Tb.

Females (or males) tshA3-2-66/T(2;3)CyOTM6B,Tb were crossed with male (or females)

∆126/T(2;3)CyOTM6B,Tb to get ∆126/tshA3-2-66 females used in this study.

All genetic markers and balancers are described by Lindsley and Zimm (1992) and

FlyBase (http://flybase.bio.indiana.edu) except as noted.

Rescue of ∆126 phenotype

For rescue and overexpression experiments the constructs UAS-tsh13 (Gallet et al.,

1998) and UAS-hh-M4 (Ingham and Fietz, 1995) were crossed to the driver P{w+mC=GAL4-

Hsp70.PB}TR2 (Bloomington).

DNA sequencing of the ∆126 revertant

Genomic DNA was isolated from L3 larvae homozygous for ∆126 allele after treatment

with a phenol/chloroform/isoamyl alcohol mixture to remove protein contaminants and

precipitation with 100% ethanol. The DNA was re-suspended in water for further

experimentation.

We sequenced and analysed 3913bp upstream of the translation start of tsh in ∆126

revertant. This region was amplified by PCR and fragments were purified and sent for

sequencing (GenomExpress). The following PCR primers were used:

5’CACAAACATTGACACCTTGCC3’;5’GACTACGCCGAACCGCCAA3’;5’CCTTCTTTTGTCCTATTCCG3’;

5’GCCAAATCTGAGCCTTAGAC3’;5’CCCAGCACCTCTTCTTTAGG3’;5’CGTCACCCTTCTTTTGTCC3’;5’

GCAGCCAAAAAGAAAAGTGG3’;5’TCGTCGTCACTACTCGAACG3’;5’GTATGGTTAAATATTGTATATGC

AA3’;5’CAAAATTTAGTTGCGAGGAGT3’;5’CGTTGTTGTTGTTTGTCAGC3’ and

5’TTAGTTGGATTTTGGAACTTAAC. Comparison of the ∆126 sequence with tsh sequence

published in FlyBase (http://flybase.bio.indiana.edu) and the genomic sequence that surrounds

the insertion site of other transposable elements inserted in the same region (P{RS3}CB-

5359-3; P{XP}cl00773; P{lacW}l(2)k16406; P{XP}cl05049) did not allowed us to identify any

mutation. Instead, we identified a 6 nucleotide sequence (AACTGC) located 1297bp upstream

of the translation start site, which is absent from the current release of the Drosophila

melanogaster genome but present in all sequences we analyzed.

99

Page 124: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

Staining ovaries

β-galactosidase activity staining was performed as described previously (Fasano and

Kerridge, 1988), but ovaries were fixed with 2.5% Formaldehyde in PBS for 10 minutes. X-Gal

stained ovaries were analysed under an Axiophot Zeiss microscope.

Antibody staining was performed based on the protocol described by Zhang and

Kalderon (2000), with the following modifications: ovaries were fixed in 1:3 heptane: 10%

DMSO and 4% paraformaldehyde in PBS for 20 minutes. The ovaries were blocked in TNB

(blocking reagent from the Tyramide signal amplification kit (PerkinElmer) for 1hour then

incubated with the primary antibody at 4°C, overnight.

Primary and secondary antibodies and their dilutions were as follows: anti-Tsh (rabbit

Tsh, a gift from S Cohen, EMBL Heidelberg, 1:1000 dilution); mouse 4-D9 anti-Engrailed

invected (Developmental Studies Hybridome bank, 1:10); anti-Hh (rabbit anti-serum against

Hh, a gift from A. Gallet, Nice University, France, 1:200); mouse anti-Arm (Developmental

Studies Hybridome bank, 1:5);mouse 7G10 anti-Fasciclin III (Developmental Studies

Hybridome bank, 1:200); rabbit anti-Vasa (a gift from R. Lehmann 1:1000); rabbit anti-

phospho-Histone H3 (Upstate Biotech, 1:500); rat anti-BrdU (abcam, 1:100); and rabbit anti-

Caspase 3 (Cell Signalling Tecnology; 1:200) Secondary are conjugated with Texas red and

FITC (Molecular Probes, 1:500).

For En and Hh staining, the signal was amplified with the Tyramide Signal Amplification

Kit (PerkinElmer) using biotinylated secondary antibodies (Jackson Laboratories, 1:500). In

these cases, the ovaries were submitted to a peroxidase treatment (3% H2O2 for 10 minutes)

to reduce the background before their blocking.

Stained ovaries were mounted in Vectashield medium with DAPI (Vectors) and analyzed

under a LSM 510 Zeiss confocal microscope.

BrdU incorporation

Follicle cells were labelled with BrdU, as described previously (Lilly and Spradling, 1996;

Shcherbata et al., 2004) with slight modifications. Ovaries from 7 days old female were

dissected in Grace’s insect medium and then incubated with 10µm BrdU (Upstate Biotech) in

the same medium for 15 minutes. To stop BrdU incorporation, ovaries were washed twice with

fresh Grace’s insect medium and then maintained in these medium for 90 minutes or 270

minutes at room temperature. The ovaries were then fixed in 3.7% Formaldehyde during 20

minutes, washed with PBST (PBS with 0.1% Triton X-100), then treated with 2N HCl for 30

minutes. Neutralization was performed in 100mM Borax for 2 minutes. Tissues were rinsed

with PBST three times (10 minutes), blocked and stained as described above.

100

Page 125: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

Rescue of ∆126/ tshA-3-2-66 mutant phenotype

∆126; UAS-tsh13/T(2;3)CyOTM6B,Tb or ∆126; UAS-hh-M4/T(2;3)CyOTM6B,Tb males

were crossed to tshA-3-2-66; hsGal4/T(2;3)CyOTM6B,Tb females. In the day of the eclosion,

hsGal4>UAS-tsh13; ∆126/ tshA-3-2-66 or hsGal4>UAS-hh-M4; ∆126/ tshA-3-2-66 females were

submitted to a heat-shock: tubes were immersed in a water bath at 37°C, for 1hour, twice a

day, in 3 consecutive days. The 4th day, ovaries were dissected and stained with FasIII and

Vasa antibodies. Females were grown at 18°C or at 25°C, until the adult stage and during the

period between heat-shocks.

Tsh and Hh overexpression

UAS-tsh13/T(2;3)CyOTM6B,Tb or UAS-hh-M4/T(2;3)CyOTM6B,Tb males were crossed

to hsGal4/T(2;3)CyOTM6B,Tb females. In the day of the eclosion, hsGal4>UAS-tsh13 or

hsGal4>UAS-hh-M4 females were heat-shocked at 37°C, for 1hour, twice a day, in 3

consecutive days. In the 4th day, ovaries were dissected and stained with FasIII and Vasa or,

Tsh and En antibodies. Females were grown at 25°C, until adult stage and between heat-

shocks.

Results

1-Tsh is expressed in Escort cells and co-expressed with En in Terminal

Filament cells and Cap cells

To analyse expression of the tsh gene in ovaries, we first took advantage of the

enhancer trap line tsh-LacZ, which was shown to faithfully reproduce the pattern of the

endogenous tsh gene in embryos and imaginal discs (Fasano et al., 1991). ß-galactosidase (ß-

gal) activity was detected in terminal filament (TF) and cap cells (CC) at the tip of the adult

germarium (Fig. 1B). Thus, in adult ovary, Tsh expression pattern resembles expression

patterns previously reported for Hh (Fig. 1D; Forbes et al., 1996a). To better characterise the

Tshz-positive cell population, we next performed double immunostaining for Tsh and Engrailed

(En). Expression of the Tsh protein was confirmed in the nuclei of TF and CC where it

colocalizes with En (Fig. 1D-D’). Unlike En, Tsh was also strongly detected in a group of

somatic cells, in region 1, 2b, and in the boundary of the 2a/2b region of the adult germarium

(Fig. 1D-D’), The location and the morphology of theses cells let us think that they are

probably the escort cells (EC) and the posterior EC (Fig. 1D arrow), respectively, which

participate in FSC niche (Kirilly and Xie, 2007). In addition, low level of Tsh was observed in

recently produced FC that surround germline in region 3 of the germarium (stage S1) and,

sometimes, this weak staining was still detectable in FC belonging to S2 egg chambers. Tsh

101

Page 126: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

was never observed in germline cells. As flies age, Tsh staining becomes weaker in the apical

half of the TF, but remains high in more proximal TF cells and CC, especially in flies older than

9 days. This fading coincides with the condensation of the CC and some proximal TF cells into

a cluster within the germarium tip (data not shown).

Figure 1- en co-localises with a tsh-

subset domain in Drosophila ovaries.

(A) Diagram of the Drosophila germarium and the

location of stem cells (in Kirilly and Xie, 2007). The

four sub-regions of the germarium are indicated

below the diagram. (B) X-Gal staining (blue) of tsh1

adult ovariole. (B’) Close up of the anterior end of

the germarium shown in B.

Germarium of late pupae (C and C’), and 7-day old

adult (D and D’) stained for Tsh (green), En (red)

and nuclei (DAPI, blue). (C and C’) Tsh positive and

En negative CC are observed in germaria from late

pupae (arrow). (D) In adult, Tsh is strongly detected

in the terminal filament cells (TF), the cap cells (CC;

arrowhead), the escort cells (EC) including escort

stem cells (ESC) and the posterior EC (arrow). (C’

and D’) Staining for Tsh, in the same confocal plane

as in C and D, respectively. In all panels, anterior is

to the left. In B and C, the scale bars represent

20µm and is the same in panels C-D’. CB, cystoblast;

CC, cap cell; DC, 16-cell developing cyst; ESC, escort

stem cell; FC, follicle cell; GSC, germline stem cell;

IGS (EC), inner germarial sheath cell, or escort cell;

SS, spectrosome; SSCs, somatic follicle stem cell,

(also know by FSC).

To investigate the onset of tsh expression in the germarium, late pupal gonads were

double stained with anti-En and anti-Tsh. As in the adult ovaries, Tsh and En were co-

expressed in TF and in CC precursors (Fig. 1C-C’) but Tsh was absent from the EC and newly

born FC. The presence of Tsh positive and En negative CC precursors suggests that Tsh

expression precedes En expression (Fig. 1C-C’).

2- teashirt expression is perturbed in P{lacW}A3-3-66∆126/tshA-3-2-66 ovaries

The cytogenetic location of tsh (40A) has precluded the analysis of tsh function during

oogenesis by germline clone analysis. To analyze the function of tsh in the adult ovary we

sought after female sterile alleles of tsh. To this aim, we first screened for P-element insertions

102

Page 127: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

at the 38A6-40B1 interval that fail to complement the deficiencies Df(2L)TW161, Df(2L)305

and the tshNG1 and tsh8 alleles (Salazar and Gomes, 1999; GR unpublished data). Based on this

criteria, we selected the lethal P{lacW}A3-3-66 (tshA3-2-66). Moreover, in this line white was

expressed as previously reported for other P{lacW} inserted in the tsh locus (Fig. 2A) (Sun et

al., 1995; Bhojwani et al., 1995; FL unpublished data).

Figure 2- Expression level variation

of tsh in ∆126/tshA3-2-66 ovarioles.

(A) Drosophila eyes from W;tshA3-2-66/CyO (left

side) and W;∆126/tshA3-2-66∆126 (right side)

adults. ∆126/tshA3-2-66 have white eyes, despite

the presence of one copy of the P{lacW}A3-3-66

responsible for the orange eyes phenotype of

the W;tshA3-2-66/CyO female.

(B-D') ovaries were stained for Tsh (green) and

nuclei (DAPI, blue). (B, C and D) confocal plane

and (B’, C’ and D’) projection of a confocal

image stack. In all panels, anterior is to the left.

The scale bar (in panels B) represents 20µm

and is the same for all panels.

(E) Relative abundance of the three classes of

Tsh staining patterns in trans-heterozygous

∆126/tshA3-2-66 (n=260 ovarioles) and wild-type

ovarioles (n=315 ovarioles) from three to seven

days old females grown at 25°C.

To ensure that the lethality was directly attributable to the P{lacW}A3-3-66 insertion, P-

element excisions (E{P}) of the transposon were induced by the ∆2-3 transposase. Among

2382 white-eye E{P}-jump-out lines (P{lacW}A3-3-66∆), 17 were viable in Trans with tshA3-2-66,

indicating that no second lethality exists in the tshA3-2-66 line. Out of 17 revertants, seven were

fully viable and fertile and the other ten were also viable but female sterile (two were male

sterile). Among these ten lines, we decided to further characterize the P{lacW}A3-3-66∆ (∆126)

line because 2/3 of the ∆126/tshA3-2-66 adults have white eyes, indicating that ∆126 modifies

the eye patterned white expression observed in the tshA3-2-66 adult eyes (Fig. 2A). As the

expression patterns of white may reflect the activity of regulatory elements belonging to the

tsh gene, we hypothesised that the ∆126 chromosome could bare a mutation in the tsh locus

103

Page 128: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

(Sun et al., 1995; Bhojwani et al., 1995; FL unpublished data). This observation, prompted us

to investigate whether the ∆126/tshA3-2-66 female sterile phenotype could be associated to an

altered expression of tsh in the ovaries.

To this aim, we analyzed the expression of Tsh in ∆126/tshA-3-2-66 ovaries (Fig. 2B-E).

We observed that Tsh was abnormal in 75% of the ovaries (Fig. 2E). Analysis of the variability

in Tsh staining allowed us to distinguish three types of patterns (type I, II and III) (Fig. 2E).

25,4% of ∆126/tshA-3-2-66 ovarioles presented an apparent wild-type distribution of Tsh in TF,

CC, EC and in newly born FC (type I, Fig. 2B-B’). In 50,0% of ∆126/tshA-3-2-66 ovarioles Tsh

was mostly detected in the TF and CC (type II; Fig. 2C-C’). Type III corresponds to 24,6% of

the ovarioles into which Tsh was absent from the germarium (Fig. 2D-D’).

Table 1- List of the tsh alleles crossed with P{lacW}A3-3-66∆126 and tshA3-2-66 to test for the viability and fertility

of the adult female.

Alelle Characterization Viability/Fertility in Trans with:

References

Bloomington stock

symbol P-element insertion

∆126 tshA3-2-66

Df(2L)tsh 8 VF L Fasano et al., 1991

tshNG1 VF L Salazar & Gomes, 1999

167 Df(2L)TW161 nd L

Df(2L)305 nd L induced by B. Wakimoto

10842 tshA3-2-66 1652 bp upstream ATG VS L

F= fertile; L= lethal; nd= not determined; S= sterile and V= viable

∆126 is a homozygous lethal mutation and a PCR-based analysis failed to reveal that

the P{lacW}A3-3-66 element reinserted. The allelism to tsh was tested by crossing ∆126 with tsh

alleles (Table 1) and we performed a sequence analysis of about 4 kbp upstream of the

translation start of tsh from the ∆126 chromosome; both analysis failed to validate that ∆126

is a new tsh allele and future experiments may help to answer this question. Nevertheless, the

∆126/tshA-3-2-66 females gave us the opportunity to investigate for the first time the

consequence of the perturbed expression of tsh on ovaries development.

3- Wrapping of the germ line cysts by the somatic cells is defective in

P{lacW}A3-3-66∆126/tshA-3-2-66 ovaries

To analyze the function of tsh in adult ovary, we used ∆126/tshA-3-2-66 trans-

heterozygous sterile females. ∆126/tshA-3-2-66 trans-heterozygous females didn’t lag eggs and

show atrophied ovaries.

Ovaries were stained for Vasa and Fasciclin III (FasIII), which respectively mark the

germline and follicular cells (Fig. 3; Ruohola et al., 1991). In wild-type ovaries, FasIII stained

104

Page 129: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

all the FC between the 16 cell cysts that leave the germarium and becomes restricted to the

polar cells around stage 3 (Fig. 3A).

Figure 3- ∆126/ tshA3-2-66 ovaries

display abnormal germline cyst

encapsulation.

Ovarioles from wild-type (A, B) and ∆126/tshA3-

2-66 flies (C-F) stained for FasIII (membrane of

the follicle cells; green), Vasa (germline; red)

and nuclei (DAPI, blue). (A) In wild-type

ovaries, FasIII becomes restricted to the polar

cells around stage 3 (arrows). (B) germarium

shown in panel A. (C) in ∆126/tshA3-2-66ovaries

oogenesis is arrested in S2/S3 stage. (D)

∆126/tshA3-2-66 swollen germaria showing

aberrant germline accumulation in region 1-2b.

Arrows indicate polyploid nurse cells typical of a

later stage. (E) Compound egg chambers with

30 nurse cells and two oocytes suggesting that

the follicle cells surrounded two independent 16

cell-cysts. In this confocal plane, the possible

position of each oocyte is indicated (O1 and O2).

(E’) 30 nurse cells (numbered from1-30) were

counted in a projection of a confocal image

stack of the same egg chamber shown in E. (F)

Example of a fused egg chamber. An egg

chamber has failed to bud off the germarium

and remained connected to the next one. Notice

the absence of stalk cells.

The three regions of the germarium are

indicated in panels B and D. In all panels,

anterior is to the left. The bar (in panels A, B, E,

E’ and F) represents 20µm. The panels A and C

and, B and D are at the same scale. GSC,

germline stem cell; FC, follicle cell.

Analysis of ∆126/tshA-3-2-66 ovaries revealed that the level of FasIII remains abnormally

high in mutant follicle cells (Fig. 3C). In the vitelarium, the follicle cells that packing the egg

chambers resemble the wild-type precursor for this population in the S1 stage, based in the

FasIII contents. Moreover, in ∆126/tshA-3-2-66 ovaries no polar cells were identified, the apical

localisation of the main body nucleus was perturbed and the stalk cells fail to maintain the

alignment in an organized monolayer of 5-7 cells.

105

Page 130: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

The germline stem cells (GSC) were identified as the 2-3 biggest Vasa-expressing cells

at the tip of the germarium (Fig. 3B). In tsh mutants, the number and morphology of GSC was

apparently normal (Fig. 3D), but ovaries shown swollen germaria with an increase in the size

of both (anterior/posterior and specially the dorsa/ventral) axis. Germline cysts abnormally

accumulate inside the germarium, especially prior to the 2a/2b border. In addition, within

region 3, cysts maintained the “lens shape” characteristic of region 2b and some cystoblasts

display a size comparable to the GSC, suggesting polyploidy levels characteristic of later stages

(Fig. 3D_arrows). Finally, in S2/S3 egg chambers, nurse cells display low levels of polyploidy.

All these observations suggested an oogenesis arrested before S3 stage.

Beside the arrested before S3 stage, the ∆126/tshA-3-2-66 ovaries displayed more than 5

consecutive egg chambers per ovarioles. In 24,4% of the ∆126/tshA-3-2-66 ovarioles, we

observed abnormal egg chambers (Table 2), 50% of which contain more than 16 germline cells

that could result from the abnormal wrapping of more than one 16-cell cyst (compound egg

chambers, Fig. 3E and E’). These compound egg chambers contain two or more groups of 16

germ line cells. In a compound egg chamber corresponding to a 32-cell cyst (Fig. 3E’), analysis

of confocal stack images and DAPI staining suggested the presence of two oocyte nuclei (Fig.

3E). This result supports that the nurse cell to oocyte ratio (15:1) is normal and that these egg

chambers result from the packaging of two consecutive cysts by the same epithelium, rather

than from an extra round of germline nuclear division.

The other 50% defective egg chambers correspond to fused egg chambers resulting

from normal encapsulation of 16-cell cyst, which failed to bud off the germarium (Fig. 3F). As

a consequence, they remain connected to each other and are not separated by stalk cells.

We noticed that encapsulation defects were more severe when trans-heterozygous flies

were grown for three to six days at 29°C, whereas under the same conditions wild-type flies

were fertile and have normal ovaries (data not shown).

Our data suggest that in ∆126/tshA-3-2-66 ovaries the wrapping of the cysts by the follicle

cells is perturbed and/or the budding-off from the germarium is delayed.

4- P{lacW}A3-3-66∆126/tshA-3-2-66 follicle cells are delayed in cell cycle

progression

To evaluate whether a diminution of Tsh activity has an effect on the proliferative

response of FSC and progeny, ∆126/tshA-3-2-66 ovaries were stained for phospho-histone H3

(PPH3), a mitotic cell marker. In the ∆126/tshA-3-2-66 ovaries, we observed ovarioles containing

more than 5 egg chambers but we never observed egg chambers beyond the S3 stage of

maturation. Thus, we performed the quantification and comparison of PPH3 positive FC in the

first three consecutive egg chambers (S1 to S3), since only these egg chambers display

comparable maturation stage in wild type and ∆126/tshA-3-2-66 ovaries. This analysis revealed

106

Page 131: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

that the number of the PPH3 positive cells was higher in ∆126/tshA-3-2-66 ovaries compared to

control ovaries (Fig. 4A-B and E).

Figure 4- Cell cycle progression is

affected in ∆126/tshA3-2-66 ovarioles.

(A, B) Mitotic follicle cells stained for PPH3 (green)

and nuclei (DAPI, blue); in wild-type (A) and in

∆126/tshA3-2-66 (B) ovarioles (projection of confocal

image stack). (C, D) Ovaries from wild-type (C) and

∆126/tshA3-2-66 (D) stained for activated-Caspase-3

(green) and nuclei (DAPI, blue). (E) Distribution of

the mitotic follicle cells labelled by Phospho-histone

H3 (PPH3) in the first three consecutive egg

chambers starting from stage S1 in the germarium.

PPH3 positive follicle cells were counted in wild-type

(n=69) and ∆126/tshA3-2-66 (n=98) ovarioles. (F, G,

H, I) Wild-type (F, G) and ∆126/tshA3-2-66 (H, I)

ovarioles stained for BrdU (green) and nuclei (DAPI,

blue) (projection of confocal image stack). Ovarioles

cultured for 90 minutes (F, H) or 270 minutes (G, I);

BrdU (10µM) was added for 15 minutes for pulse

labelling. In all panels, anterior is to the left. The bar

in panels A, C and F represents 50µm; the scale is

the same for all panels.

In the ∆126/tshA-3-2-66 ovaries, the number of PPH3 positive FC was always higher in the

egg chambers analysed (S1-S3) and this difference was maximize in the S1 stage, where we

observed three more mitotic FC compare to control (3,2 versus 0,8 FC/egg chamber ;

Supplementary Table 1). This result suggested that the perturbed expression of tsh in the

∆126/tshA-3-2-66 ovaries afected FC proliferation. The increase in mitotic FC number could result

(1) from a FC overproliferation or (2) from a mitotic delay of the FC. In the ∆126/tshA-3-2-66 egg

chambers, the FC nucleus seems significantly less compact and the S2/S3 egg chambers size

seems comparable to wild-type. Thus, no evidences of an increase of total FC cells, was

107

Page 132: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

observed in ∆126/tshA-3-2-66 ovaries to justify the FC overproliferation, suggesting that an

increase in mitotic FC result from a mitotic delay.

The global analysis of the distribution of the mitotic FC suggested that, in ∆126/tshA-3-2-

66 ovaries, the number of mitotic FC increase until the 4rd consecutive egg chambers showing

an abrupt reduction in the 5th egg chamber (Supplementary Table 1). This reduction could be

related with an excess of apoptosis observed in ∆126/tshA-3-2-66 ovaries beyond the 4th egg

chamber, as revealed by analysis of activated Caspase-3 activity (Fig. 4C, D). In addition, high

level of activated caspase-3 was also visible in germline.

To test for a delay in cell cycle progression in ∆126/tshA-3-2-66 ovaries, we performed 5-

bromo-2-deoxyuridine (Brdu) incorporation experiments (Fig. 4F-I). Dissected wild-type

(control) and mutant ovarioles were pulse labelled with BrdU for 15 minutes and then cultured

for 90 minutes or 270 minutes. In ∆126/tshA-3-2-66 ovaries cultured for 90 minutes, the BrdU

staining appears brighter and there was more BrdU positive FC compared to the control (Fig. 4

F, H). This result supports an extension in the length of the cell cycle in the ∆126/tshA-3-2-66

ovaries.

When wild-type ovarioles were cultured for 270 minutes, the majority of FC that have

incorporated the BrdU undergoes further division, which results in the dilution of the BrdU. In

the ∆126/tshA-3-2-66 ovaries, we observed more BrdU positive FC, especially in the first egg

chambers, showing that some FC in ∆126/tshA-3-2-66 are cycling very slowly or are cell cycle

arrested (Fig 4G, I).

All together, these results suggest that in follicular cells, tsh is required for the cell

cycle progression.

5- Down-regulation of teashirt during Drosophila oogenesis leads to the loss of

engrailed and hedgehog expression

In ∆126/tshA-3-2-66 ovarioles, proliferation of the FC was reduced and, as a consequence,

the wrapping of the cysts by the FC was perturbed and/or the budding-off from the germarium

was delayed. In the germarium, Tsh was detected with Hh and En in the TF cells and CC and,

Hh signalling has been shown to be critical to modulate proliferation of FC (Forbes et al.,

1996a; Zhang and Kalderon, 2000).

To investigate a possible regulatory relationship between tsh, hh and en in the adult

ovary, we examined the distribution of En and Hh in tsh mutants. In all ∆126/tshA-3-2-66

ovarioles, En was strongly reduced or totally absent in CC, nevertheless CC were correctly

enriched in Arm; En was normally detected in TF cells (Fig. 5B). In all ∆126/tshA-3-2-66 ovaries,

Hh staining was lost from the tip of the germarium (Fig. 5D).

These data suggest that expression of Tsh has to be tightly controlled to guarantee the

correct expression of en and hh respectively in CC and TF cells during Drosophila oogenesis.

108

Page 133: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

Figure 5- Tsh is required for the proper

expression of En and Hh in the germarium.

(A, B) Wild-type (A) and ∆126/tshA3-2-66 ovaries (B),

stained for Arm (green), En (red) and nuclei (DAPI, blue).

(C, D) Wild-type (C) and ∆126/tshA3-2-66 ovaries (D),

stained for Hh (green) and nuclei (DAPI, blue). Only the

TF, region 1 and region 2a of a single germarium are

shown. In all panels, anterior is to the left. The bar in

panel A represents 20µm; all panels are at the same scale.

- Ectopic tsh expression in the ovary induces ectopic En and phenocopies

ectopi

the role played by tsh in egg chamber formation, we ectopically

expres

excess of FSC derivatives

betwee

vitellog

and Hh lead to similar phenotypes, we tested whether ectopic

expres

6

c hh expression

To further analyze

sed tsh in wild-type adult ovaries. To this aim, hs-Gal4>UAS-tsh13 flies that carry one

copy of the UAS-tsh13 and hs-Gal4 constructs were heat-shocked.

Ectopic expression of tsh induces the accumulation of an

n egg chambers forming giant “stalk” like structures (Fig. 6A-A’). A similar phenotype

was observed when hh was ectopically expressed under the same conditions (Fig. 6B-B’;

Forbes et al., 1996a; Forbes et al., 1996b; Zhang & Kalderon 2000; Zhang & Kalderon, 2001).

In control egg chamber, FasIII is expressed in all FC during the germarial and early

enic stages but becomes restricted to the two polar follicles cells at each end of the egg

chamber by the stage 4 (Fig. 6C-C’). In contrast, in hs-Gal4>UAS-tsh13 and hs-Gal4>UAS-hh-

M4, FasIII was detected in FC at later stages and in all the excessive FSC progeny

accumulated between egg chambers (Fig. 6A-B’). These results suggest that ectopic tsh, like

ectopic hh, promotes cell proliferation and affects FasIII expression in FC, delaying follicle cells

fate acquisition. Of note, tsh was less efficient than hh in inducing giant “stalk” like structures

(Fig. 6A-B’); the giant “stalks” were smaller and less frequent in UAS-tsh13 (37%) than in

UAS-hh-M4 (62%) ovaries.

Because ectopic Tsh

sion of tsh may impact on the Hh signalling pathway. We analyzed the expression of En

in hs-Gal4>UAS-tsh13 heat-shocked ovaries and found that ectopic expression of tsh induces

ectopic expression of En (Fig. 6E-F’; compare to Fig. 6D-D’).

109

Page 134: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

Figure 6- Ectopic expression of hh and tsh

induces similar effects.

Ovarioles

hs-Gal4/+ (C, C’), were heat-

-Rescue of the P{lacW}A3-3-66∆126/tshA-3-2-66 phenotype

order to demonstrate that the perturbed expression of tsh contribute to the defects

observ ectopically provided Tsh

could rA-3-2-

from hs-Gal4>UAS-tsh13 (A, A’), hs-Gal4>UAS-

hh-M4 (B, B’) and control

shocked (kept at 25°C between heat-shocks) and stained

for FasIII (green), Vasa (red) and nuclei (DAPI, blue). (A’,

B’, C’) Enlargements of the stalk between the 1st and 2nd

egg chambers of the vitellarium signal with a arrow in the

respective left panels (B, C, D). Ectopic expression of tsh

(A, A’) and hh (B, B’) result in hyperproliferation of somatic

cells and formation of giant interfollicular stalk-like

structures (arrows). (C, C’) In control, consecutive egg

chambers are separated by 5-7 stalk cells (arrows). (D, D’)

heat-shocked hs-Gal4/+ and (E-F’) heat-shocked hs-

Gal4>UAS-tsh13 ovarioles were kept at 18°C between heat

shock and stained for Tsh (green), En (red) and nuclei

(DAPI, blue); (D’, E’ and F’) only show En (red) and nuclei

(DAPI, blue) staining. (F and F’) enlargements of the S3

egg chambers framed in E and E’, respectively. The

majority of the follicle cells into which Tsh was ectopically

produced were En positive (arrows). Anterior is to the right

and posterior is to the left. The bar (in panels A, A’, D, F)

represents 20µm. The panels (A, B, C), (A’, B’, C’), (D-E’)

and (F, F’) are at the same scale.

7

In

ed in the ∆126/tshA-3-2-66 ovaries, we decided to test whether

escue the ∆126/tshA-3-2-66 phenotype. To this aim, we ectopically expressed tsh in hs-

Gal4>UAS-tsh13; ∆126/tsh 66 mutant ovaries.

110

Page 135: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

Figure 7- Tsh rescues the imperfect proliferation phenotype of ∆126/tshA3-2-66 mutants and allows

the progression of oogenesis until stage S5/S6.

hs-Gal4/+ heat-shocked ovaries used as a control (A, D, G), ∆126/tshA3-2-66 (B, E, H) and ∆126/tshA3-2-66;UAS-

tsh13/hs-Gal4 (C,F, I). (A, B, C) ovaries (from flies maintained at 25°C) stained for FasIII (green) and Vasa (red).

Insets on bottom left show S5/S6 (A’ and C’) or S2/S3 (B’) egg chambers. Insets on bottom right show the stalk

between consecutive egg chambers (A’’, B’’ and C’’). (D-F) ovaries (from flies maintained at 18°C) stained for En

(red). (G-I) ovaries stained for Hh (green). Arrow in panel F and I indicate the restoration of En in CC and Hh

expression in TF cells, respectively.

In all panels, nuclei are stained with DAPI (blue), anterior is to the right and posterior is to the left, the scale bars in

(A-A’’) represent 50 µm and is the same for the panels (A-C), (A’-C’) and (A’’-C’’) respectively. The scale bar in (D and

G) represent 20µm and is the same for panels (D-F) and (G-I), respectively. CC, cap cells; TF, terminal filament.

This rescue assay is not ideal because (1) hsGAL4 produce a generalised tsh ectopic

expression (even out of the normal wild-type tsh context, what could have a toxic effect) and

(2) not guarantee the simultaneous and continue ectopic tsh expression in the TF and CC in all

ovarioles of the same ovary, (bab-GAL4, for example restrict expression to TF and CC).

Independently of the Gal4 driver used, the Tiptop (Tio) and the Tsh endogenous could

contribute, at least partially, to repress the ectopic tsh expression.

Previously, it was reported that tio and tsh repress each other during embryogenesis

(Laugier et al., 2005). In ∆126/tshA-3-2-66 ovaries Tio expression is increase to all TF cells, while

in wild-type it’ is restrict to the apical TF cells only (data not show). Thus it is predicable that

tsh and tio maintain the mutual repression in the tip of the germarium. It’s tempting to

suspect that ectopic tsh expression in TF cells will be affected by the presence of Tio in all

∆126/tshA-3-2-66 TF cells.

111

Page 136: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

Table 2- Quantification of the ∆126/ tshA3-2-66 and the ∆126/ tshA3-2-66;UAS-tsh13/hs-Gal4 ovaries phenotype.

∆126/tsh A3-2-66 ∆126/tsh A3-2-66; UAS-tsh13/hsGal4

Total ovarioles (n=) 275 188 Ovarioles with swollen germarium (%) 100 75,0 Ovarioles with compound and fused egg chambers (%) 24,4 4,2 Ovarioles with S3/S4 egg chambers (%) 0 32,4 Ovarioles with S4/S5 egg chambers (%) 0 9,6 Ovarioles with S5/S6 egg chambers (%) 0 4,8 Ovarioles with En in CC in the tip of the germarium (%) 0 22,2 (1)

(1) n=18 ovarioles

Beside the limitations of our rescue assay, the ectopic expression of tsh in ∆126/tshA-3-2-

66 ovaries partially rescued the tsh proliferation phenotype (Fig. 7C; compare with Fig. 7B). In

∆126/tshA-3-2-66 ovaries oogenesis was arrested before S3 stage (in 100% of the ovarioles) with

poorly polyploid nurse cells and a disorganised FC epithelium. In contrast, in the rescued

ovaries, oogenesis progressed beyond the S2/S3 stage in 46,8% of the ovarioles, the

polyploidy of the nurse cells was improved and the oocyte was located close to the posterior

polar cell (Table 2 and Fig. 7C; compare with Fig.7B and 3C,D). In addition, follicle cell

epithelium nuclei localisation (apical) and density were similar to the wild-type. Stalk cells

were correctly intercalated and formed normal stalks with 5 to 7 cells. Furthermore, in 4,8% of

the ovarioles, the maturation of the egg chambers reached the S5/S6 stage and, the frequency

of ovarioles with compound or fused egg chambers was reduce to only 4,2% (Table 2 and Fig.

7C’-C’’). In 24,5% of the hs-Gal4>UAS-tsh13; ∆126/tshA-3-2-66 ovarioles, the size of the

germaria was improved, but remains slightly bigger than in wild-type. These results show that

expression of tsh can rescue the ∆126/tshA-3-2-66 mutant phenotype.

We then asked whether the rescued ∆126/tshA-3-2-66 mutant phenotype could depend on

the capacity of the ubiquitously provided tsh to restore hh and/or en expression. In heat-

shocked hs-Gal4>UAS-tsh13; ∆126/tshA-3-2-66 ovaries, we found that tsh restored en and hh

expression (Fig. 7 F and I; Table2). This result is consistent with the previous finding that tsh

is required for the expression of hh and en respectively in the TF cells and CC. These data

suggest that the absence of hh expression in ∆126/tshA-3-2-66 ovaries could be due to the down

regulation of tsh. However Hh that was loss in both, TF and CC, in ∆126/tshA-3-2-66 ovaries, it

was restored only in CC of the hs-Gal4>UAS-tsh13; ∆126/tshA-3-2-66 ovaries. In ∆126/tshA-3-2-66

ovaries Tio expression is extended to all TF cells, while in wild-type it’ is restrict to the apical

TF cells only, indicated a mutual repression between the two paralogue, as explained above

(data not show, for details see chapter II part II of this manuscript). Thus, it is predicable that

in hs-Gal4>UAS-tsh13; ∆126/tshA-3-2-66 ovaries, the presence of Tio in all TF cells could

112

Page 137: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

diminish the tsh ectopic expression in all TF, justifying the presence of ectopic hh expression

only in CC, where no Tio was observed.

Finally, to test whether ectopic Hh could rescue the tsh mutant phenotype, ∆126/tshA-3-

2-66;hs-Gal4/UAS-hh-M4 flies were heat-shocked (Ingham and Fietz, 1995). Under these

conditions, the germline/somatic cells distribution resembles the wild-type and the germarium

size was comparable to hs-Gal4>UAS-tsh13; ∆126/tshA-3-2-66 (Supplementary Fig. 1).

Furthermore, the distribution of the follicle epithelium nuclei and stalk cells intercalation were

also improved. Nevertheless, oogenesis was unable to proceed beyond the S3/S4 stage and

egg chambers contained nurse cells with low levels of polyploidy and absence of differentiated

polar cells. Thus, the ∆126/tshA-3-2-66 mutant phenotype can also be rescued by over

expressing hh, suggesting that Tsh is acting upstream of hh.

Discussion

Whereas the function of tsh has been extensively studied during embryogenesis and

imaginal discs development, the function of tsh in the ovaries was never investigated. Here we

showed that Tsh is co-expressed with Hh at the tip of Drosophila germarium, in terminal

filament (TF) cells and in cap cells (CC). We demonstrated that overexpression of tsh mimics

the phenotype associated with hh overexpression. Phenotypic characterisation of ovaries into

which tsh expression is perturbed allowed us to suggest that hh and engrailed (en) expression

might depend on tsh. In these mutant ovaries, ectopically provided Tsh partially rescues the

defects and restores hh and en expression in CC. In addition, ectopic expression of Tsh induce

ectopic En expression. Our results suggest that in adult ovaries expression of Tsh might play a

critical role in regulating hh expression contributing for the regulation of FSC proliferation and

the specification of somatic cell identity.

Control of FSC proliferation upstream of the Hh pathway

At the tip of the Drosophila adult ovary, the germarium contains three identified stem

cell populations [germline stem cells (GSC), follicle stem cells (FSC) and escort stem cells

(ESCs)], making it an excellent model system for studying stem cell biology. Our data suggest

that Tsh might play a role in FSC proliferation. We showed that Tsh, like Hh, is probably

required for the proper packing of 16-cell germ-line cyst; the absence of Tsh correlates with de

disruption of the coordination between the 16-cell germ-line cyst formation and FSC

proliferation. Our data support that the frequency of the encapsulation defects increases with

the lowering of the Tsh level in the germarium.

113

Page 138: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

The Hh pathway has been implicated in the FSC proliferation (Forbes et al., 1996a and

1996b; Zhang and Kalderon, 2000 and 2001). We show that in wild-type ovaries the ectopic

expression of tsh mimics the constitutive Hh phenotype, that is the accumulation of an excess

of FSC derivates between egg chambers as a result of FSC over-proliferation. This result

supports that Tsh positively affects FSC proliferation and, either by a direct or indirect manner,

negatively affects FC differentiation, as previously reported for Hh. In our experimental

conditions, we noticed that the hs-Gal4>UAS-hh-M4 ovaries showed a more penetrating

phenotype, when compared to hs-Gal4>UAS-tsh13 ovaries; the giant “stalks” structures are

bigger and more frequent.

Ubiquitous expression of tsh in ∆126/tshA-3-2-66 ovaries partially rescues the proliferation

defect, significantly improved the encapsulation process and the progression of oogenesis.

More important, in the rescued ovarioles, hh and en expression was restored in the CC,

suggesting that the low levels of Tsh detected in ∆126/tshA-3-2-66 ovaries could account for the

proliferation defect.

The presence of polyploid nurse cells characteristic of a later stage in S1 egg chambers

indicates a delayed budding off from the germarium. Accordingly, PPH3 staining and BrdU

incorporation revealed that ∆126/tshA-3-2-66 ovaries exhibit a delay in cell cycle progression.

The quantification of the PPH3 positive FC per egg chamber in wild-type and ∆126/tshA-3-2-66

ovaries supports the idea of a delay to complete the sequence of approximately five mitotic

cycles downstream from the stem cell until leave the germarium and probably an incapacity

for the FC to undergo the next three to four mitotic cycles from 2-6 stage. In the Drosophila

eye, a recent study indicated that blocking the Hh pathway reduces the number of retinal

progenitors that take up BrdU as a consequence of a slower cell cycle kinetics (Locker et al.,

2006). Thus it is attempting to propose that the delay in cell cycle progression observed in

∆126/tshA-3-2-66 ovaries could be a consequence of the down-regulation of hh, which is known

to promote follicle-cell proliferation (Forbes et al., 1996a; Zhang and Kalderon, 2000). In

vertebrates, mutations in the SHH pathway have been shown to affect cell cycle progression

(Adolphe et al., 2006; Barnes et al., 2001). In the future, it would be interesting to analyze

whether vertebrate orthologues of tsh affect cell cycle progression.

Regulation of En and Hh expression in the Tip of the germarium

In adult Drosophila ovary, Tsh was detected in proximal TF, CC, ESs (including ESC)

and FC in region 3 of the germarium and the expression levels of Tsh in TF cells and CC vary

with aging. In the TF cells and CC, Tsh was co-detected with Hh, and Tsh and En proteins were

detected in the same group of somatic cells at the tip of the germarium. In the absence of Tsh,

hh expression was drastically reduced in TF and CC whereas En was absent from CC but was

still detectable in TF cells. In the ovaries, the regulatory relationships between hh and en

114

Page 139: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

remain poorly understood. Our analysis propose that en expression is maintained in the TF

cells in absence of hh, suggesting that in the TF cells en expression might not depend on hh.

It has been shown that the ectopic expression of hh and en in the adult ovary cannot

induce the expression of the other outside its normal domain of expression (Forbes et al.,

1996). Here we show that ectopic expression of tsh induces expression of en in FC. These

results suggest that tsh can act upstream of en and hh, and that tsh could participate to the

regulation of en and hh expression in Drosophila ovaries. Interestingly, we noticed that, as

flies age, while En continues to be strongly expressed in all non-somatic cells in the tip of the

germarium, Tsh was down regulated in the most distal TF cells. This observation suggests that

Tsh not only co-localises with, but also displays the same dynamic expression in TF and CC

reported for Hh (Forbes et al., 1996a; Forbes et al., 1996b). Our experiments suggest that the

down regulation of hh may be tsh-dependent and, because of the absence of a suitable tsh

temperature-sensitive allele and the impossibility to make tsh clones, we were unable to

further analyze these regulatory mechanisms.

The mechanisms by which Tsh may regulate hh and/or en expression are still poorly

understood. In vertebrate and Drosophila, Tsh factors can recrute the corepressor CtBP and in

vertebrate TSHZ3 has been shown to recrute HDAC1/2 activities (Manfroid et al., 2004;

Kajiwara et al., 2009). Moreover, in Drosophila, Tsh has been shown to behave as a repressor

(Taghli-Lamallem et al., 2007). Thus, if in the TF cells and CC tsh regulates the expression of

hh and/or en, it is likely that this regulation might not be direct but rather indirect through the

control of the activity of a repressor.

In the future, our findings may help to further identify and characterize factors that

participate to the exquisite control of Hh expression during embryonic development and adult

tissue homeostasis.

115

Page 140: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART II

Supplementary Data

Supplementary Figure 1- Rescue of the ∆126/tshA3-2-66 proliferation phenotype by ectopic Hh

(A) hs-Gal4/+ (B) ∆126/ tshA3-2-66; UAS-hh-M4/ hs-Gal4 ovarioles stained for FasIII (green), Vasa (red) and

nuclei (DAPI; blue).

In (B) ∆126/ tshA3-2-66; UAS-hh-M4/ hs-Gal4 ovarioles the germline/somatic cells distribution and the germarium size

are approximated to the observed in wild-type. Furthermore, some improvements are also observed in the distribution

of the follicle epithelium nuclei and stalk cells intercalation. Nevertheless, oogenesis was unable to proceed beyond the

S3/S4 stage and egg chambers contained nurse cells with low levels of polyploidy and absence of differentiated polar

cells.

In all panels, anterior is to the right and posterior is to the left. The scale bar (in panel A) represents 50 µm and is the

same for both panels.

Supplementary Table 1- Quantification of the positive P-Histone H3 FC in the first 5 egg chambers of wild-

type (n=69) and ∆126/ tshA-3-2-66(n=98) ovaries.

Egg Chamber position

FC/ovariole in(1):

WT ∆126/tshA-3-

2-66

1 0,8 3,2 2 1 2,2 3 2,6 3,2 4 5,5 2,6 5 3,7 0,8

(1)FC/ovarioles= Total FC counted in the 1st (2nd, 3rd, 4th or 5th) egg chamber/ total number of ovarioles

screened

116

Page 141: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_Part III:

Page 142: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

118

Page 143: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Teashirt and Tiptop repress each other in Terminal Filament Cells

during Drosophila oogenesis

Silvia Pimentel1,2, Isabel Salazar1, Christine Vola2, Rui Gomes 1 and Laurent Fasano2†

1 Universidade de Lisboa, Faculdade de Ciências, Departamento de Biologia Vegetal, Campo

Grande, 1749-016 Lisboa, Portugal 2 Institut de Biologie du Développement de Marseille Luminy (IBDML), UMR 6216, CNRS-

Université de la Méditerranée, Campus de Luminy, Case 907, 13288 Marseille Cedex 09,

France.

†Authors for correspondence: (emails: <[email protected]>)

119

Page 144: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal
Page 145: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

Abstract

In Drosophila, the teashirt (tsh) gene encodes a zinc finger protein known to be

required for proper specification of the trunk segments in the developing embryo. The Tsh

protein is expressed throughout development and within a number of different tissues.

Recently, it was described a new member of the tsh family in Drosophila, call Tiptop (Tio),

which encodes a protein of 1024 amino acids with a fourth Zinc finger motif in the C-terminal

part. Priviously was described the mutual repression between Tsh and Tio during

embryogenesis, as the ectopic expression of Tio masked a crucial role for Tsh in trunk

patterning. Here we report that, during oogenesis, Tsh and Tio present a complementary

expression pattern in terminal filament cells (TF). In ∆126/tshA3-2-66 ovaries, which do not

express Tsh properly, Tio is expressed ectopically in all TF, and the reverse is observed for Tsh

in tio473 homozygous ovaries. The construct hsGal4>UAS-tioFL rescues the ∆126/tshA3-2-66

phenotype as efficiently as the hsGal4>UAS-tsh13. The ectopic expression of tio induces

similar effects to those described after overexpression of tsh, but preferentially in background

defective for tsh. We found that, like it was reported during Drosophila embryogenesis, Tsh

and Tio repress each other in Drosophila germarium. During oogenesis, Tio has, at least, a

partial redundant function, since the lack of Tio may be rescued by the tsh expression in

tio473mutants.

Key words: Drosophila; oogenesis; terminal filament cells; teashirt; tiptop.

121

Page 146: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

Materials and Methods

Drosophila strains

All genetic markers and balancers are described by Lindsley and Zimm (1992) and

FlyBase (http://flybase.bio.indiana.edu) except as noted.

The hypomorphic allele tshA3-2-66 (Bloomington) has a P{lacW} insertion at 1652bp

upstream of the tsh translation start (see chapter II part I of this manuscript).

The ∆126 line results from a tshA3-2-66 P-element excision mutagenesis (for details see

chapter II part I and part II of this manuscript). Among 2382 revertants, 17 were viable in

Trans with tshA3-2-66. From this 17 viable revertants, seven were fully viable and fertile and the

other ten were also viable but female sterile. The P{lacW}A3-3-66∆ (∆126) was one of the ten

lines that presented females viables but sterile. In addition, 2/3 of the ∆126/tshA3-2-66 adults

have white eyes, indicating that ∆126 modifies the eye patterned white expression observed in

the tshA3-2-66 adult eyes (for more details see chapter II part I and part II of this manuscript).

Our analysis failed to validate that ∆126 is a new tsh allele, nevertheless, in ∆126/tshA-3-2-66

ovaries tsh expression is perturbed, which constitute a opportunity to investigate the effect of

tsh in the expression of his paralogue tio during oogenesis.

Females (or males) tshA3-2-66/T(2;3)CyOTM6B,Tb were crossed with male (or females)

∆126/T(2;3)CyOTM6B,Tb to get ∆126/tshA3-2-66 females used in this study.

The allele tio473 was identified as a tio deletion that removes tio and surrounding DNA,

3,6Kb upstream of the start codon and 0,9Kb downstream of the stop codon. Neither tio mRNA

nor protein are detected (Laugier et al., 2005).

For rescue and overexpression experiments the UAS-tsh13 construct (Gallet et al.,

1998), the UAS-tio construct (Laugier et al., 2005) or the UAS-hh-M4 construct (Ingham and

Fietz, 1995) were crossed to the driver P{w+mC=GAL4-Hsp70.PB}TR2 (Bloomington).

Staining ovaries

Antibody staining, was performed as described by (Zhang and Kalderon, 2000), with

the modifications previously reported in chapter II partII of this manuscript.

Primary and secondary antibodies and their dilutions were the following: anti-Tsh

(rabbit Tsh, a gift from S Cohen, EMBL Heidelberg, 1:1000 dilution); anti-Tio 1:500 (rat Tio,

Laugier et al., 2005, 1:500 dilution) mouse 4-D9 anti-Engrailed invected (Developmental

Studies Hybridome bank, 1:10 dilution); mouse 7G10 anti-Fasciclin III (Developmental Studies

Hybridome bank, 1:200 dilution) and rabbit anti-Vasa (gift from Lehaman, 1:1000 dilution).

Secondary were conjugated with Texas red or FITC (Molecular Probes, 1:500 dilution).

For En and Tio staining, the signal was amplified with the Tyramide Signal Amplification

Kit (PerkinElmer) using biotinylated secondary antibodies (Jackson Laboratories, 1:500). In

122

Page 147: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

these cases, the ovaries were submitted to a peroxidase treatment (3% H2O2 for 10 minutes)

to reduce the background, before their blocking.

Stained ovaries were mounted in Vectashield medium with DAPI (Vectors) and analyzed

under a LSM 510 Zeiss confocal microscope.

Rescue of ∆126/ tshA-3-2-664 mutant phenotype

∆126;UAS-tsh13/T(2;3)CyOTM6B,Tb or ∆126;UAS-tioFL/T(2;3)CyOTM6B,Tb males

were crossed with tshA-3-2-66;hsGal4/T(2;3)CyOTM6B,Tb females. In the day of the enclosion,

∆126/tshA-3-2-66;UAS-tsh13/hsGal4 or ∆126/tshA-3-2-66;UAS-tioFL/hsGal4 females were

submitted to heat-shock; tubes were immersed in a water bath at 37°C, for 1hour, twice a

day, during 3 consecutive days. In the 4th day, ovaries were dissected and stained with FasIII

and Vasa antibodies. Females were grown at 25°C until the adult stage and between the heat-

shocks.

Tsh, Tio and Hh overexpression

UAS-tsh13/T(2;3)CyOTM6B,Tb, UAS-tioFL/T(2;3)CyOTM6B,Tb or UAS-hh-

M4/T(2;3)CyOTM6B,Tb males were crossed with hsGal4/T(2;3)CyOTM6B,Tb females. In the

day of the enclosion, UAS-tsh13/hsGal4, UAS-tioFL/hsGal4 or UAS-hh-M4/hsGal4 females were

submitted to a heat-shock at 37°C, for 1hour, which was repeated twice a day, during 3

consecutive days. In the 4th day, ovaries were dissected and stained with FasIII and Vasa.

Females were grown at 25°C, until adult stage and between heat-shock.

Results

1-Tsh and Tio show complementary localisation in Terminal Filament cells

Previous results have shown that, during early embryogenesis, tsh and tio expression is

detected in distinct domains (Laugier et al., 2005). Tsh is located in the trunk and Tio in parts

of the head and tail. In later embryogenesis, they present both common and tissue-specific

expression patterns. The loss of tio caused ectopic expression of tsh in the clypeolabrum,

where tio is normally expressed (Laugier et al., 2005). Similarly, loss of tsh caused ectopic

expression of tio in the trunk. In different mutant situation it was showed that in the trunk and

head, they repress each other’s expression (Laugier et al., 2005). In contrast, during adult

development, it was recently shown that tsh and tio were co-expressed in imaginal discs

(Besssa et al., 2009).

123

Page 148: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

In order to find out whether Tsh and Tio behave during oogenesis, in a similar way to

embryos or to imaginal discs development, we looked for Tsh and Tio localisation in wild-type

and mutant Drosophila ovaries.

In wild-type, Tsh is present in terminal filaments (TF) cells, cap cells (CC) and in escort

cells (EC) including escort stem cells (ESC) and recently formed follicle cells (FC; see chapter

II part II of this manuscript and Fig. 1A). Tio is only observed in TF (Fig. 1A). Double staining

in wild-type adult ovaries, with anti-Tsh and anti-Tio, show a complementary localisation of

both proteins in the nuclei of TF. Tio is localised in more apical TF, where Tsh staining is weak,

while Tsh is stronger in more proximal TF cells, where Tio is not detected (Fig. 1A). Although

the complementar expression in TF, tsh has a more extense domain of expression suggesting a

more important role in oogenesis. Interestingly, a precise deletion of tio is homozygous viable

and fertile (Laugier et al., 2005). tio473 homozygotes show perfectly normal ovaries, without

detectable anomalies (data not show). While ∆126/tshA3-2-66 female are sterile, with atrophied

ovaries, in wich oogenesis is arrested before S3 stage (Fig. 2B, for more details see also

chapter II part II of this manuscript).

Figure 1- Tsh and Tio co-repress each other.

(A) wild-type; (B) ∆126/tshA3-2-66 and (C) tio473 homozygous germarium stained with Tsh (green) and Tio (red)

antibodies. tsh is expressed in cap cells (CC), escort cells (EC) and also, in new follicle cells in S1 stage. (A) In

terminal filament, Tsh is observed in more proximal cells (TFp), while tio is expressed only in more apical cells (TFa).

(B) Tsh is lost from all germarium ∆126/tshA3-2-66 (phenotype III) while, Tio is detected in all terminal filament cells

(TF). (C) In tio473 homozygous germarium, Tsh is observed in all terminal filament cells (TF) and in cap cells (CC),

escort cells (EC) and also, in new follicle cells in S1 stage.

Anterior is to the right and posterior is to the left. The scale bar (in panel A) represents 20µm and is the same for all

panels.

In order to avalaible the behavoir of Tsh in the absence of Tio and vice-versa, we

analysed the Tsh expression in the germarium of tio473 homozygotes, and the Tio expression in

the germarium of ∆126/tshA3-2-66. We observed ectopic Tsh staining in the apical TF cells of the

124

Page 149: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

tio473 germaria; Tsh was detected in all TF, CC and EC, and a weak Tsh staining continues to be

observed in recently produced follicles cells (Fig.1C). In ∆126/tshA3-2-66 ovarioles, Tio was

detected in all TF but never in CC or EC, nor even in ovarioles where Tsh is missing in all the

germarium (phenotype type III; Fig.1B; for more detail see chapter II part II of this

manuscript). For note, we suspected that ∆126/tshA3-2-66 ovaries are an allele of regulation, and

a residual quantity of Tsh (not detectable by immunostainig) could be enough to repress tio in

CC of ∆126/tshA3-2-66ovaries.

2- The gene tio show similar functional properties to tsh in the tip of the

ovaries and can partially substitute it during oogenesis

As tio and tsh have been shown to be functionally similar during embryogenesis and

imaginal development (Laugier et al., 2005; Bessa et al, 2009), we dicide to investigated if

this was also the case during oogenesis. We preformed three experiments to test this point.

2.1-UAS-tioFL expression can partially rescue ∆126/tshA3-2-66phenotype

We tested the ability of tio to rescue the ∆126/tshA3-2-66 phenotype using the UAS/Gal4

system (Brand and Perrimon, 1993). We used the hsGal4 driver, where Gal4 is under the

control of the heat-shock promoter hsp70, because no other Gal4 driver mimicking tsh

expression in ovaries was available. Flies carrying a single copy of UAS-tsh13 or UAS-tioFL and

hsGal4 will be referred subsequently as ∆126/tshA3-2-66;UAS-tsh13/hsGal4 or ∆126/tshA3-2-

66;UAS-tioFL/hsGal4, respectively.

In ∆126/tshA3-2-66 ovaries, oogenesis was arrested before S3 stage (in 100% of the

ovarioles) with poorly polyploid nurse cells and a disorganised FC epithelium. These ovaries

present enlarged germaria, with frequent abnormal egg chamber (24,4% of the ovaioles

presented at least one compound or fused egg chambers; Fig.2B, for more detail see chapter

II part II of this manuscript ). If Tio/Tio were functionally equivalent, we expect that Tio can

rescue the tsh loss of function, when expressed in the tsh domain.

The ectopic expression of tio in ∆126/tshA3-2-66 ovaries partially rescues the proliferation

phenotype (Fig.2C-D). In these flies, oogenesis progress until S5/S6 stage, nurse cells improve

ther ploidy levels and it is possible to distinguish the oocyte close to the posterior polar cell

(Fig.2 D arrowhead and arrow, respectivelly). Follicle epithelium nuclei are perfectly placed in

the apical part of the epithelium, and their density resembles the wild-type. Stalk cells are

correctly intercalated forming normal stalks. The size of the germaria is smaller than in

∆126/tshA3-2-66.

Unexpectedly, in some aspects Tio seems more efficient than Tsh in the rescue of the

∆126/tshA3-2-66 phenotype. The S5/S6 egg chambers were more frequent whenever we

125

Page 150: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

ectopically expressed tio. In addition, and unlike tsh ectopic expression, in ∆126/tshA3-2-

66;UAS-tioFL/hsGal4 ovaries, we also observed an improvement in the germ-line/ somatic cells

organization in the germarium.

Together, these results suggest that, ectopic tio is able to restore the absence of tsh in

∆126/tshA3-2-66ovaries, and surprisingly, in some aspects, tio was even more efficient than tsh

to rescue the proliferation phenotype observed in ∆126/tshA3-2-66ovaries.

Figure 2- Tsh and

Tio rescue the imperfect

proliferation phenotype

of the ∆126/tshA3-2-66

mutant and allow the

progression of oogenesis

until stage S5/S6.

(A) hsGal4/+; (B)

∆126/tshA3-2-66; (C)

∆126/tshA3-2-66; UAS-

tsh13/hsGal4 and (D)

∆126/tshA3-2-66;UAS-

tioFL/hsGal4 ovarioles were

stained with antibodies to

FasIII (green) in the

membrane of the follicle

cells and Vasa (red) in

germ-line. DNA (blue) was

labelled with DAPI.

(C, D) Tsh and Tio ectopic

expression rescues the

imperfect proliferation

phenotype displayed by

∆126/tshA3-2-66 ovaries; it is

possible to observe S5/S6

egg chambers (insert in

bottom right in C and

biggest egg chamber in D)

with the oocyte (arrowhead)

located close to the

posterior polar cell (arrow), whereas nurse cells improved their polyploidy. The density and the arrangement of the

nuclei of the follicle epithelium, in both cases (C, D) are comparable to the wild-type (A). Stalk cells are correctly

intercalated. The size of the ∆126/tshA3-2-66;UAS-tsh13/hsGal4 (C) and ∆126/tshA3-2-66;UAS-tioFL/hsGal4 (D) germaria

are smaller than in ∆126/tshA3-2-66(B).

Anterior is to the right and posterior is to the left. The scale bar (in panel A) represents 50µm and is the same for all

panels and insert.

126

Page 151: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

2.2- Tio maintained engrailed (en) expression in all TF cells of ∆126/tshA3-2-66

germarium, while in tio473 homozygous ovaries en expression is maintened by Tsh.

We found that Tsh is required for proper engrailed (en) and hedgehog (hh) expression

in the tip of the germarium (see chapter II part II of this manuscript and Fig.3). In the wild-

type, En is co-localised with Hh in the nuclei of the same group of somatic cells. Hh is

expressed selectively in the membrane of specialized non–proliferating somatic TF and CC, at

the anterior tips of the germarium (Forbes et al., 1996a; Forbes et al., 1996b Fig.3A). To

evaluate the possibility of tio to be able to replace tsh in the regulation of En expression in TF,

and vice versa, we stained ovaries from ∆126/tshA3-2-66 and tio473 with antibodies against En

and Armadillo (Arm; Fig. 3).

En is detected in all TF and CC in homozygous tio473 germaria (Fig.3C). In this

background, Tsh replaces Tio in apical TF and, in consequence, En is detected like it was

observed in wild-type ovaries. Since Tsh can completely replace the absence of tio, at least in

the maintenance of En expression, homozygous tio473 ovaries are perfectly normal and female

fertile.

Figure 3- Localisation of En in wild-type, ∆126/tshA3-2-66 and tio473 germaria.

(A) wild-type; (B) ∆126/tshA3-2-66and (C) tio473 homozygous germaria stained with Arm (green) and En (red)

antibodies. DNA (blue) was labelled with DAPI. (A, C) In wild-type and tio473 germaria, En is detected in terminal

filament (TF) cells and cap cells (CC). (B) In ∆126/tshA3-2-66 germarium En is lost from cap cells (CC) that continue to

display normal levels of Arm.

Anterior is to the right and posterior is to the left. The scale bar (in panel A) represents 20µm and is the same for all

panels.

Thus, ectopic tsh expression perfectly mimics tio expression in TF in tio473 ovaries and

apparently could replace the absence of Tio, since tio473 homozygous female are perfectely

fertile.

127

Page 152: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

In ∆126/tshA3-2-66 ovaries, Hh is completely absent from the tip of the germarium. En is

strongly reduced or totally absent in CC (although these last cells are correctly enriched in

Arm). Curiously all TF cells still expressed en (Fig.3B and for more detail see chapter II part II

of this manuscript). The absence of Hh from the TF and CC at the tip of ∆126/tshA3-2-66 ovaries

suggests that Tio alone is insufficient to substitute Tsh in the maintenance of Hh expression

(data show, see chapter II part II of this manuscript).

The maintenaice of En in TF but not in CC suggest that Tio can replace tsh in all TF, and

maintain En expression in this domain of the ∆126/tshA3-2-66 germarium. However, since tio is

not ectopically expressed outside TF, En expression is not maintained in CC. Thus, Tio is

expressed in all TF cells (being ectopic only in proximal TF cells) of ∆126/tshA3-2-66ovaries and it

is insufficient to matained En in this domain in the absence of tsh.

These show that Tsh can replace Tio but not the inverse and suggests that Tio may

have a redundant function with Tsh during oogenesis, resembling their relation during

embryogenesis. However, at least in ∆126/tshA3-2-66;UAS-tioFL/hsGal4 ovaries the ectopic

expression of tio induce ectopic en expression out of their wild-type domain.

2.3- Ectopic expression of tio mimic ectopic expression of tsh specially in a

mutant background context

In embryos the ectopic expression of Tsh and Tio in embryo and in imaginal discs

induced similar effects (Laugier et al., 2005; Bessa et al., 2009). It was showed that Tio is less

efficient than Tsh to induce naked cuticle identity but, from stage 13 onwards, ectopic tsh and

tio activated and maintained wg transcription in a similar way (Laugier et al., 2005).

The ectopic expression of tsh generated giants stalk-like structures, (see chaper II part

II of this manuscript and Fig.4C) and mimic the ectopically expression of hh (Forbes et al.,

1996a; Forbes et al., 1996b; Zhang & Kalderon 2000; Zhang & Kalderon, 2001).

In wild-type, two consecutive egg chambers are separated by 5-7 stalk cells (Decotto

and Spradling, 2005; Fig.4A). In hsGal4>UAS-tsh13 ovaries, we observed the overproliferation

of the follicle stem cells (FSC) and/ or progeny, which accumulated between egg chambers to

give giants stak-like structures (Fig.4D). In hsGal4>UAS-tioFL ovaries the giants stak-like

structures was rarely observed (Fig.4E). However, in ∆126/tshA3-2-66;UAS-tioFL/hsGal4 these

abnormal stalks were frequent and particularly enriched in FasIII, resemble the observed for

hsGal4>UAS-tsh13 and for hsGal4>UAS-hh-M4 giants stak-like structures(Fig.4F).

These data suggest that ectopic tio can induce the same Hh overactivation phenotype,

preferentially, in the absence of tsh.

Unlike to hsGal4>UAS-tsh13 and hsGAL4>UAS-hh-M4, the ectopic expression of tio

produced bigger germaria specially increased in region 1-2b. These bigger germaria seem to

have extra germ-line cysts, at least two rows of disc shaped cysts surrounded by follicle cells

128

Page 153: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

in region 2b. However, these germaria conserved the germline organization observed in wild-

type. Unlike to ∆126/tshA3-2-66 germaria, the hsGal4>UAS-tioFL germaria presented typical disc

shaped cysts in region 2b and were correctly intercalated by the somatic follicle cells.

Moreover, in region 3, the germ-line cysts were perfectly circular and well surrounded by the

follicle cells (Fig.4E-inset). In these ovaries (hsGal4>UAS-tioFL), we failed to detect

encapsulation problems, compound or fused egg chambers. In a wild-type background, the

ectopic expression presented a more evident effect in GSC rather than in FSC proliferation.

More work is need to confirme this possible implication of Tio in GSC proliferation, for exemple,

specific GSC staining and posterior counting, should be performed.

Figure 4- Ectopic expression of

hh, tsh and tio induces a similar

phenotype.

(A) hsGal4/+; (B) ∆126/tshA3-2-66; (C)

hsGal4>UAS-tsh13; (D) hsGal4>UAS-hh-

M4; (E) hsGal4>UAS-tioFL and (F)

∆126/tshA3-2-66;UAS-tioFL/hsGal4 stained

with antibodies to FasIII (green) and Vasa

(red) and with DAPI for DNA (blue).

In wild-type (A), consecutive egg

chambers are separated by 5-7 stalk cells.

Hyperactivation of tsh (C) or hh (D) results

in hyperproliferation of somatic cells and

the formation of giant interfollicular stalk-

like structures (arrowheads in panels C, D

and F)). The same phenotype, caused by

tio overexpression, is more frequent in a

tsh mutant background (F); giants stak-

like structures are rare in hsGal4>UA-tioFL

(E).

An enlargement of each germarium is

shown in the bottom right inset of each

panel. In hsGal4>UAS-tioFL ovarioles (F)

the bigger germaria seem to result from an

increase in germline-cyst production. It is

possible to observe at least two rows of

disc shaped cysts (arrows in the bottom

right inset in panel E). In contrast to

∆126/tshA3-2-66germaria (B), the

hsGal4>UAS-tioFL germaria (F) maintain

the germline/ soma organization

characteristic of wild-type (A).

Anterior is to the right and posterior is to

the left. The bar (in panels A) represents

50µm. All the panels are at the same scale.

129

Page 154: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

Discussion

Teashirt and Tiptop are localised in the left arm of chromosome 2 and are transcribed in

the sam

Tio and Tsh proteins is about 28%. The best

percen

other duplicated genes in the Drosophila genome are important during its

develo

uplication event

characteristic of

utual repression between teashirt and tiptop

tio473 ovaries shows that Tsh

and Tio TF) cel

s

orm

e orientation 5’-3’. These genes are spaced 183 Kb from each other and code for a

protein with zinc finger (Znf) C2H2 type motifs.

The global percentage of identity between

tage of identity coincides with the three Znf motifs and is around 51%. Tio has a fourth

Znf motif conserved in the C-terminal part, which is also present in all other Tsh family

members, except in Tsh (Caubit et al., 2000; Laugier et al., 2005; Herke et al., 2005; Onai et

al., 2007).

Many

pment. For example, engrailed (en) and invected (inv; Poole et al., 1985), knirps (kni)

and knirps-related (knrl; Rothe et al., 1989), sloppy paired 1 (slp1) and sloppy paired 2 (slp2;

Grossniklaus et al., 1992), zerknüllt 1 (zen1) and zerknüllt 2 (zen2; Rushlow et al., 1987),

disconnected (disco) and disco-related (disco-r; Mahaffey et al., 2001) or distal antenna (dan)

and distal antenna related (danr; Emerald et al., 2003). These genes are characterised by

close genetic linkage, by extensive sequence homology and, similarly to the duplication of tsh

tio, they are also duplicated in tandem (Laugier et al., 2005; Laugier, 2005).

The tsh tio gene pair seems to have arisen from a recent d

Drosophilidae. Phylogenetic analysis of the sequence of Tsh/Tio family protein

in the arthropods showed that Drosophila Tio protein is closer to the other arthropod proteins

than to Drosophila Tsh (Laugier et al., 2005; Laugier, 2005). It seems that tio is closer to the

ancestral gene than to tsh, and as is often the case for duplicated genes in Drosophila, one

plays a dominant role as compared to the other (Gonzalez-Gaitan et al., 1994; Gustavson et

al., 1996); in this case tsh played the dominant role (Laugier et al., 2005; Laugier, 2005). The

duplication arose only after the separation of the Drosophila and Glossina lineages, between 60

and 140 million years ago. The fact that the embryonic pattern of expression of the single Tc

tiotsh gene in the flour beetle roughly resembles the summation of the patterns of tsh and tio

in Drosophila embryo points to a possible subfunctionalization of tsh and tio after the

duplication (Lynch and Force, 2000; Shippy et al., 2008; Bessa et al., 2009).

M

The expression pattern analysis in ∆126/tsh A3-2-664 and

repress each other in the terminal filament ( ls. In tio473 ovaries, Tsh is detected

in all TF cells. When tsh activity is removed (∆126/tshA3-2-66), Tio i detected in all TF cells,

including the more proximal TF cells, where Tsh is n ally detected, showing that Tsh

represses the expression of tio and the inverse. This is in accordance with the reports during

130

Page 155: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

embryogenesis and eye development, where the loss of one gene leads to the transcriptional

de-repression of the paralog resulting in the compensation by one paralog for the loss of the

other (Laugier et al., 2005; Bessa et al., 2009; Datta et al., 2009).

Curiously, Tio was never detected in cap cells (CC) of ∆126/tshA3-2-66 ovaries, even in

the cas

fertile, ∆126/tshA3-2-66 flies are sterile

and ar

at, as fly age, Tsh was down regulated in the most distal TF cells. Could

be inte

ess each other but also act synergistically

in som

to what have been reported for embryos (Laugier et al., 2005; Laugier, 2005),

our ob

e of ovarioles, where no Tsh is detectable by imunostainig in the germarium (phenotype

III, see chaper II part II of this manuscript). For note, in contrast to the homozygous tio473,

the trans-heterozygote ∆126/tshA3-2-66 is not a null mutant. Thus, this may reflect that even a

tiny amount of tsh expression is enough to repress tio.

Although homozygous tio473 flies are viable and

rest oogenesis before S3 stage. This reinforces Tsh as having the major role of the two

duplicated genes during oogenesis. And suggests that, tio plays at least in part, a commum

function with tsh during Drosophila oogenesis, similar to what it happens during embrogenesis

and eye formation.

We noticed th

resting available if this downregulation is related with Tio, since this is the late Tio

domain. Our Tio/Tsh immunostainig was performed in flies with around 8 days old. In future

would be interesting performed these analyses in younger flies. In addition, the Tio/ Tsh

behaviour during late pupal stage should also be interesting to study, since Tsh is expressed in

all TF at this stage. Tio start to be expressed after Tsh start to be donwregulated, or initial the

two paralogue share the same expression domain?

In embryos, we may say that tsh and tio repr

e way, and that this may fine tune the development (Laugier et al., 2005; Laugier,

2005). It was showed that Tio can partially replace the loss of tsh function, while homozygous

tio473 embryos are viable, the tsh8 embryos (double mutants for tio and tsh) present a more

severe phenotype than tsh mutants alone (for example tsh2757R8; Laugier et al., 2005; Laugier,

2005). In order to find out if the absence of both proteins aggravates the ∆126/tshA3-2-664

ovary phenotype, we tried to deplete simultaneously tsh and tio during oogenesis using UAS-

RNAi-tsh in tio473 ovaries (a gift from Casares). However, the absence of a specific Gal4 driver,

that mimic tsh expression in their wild-type domain during all oogenesis unable us to efficienly

depleted Tsh.

Similarly

servations did not allowed us to distinguish if the regulatory mechanism between Tsh

and Tio repression is direct or indirect. In a simple model, Tsh may repress tio and vice versa.

The molecular similarities between the two paralogs support a possibility of a negative auto

and cross-regulatory loop (Bessa et al., 2009).

131

Page 156: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

Tio show similar functional properties to tsh in the tip of the ovaries and can

partially substitute it during oogenesis.

We showed that the ectopic expression of tio is able to rescue the ∆126/tshA3-2-66

phenotype as efficiently, or even better, than the tsh ectopic expression. This suggests that, in

the ectopic situation, when tio is expressed out of its normal expression domain, he is able to

compensate the loss of tsh and restore the normal morphology of the ∆126/tshA3-2-66 ovarioles

allowing the progression of the oogenesis until S5/S6 stage.

The lower toxicity of the ubiquitus expression of the UAS-tio construct, comparatively to

the ubiquitus expression of the UAS-tsh construct could justify the differences in the efficacy of

the 2 constructs. Accordingly to what has been described for embryogenesis, Tsh may have

more number of target genes than Tio during oogenesis, and in consequence of that, the

ectopic expression of tio affect the expression of a more restrict number of target gene and

therefore is less toxic in ovaries than the ectopic expression of tsh. Other simple, explanation

could be just a mater of efficiency of the construct, independently of the gene associated.

The overexpression of tio is able to mimic the overactivity of Tsh mediating Hh in FSC

proliferation during oogenesis, preferencially in a mutant context for tsh. The production of

giants stalk-like structures, delayed in their fate acquisition, was more frequently observed in

∆126/tshA3-2-66;UAS-tioFL/hsGAL4 than in hsGAL4>UAS-tioFL.

Interestingly, the presence of Tsh seems to prevent ectopic tio to induce giants stalks-

like structures. In a mutant context for tsh, the ectopic tio was able to mimic overproliferation

of FSC and delaying FC fate acquisition, as described for overexpression of Hh signalling. This

suggests that, in hsGAL4>UAS-tioFL ovaries, the endogenous Tsh is able to repress the ectopic

expression of Tio or it is capable to inhibit the activation of Hh by ectopic Tio. Tsh and/or Tio

levels are critical for normal development. Therefore, it would be predicted the tightly

regulation of the total concentration of Tio and Tsh presented in a cell. Transcriptional cross

regulation between tio and tsh ensure that their protein levels are kept constant as has been

show in the embryo (Laugier et al., 2005) and imaginal discs (Bessa et al., 2009). Thus in

hsGal4>UAS-tioFL ovaries is probable that the endogenous Tsh prevent a generalised ectopic

expression of Tio, and in consequence a few giants stalk-like structures are observed. In

contrast, in a mutant context background, the ubiquitous expression of Tio will be effective,

and more giants stalk-like structures enriched in FasIII will be produced.

On the other hand, these results indicated that ectopic Tio is able to positively affect

the expression of Hh, at least in the absence of tsh, suggesting that Tio can induce the same

target known for tsh in oogenesis, which is Hh and En. In addition, it was observed ectopic en

(out of their normal domain: TF and CC) in ∆126/tshA3-2-66;UAS-tioFL/hsGal4 (supplementary

Fig.1J).

In the TF cells of the ∆126/tshA3-2-66 ovaries, Tio is ectopic in the Tsh domain, localizing

in all TF cells but never in CC, curiously En display the same domain of expression in this

132

Page 157: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

context of Tsh absence (∆126/tshA3-2-66;Fig 3 and Supplementary Fig. 2D-F). Thus, it is

tempting propose that Tio can contributed to the maintenance of en expression in the

∆126/tshA3-2-66 ovaries. However the absence of Hh from the all TF cells and CC in this ovaries

(see chapter II part II of this manuscript) indicating clearly that in a tsh reduction context, Tio

was unable to maintain hh in TF. It is not clear whether Tio contribute to the maintenance of

en expression in ∆126/tshA-3-2-66 mutant but this result suggest that Tsh might regulate hh

expression in different ways or in a regulatory mutant context (like we suppose that is the case

of the ∆126/tshA3-2-66 ovaries) the reminiscent Tsh prevent the Hh maintenance by Tio, for

example, preventing tio expression in CC. We question, if in a tsh null mutant context, Tio

could not keep En and Hh in all TF and CC. Further experimental work must be performed to

clarify how Tsh and Tio regulates en and hh expression during oogenesis.

In addition, we observed that, unlike the overexpression of tsh or hh, the ectopic

expression of tio produced bigger germarium suggesting extra GSC and germ-line cysts, that

conserve the germline organization observed in wild-type. Unlike to the ∆126/tshA3-2-66

germaria, the hsGal4>UAS-tioFL germaria presented a normal germline/ somatic cell

organization. It is know, that beside the importance of the Hh function for self-renewal and

proliferation of the FSC, this signalling plays a non-essential role in maintaining the GSC, by

having redundant function with the piwi–mediated pathway (King et al., 2001). However the

overexpression of hh stimulates a slight increase in the number of GSC (King et al., 2001).

Thus, it is tempting to suppose that the apparent increase of the germline cyst, in the ovaries

where tio was ectopically expressed, could be the result of the effect of Tio in Hh signalling

favoring GSC instead FSC proliferation. For note, tio is normally expressed only in TF, also

component of the GSC niche. The ectopic tio expression could have affected also the number

of GSC or the rate of germ-line cyst production, originating the accumulation of the cyst in

region 1-2b of the germarium. We speculated that Tsh could mediates the FSC proliferation

while Tio could have a more preponderant role in mediates the GSC proliferation, in both

cases, through Hh signalling.

In a tsh mutant background, may be the overexpression of tio produces germaria with

normal size because the increase in GSC number and or proliferation rate was accomplished by

an increase in follicle production, documented by the more frequent presence of giants stalk-

like structures in hsGal4>UAS-tioFL /tshA3-2-66;UAS-tioFL/hsGal4 ovaries, comparatively to

hsGal4>UAS-tioFL ovaries. This suggests that, in some level, endogenous Tsh represses the

ectopic expression of tio or the ectopic effect of Tio in tsh targets during oogenesis.

In conclusion, we confirmed that tsh and tio repressed each other during oogenesis,

and we verified that tio is able to perform a redundant function with tsh in ovaries; Tio and Tsh

share some targets (perhaps indirect targets) like en and hh. We speculate that the ability to

regulate these targets may be assigned to a few sequence similarities between Tio and Tsh,

particularly to the three zinc finger region. The maintenance of En by tsh and tio has been

133

Page 158: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

already described in Drosophila embryos and imaginal development, as well as a commum

function of the two paralogs (Laugier et al., 2005; Laugier, 2005; Bessa et al., 2009). Here we

show that these functions are maintained during Drosophila oogenesis, opening the way to

clarify the relationships of this signalling pathway and its regulation within the GSC and FSC.

134

Page 159: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART III

Supplementary figures

Supplementary Figure 1- Tsh and En in wild-type, ∆126/tshA3-2-66, tio473 and ∆126/tshA3-2-66;UAS-

tio/hsGAL4 germaria.

(A, B, C) wild-type; (D, E, F) ∆126/tshA3-2-66, (G, H, I) tio473 homozygous germaria and (J) ∆126/tshA3-2-66;UAS-

tio/hsGAL4 stained with Tsh (green) and En (red) antibodies. DNA (blue) was labelled with DAPI. (A, B, C, G, H, I). In

wild-type and tio473 germaria, En is detected in all terminal filament and cap cells. (D, E, F) In ∆126/tshA3-2-

66germarium En is lost in cap cells but maintained in all terminal filament cells. (J) In ∆12 6/tshA3-2-66;UAS-tio/hsGAL4

germarium, en is ectopic out of their wild-type pattern of expression.

Anterior is to the right and posterior is to the left. The scale bar (in panel A) represents 20µm and is the same for all

panels.

135

Page 160: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal
Page 161: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_Part IV:

Page 162: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

138

Page 163: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

The knock-down of the teashirt gene causes a metaphase-like

arrest in Drosophila larval neuroblasts and S2 cells

Isabel Salazar1, Sílvia Pimentel1,2, Jorg D. Becker3, Laurent Fasano2, Stephen Kerridge2

and Rui Gomes1 *

1 Universidade de Lisboa, Faculdade de Ciências, Departamento de Biologia Vegetal,

Campo Grande, 1749-016 Lisboa, Portugal 2 Institut de Biologie du Développement de Marseille Luminy (IBDML), UMR 6216,

CNRS-Université de la Méditerranée, Campus de Luminy, Case 907, 13288 Marseille Cedex 09,

France. 3 Affymetrix Core Facility, Instituto Gulbenkian de Ciência, Rua Quinta Grande Nº6,

2780-156-Oeiras, Portugal.

Authors for correspondence: (emails: <[email protected]>)

139

Page 164: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal
Page 165: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RESULTS_PART IV

Abstract

In Drosophila, the teashirt (tsh) gene encodes a zinc finger protein known to be

required for proper specification of the trunk segments of the developing embryo. Tsh is

expressed throughout development and within a number of different tissues. Here we report

the characterisation of two new hypomorphic mutations of teashirt that cause a metaphase-

like arrest in larval neuroblasts, with the majority of mitotic figures exhibiting highly

condensed chromosomes. Metaphase-arrested figures are associated with apparently normal

mitotic spindles and regular centrosomes. Analysis of squashed preparations of tsh mutant

larval neuroblasts with specific antibodies to the checkpoint proteins (Bub1 and Rod) and to

the CENP-A homologue CID revealed normal localisation of these proteins and no premature

sister chromatid separation. Furthermore, in metaphase-arrested neuroblasts cyclin-A appears

to be degraded whereas cyclin-B remains detectable. Double mutants tsh rod and tsh bub1

display phenotypes that resemble the phenotype of the tsh single mutant, as their neuroblasts

present an elevated mitotic index and a strong metaphase-like arrest, with highly condensed

chromosomes associated with detectable levels of cyclin-B. Therefore, rod and bub mutations

cannot override the strong mitotic arrest produced by the mutant tsh alleles and progression

to anaphase is inhibited. Similarly, the depletion of tsh in Drosophila S2 cells by double-

stranded RNA interference (RNAi) disrupts mitotic progression and mimics the tsh mutant

phenotype in neuroblasts. Throughout the course of RNAi experiments, most cells arrest in a

metaphase-like configuration leading to a remarkable increase in the metaphase and mitotic

indices. Microarray data identified ihog as a significantly downregulated gene in tsh mutants.

Our results strongly suggest that Tsh is required for mitosis progression possibly acting trough

the Hedgehog-pathway.

Key words: Drosophila, teashirt, mitosis, metaphase-anaphase transition, anaphase-

promoting complex/cyclosome (APC/C), neuroblasts, mitotic mutant, hedgehog, iHog.

Page 166: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Introduction

The teashirt (tsh) gene encodes a protein with three atypical widely-spaced C2-H2 zinc

finger motifs and it is required for the patterning of the trunk segments in Drosophila

embryogenesis in collaboration with the Hox genes (Fasano et al., 1991; Roder et al., 1992).

Loss-of-function mutations in tsh lead to trunk-to-head transformation, whereas ectopic

expression of tsh results in head-to-trunk transformation (Fasano et al., 1991; Roder et al.,

1992; de Zulueta et al., 1994). More recently, it was shown that Tsh directly interact with the

Hox gene Sex combs reduced (Scr; Taghli-Lamallem et al., 2007).

Mutants in tsh also resemble segment-polarity genes, revealing that tsh acts as a

temporal and spatial modulator of the Wingless (Wg) pathway in the ventral ectoderm, and

that it is required for development of the proximal parts of the adult appendages (Erkner et

al., 1999; Wu and Cohen, 2000) and for specification of hinge structures of the wing (Azpiazu

and Morata, 2000; Bessa et al., 2002; Casares and Mann, 2000; Pan and Rubin, 1998; Soanes

et al., 2001; Pan and Rubin, 1998).

Interestingly, vertebrate Tsh proteins were isolated and their expression patterns

suggest that function has been conserved from Drosophila to the vertebrates (Caubit et al.,

2000; Manfroid et al., 2004).

Progression into and through mitosis is controlled by activation and inactivation of M-

phase promoting factor (MPF), a complex comprising the Cyclin-dependent kinase 1 (Cdk1,

Cdc2 in Drosophila) and a Cyclin regulatory subunit. In animal cells, the mitotic Cyclins A (Cyc

A) and B (Cyc B), which bind as regulatory subunits to Cdk1, are required for entry into

mitosis.

MPF activity is also modulated by post-translational mechanisms, notably

phosphorylation on inhibitory sites of CdK1, (Thr14 and Tyr15) by the kinases Myt1 and Wee1,

respectively (Porter and Donoghue, 2003). Activation of Cdk1 occurs through the combination

of three required steps: phosphorylation of Thr161, in late G2 by the Cdk-activating kinase

(CAK); nuclear localization of Cdk1/cyclin B1 is promoted by Plk1 phosphorylation of Ser147

and dephosphorylation of the Cdk1 in the Thr14 and Tyr15 by members of the Cdc25

phosphatase family (only one member in Drosophila: String; Porter and Donoghue, 2003;

Fesquet et al., 1993). Activation of Cdc25C is achieved by phosphorylation of Ser198 by Plk1

(Roshak et al., 2000).

Polo-like kinases (PlK) are well established modulators of cell cycle progressionin a wide

range of organisms participating in many aspects of cell division such as mitotic entry, bipolar

spindle formation, chromosome segregation, mitotic exit and cytokinesis (Glover et al., 1998;

Nigg, 1998).

142

Page 167: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

A series of phosphorylations catalysed directly or indirectly by MPF provokes profound

structural reorganisation of the cell and drives the formation of the metaphase spindle, while

inactivation of MPF triggers the completion of mitosis and the return to the interphase state.

During mitosis is essential to ensure the equal distribution of a complete set of

chromosomes by each daughter cell. The spindle assembly checkpoint (SAC) or metaphase

checkpoint is a quality control mechanism that prevents chromosome segregation errors. It

acts to restrain cells from entering anaphase, until all replicated chromatids have formed

proper attachments to a functional bipolar spindle. The checkpoint pathway transduces an

inhibitory signal generated by improperly attached kinetochores that delays anaphase onset.

This signal is presumably amplified and ultimately acts to halt the action of the APC (reviewed

by Hoyt, 2001; Shah and Cleveland, 2000; Amon, 1999).

The SAC acts via a signal transduction pathway composed of the Mad2 and Bub

proteins to inactivate Cdc20 (Alexandru et al., 1999; Fang et al., 1998; Hwang et al., 1998;

Kim et al., 1998). This prevents the degradation of Securin and consequent chromosome

segregation until all the chromosomes be attached to both spindle poles.

The Securins must be degraded to initiate sister chromatid separation and to allow the

onset of anaphase. All these proteins are degraded by ubiquitin-mediated delivery to the

proteosome 26S, which requires an ubiquitin ligase called the anaphase-promoting

complex/cyclosome (APC/C) (for review see Peters, 1999; Peters, 2006; Zachariae and

Nasmyth, 1999).

The APC/C is under complex control via phosphorylation Rudner and Murray, 2000) and

by binding one of two WD 40 repeat proteins: Cdc20 (Fizzy/Slp1/p55Cdc) and Cdh1 (hct1/Fizzy-

related/Srw1/ste9), which function as rate-limiting and cell cycle stage-specific activators of

the APC/C (for review see Morgan, 1999). In Drosophila, as in animal cells, Cdc20Fizzy initiates

APC/C-dependent degradation at metaphase-to-anaphase transition (Dawson et al., 1995;

Leismann et al., 2000; Sigrist et al., 1995).

The proteolytic degradation of Cyc A and B is essential for the exit from mitosis (Jacobs

et al., 2001; Jacobs et al., 1998; Lehner and O'Farrell, 1989; Parry and O'Farrell, 2001; Sigrist

et al., 1995). Once Cyclins are destroyed, activation of the APC/C by Cdc20 declines and is

replaced by Cdh1Fizzy-related until cells enter S phase (Sigrist and Lehner, 1997).

Here we describe two new tsh alleles (tshNG1and tshA3-2-66) and their molecular and

phenotypic characterization. Our analysis revealed that the Tsh protein is essential for mitotic

progression in larval neuroblasts and S2 cells. In tshNG1 and tshA3-2-66 mutants, the absence of

tsh function causes a metaphase-like arrest in neuroblasts, and the depletion of tsh in S2 cells

by RNAi mimics this mutant phenotype. Here, we discuss potential roles of this transcription

factor in mitosis regulation.

143

Page 168: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Materials and Methods

Drosophila strains and identification of tshNG1 and tshA3-2-66 mutants

All genetic markers and balancers are described by Lindsley and Zimm (1992) and

FlyBase (http://flybase.bio.indiana.edu) except as noted. The tshNG1 mutant allele was isolated

after MNNG (N-methyl-N’-nitro-N-nitrosoguanidine) mutagenesis using standard conditions

(Roberts, 1998), and lines carrying an induced lethal on the second chromosome were then re-

balanced over T(2;3)CyO-TM6B,Tb. Since TM6B carries the dominant larval/pupal marker

Tubby (Tb), the use of this translocation allows larvae homozygous for any gene of interest on

the balanced second or third chromosomes to be recognised by virtue of being Tb+. Available

deficiencies for chromosome 2 in Bloomington and Umea Drosophila Stock Centres delimited

the tshNG1 mutation to the cytogenetic interval 39D3-E1; 40A1-F on the basis of lack of

complementation with deficiencies Df(2L)305 (breakpoints 39A-E7; 40A1-F) and Df(2L)TW161

(breakpoints 38A6-B1; 40A4-B1) and full complementation with Df(2L)TW84 (37F5-38A1;

39D3-E1). tshNG was subsequently identified as a tsh mutant allele that fails to complement

tsh8 (a deficiency that removes tsh and the distal breakpoint was localised at 2Kb upstream of

the start codon of tiptop; Fasano et al., 1991; Laugier et al., 2005). The lethal P{lacW}

insertion allele tshA3-2-66 (P{lacW}A3-3-66) was identified after the screening of the Bloomington

P-element insertions at the 38A6-40B1 interval that fail to complement the tshNG1 allele, and

subsequently shown to be unable to complement deficiencies Df(2L)TW161, Df(2L)305 and

tsh8. For rescue experiments, the UAS-tsh13 construct (Gallet et al., 1998) was crossed to the

driver P{w+mC=GAL4-Hsp70.PB}TR2.

To test for embryonic lethality, tshNG1/In(2LR)CyL and tshA3-2-66/In(2LR)CyL stocks were

outcrossed to wild type and Cy+L+ male progeny were backcrossed to the balanced stocks.

Eggs collected over a 24-hour period were aged for 48 hours and examined. No embryonic

lethality of tshNG1 or tshA3-2-66 was observed. On the other hand, embryonic lethality was

detected when these alleles were combined with Df(2L)305 or tsh8. Crosses between tshNG1/+

males or tshA3-2-66/+ males and Df(2L)305/+ females or tsh8/+ females produced 24%

(tshNG1/Df(2L)305), 23% (tshNG1/tsh8), 23% (tshA3-2-66/Df(2L)305) or 22% (tshA3-2-66/tsh8) of

dead eggs (n=200 in each case).

tsh rod and tsh bub1 double mutants were constructed by standards means, using the

genetically null allele rodX5 (Karess et al., 1989, Scaerou et al., 1999, Basto et al., 2000) and

bub1 allele (l(2)k02113 (Basu et al., 1999). Both alleles were a kind gift from R. Karess

(CNRS, Gif-sur-Yvette) and the double mutant constructs were confirmed by segregation and

complementation tests.

144

Page 169: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Nucleic acid procedures and analysis

Standard protocols were used for nucleic acid preparation and in situ hybridisation

(Gonzalez and Glover, 1993; Sambrook et al., 1989). The pπ 12.20 plasmid (O'Hare and

Rubin, 1983) was used as a probe to confirm the cytological location of the P{lacW} tshA3-2-66

insertion to the interval 39E-40A. A genomic DNA fragment next to the P{lacW} insertion was

isolated by plasmid rescue: DNA from heterozygous tshA3-2-66 flies was digested with EcoRI,

self-ligated, and transformed into TOP10 E. coli competent cells (Invitrogen). The fly genomic

DNA from the chimeric plasmid was completely sequenced starting from primer P1 (5’-GAC

CAC CTT ATG TTA TTT CA-3’) and primer lacW8465 (5’-AAT AGG CGT ATC ACG AGG C-3’),

both specific for the P{lacW} transposon. Manual DNA sequencing was performed with the

Sequenase 2.0 Kit (Amersham Pharmacia) and sequences were independently confirmed by an

automated ABI377 v3.0 DNA Sequencer (Toplab, Germany). Homology searches using the

BLASTN 2.0 programs (Warren Gish, unpublished; Altschul et al., 1990) identified a fragment

within the sequence AE003781 (GenBank) which includes the teashirt gene. The structure of

the mutant allele tshNG1 was determined by primer walking and sequencing of PCR-generated

fragments starting from the primers: (5’-AGC AGG ACG GCG CAT GCG A-3’) and (5’-GGC GGT

CTT CTC CTT CTT CA-3’). These fragments were subcloned into pCR2.1-TOPO (Invitrogen) and

double–stranded sequenced by an automated ABI377 v3.0 DNA Sequencer (Toplab, Germany).

For Northern analysis, poly(A)+ RNA was extracted from wild type, revertant,

transformant and homozygous tsh third instar larvae using the QuickPrep Micro mRNA Kit

(Amersham Pharmacia). Twenty microgram samples of poly(A)+ RNA were subjected to

electrophoresis in 1.2% agarose gels containing formaldehyde. The tsh probe used for

Northern was a mixture in equal parts of the following cDNA clones (ESTs) from the Berkeley

Drosophila Genome Project (BDGP) and Research Genetics: CK00422, CK01473, LP11282,

LP07848 and LP44646. As a control for loading, the Northern was re-probed with specific ESTs

for the NUCB1 gene (Otte et al., 1999) made from an equal mixture of the following cDNA

clones from BDGP and Research Genetics: LD40692, LP03575, LP08122, GH05163 and

CK01389.

Rescue of tsh mutants

Males from transformed lines carrying the transgene on the third chromosome (w;

CyO/Sp; UAS-Tsh13/ UAS-Tsh13) (Gallet et al., 1998) were crossed to T(2;3)CyO

TM6,Tb/tshNG1. In parallel, females In(2L)Cy In(2R)Cy, al2 Cy lt3 cn2 L4 sp/S Sp Bl bwD were

crossed to w;TM3,P{w+mC=GAL4-Hsp70.PB}TR2,Ser/Sb males. Among the F1 progeny,

females CyO/tshNG1; UAS-Tsh13/+ were selected and mated to bwD/+; TM3, P{w+mC=GAL4-

Hsp70.PB}TR2, Ser /+. F2 males bwD/tshNG1; TM3, P{w+mC=GAL4-Hsp70.PB}TR2, Ser / + or

UAS-Tsh13 were crossed to CyO/tshNG1; UAS-Tsh13/+ females. Offspring were heat shocked in

145

Page 170: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

a water bath at 37ºC for 30 minutes twice every day and allowed to develop at 25ºC. Animals

bearing the same construct without heat shock treatment were prepared in parallel. An

analogous procedure was followed to examine the ability of the transgene to rescue the mitotic

defects and viability of tshA3-2-66 flies.

The production of ectopic Tsh protein rescued the mitotic defects in the central nervous

system (CNS) in third instar larvae of tshNG1/ tshNG1; TM3, P{w+mC=GAL4-Hsp70.PB}TR2, Ser /

UAS-Tsh13 and tshA3-2-66/ tshA3-2-66 ; TM3, P{w+mC=GAL4-Hsp70.PB}TR2, Ser / UAS-Tsh13, and

the development proceeded shortly after the eclosion from the pupal case. Animals bearing the

same constructs without heat shock treatment failed to rescue the abnormal phenotypes, as

well as control tshA3-2-66 and tshNG1 mutants carrying either UAS-tsh13 or the hs-GAL4 driver

when subjected to the same heat shock treatment.

Cytology and Immunofluorescence Observations

Larval brain squashes and immunostaining of whole-mount third instar larval CNS and

imaginal discs were carried out as described (Gonzalez and Glover, 1993). Microtubules were

detected with rat anti-α−tubulin antibody, YL1/2 (1:100, Sera-Lab) and Alexa 488-conjugated

anti-rat (1:200, Molecular Probes). Centrosomes were revealed with mouse anti-γ-tubulin

GTU88 (1:100, Sigma) and an FITC-conjugated horse anti-mouse secondary antibody (1:50;

FI-2001, Vector Laboratories). Cyclin A and B (Whitfield et al., 1990; a kind gift from A.

Tavares, IGC, Lisboa), Polo Ma294 (Llamazares et al., 1991; a kind gift from A. Tavares, IGC,

Lisboa), Rod (Scaerou et al., 1999, Scaerou et al., 2001), Bub1 (Basu et al., 1999), CID

(Blower & Karpen; a kind gift from R. Karess, CNRS, Gif-sur-Yvette) were detected with

specific rabbit antibodies and used at 1:100, 1:200, 1:100, 1:500, 1:1000 and 1:1000

respectively. Secondary antibodies were made in goat and conjugated to FITC or rhodamine

(Jackson ImmunoResearch Laboratories). DNA was stained with 1µg/ml of propidium iodide or

Hoechst 33258 and the brains were mounted in Vectashield (Vector Laboratories).

Immunoblotting

Standard protein and immunological techniques were followed (Harlow and Lane, 1988;

Sambrook et al., 1989). For immunoblotting, secondary antibodies were all peroxidase-coupled

and detection was performed using the Lumi-Light Plus Western Blotting Kit (Roche). Anti-rat-

POD was bought from Sigma. Total protein samples from Drosophila third instar larvae and

Schneider S2 cells were prepared by homogenisation in SDS sample buffer. We used the

follow primary antibodies: rat anti-Tsh (1:500); mouse anti-Fizzy (1:20; a kind gift from C.

Lehner) and rabbit anti-Polo MA294 (1:500, a kind gift from A. Tavares, IGC, Lisboa).

146

Page 171: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

RNAi treatment and analysis of S2 Tissue culture cells

RNAi was performed according to the methods of Clemens et al. (2000) using target

sequences that exhibited minimal homology with others genes as determined by BLAST

comparison. The template for in vitro transcription was generated using the primer pairs: 5´-

TAA TAC GAC TCA CTA TAG GGA GAC CAC AGC GGG TCA ATT CCC TGA CT-3´ and 5´-TAA TAC

GAC TCA CTA TAG GGA GAC CAC GAT TTG CAT GGC CTG TTC CA-3´. The Purified PCR product

(~650 bp in length) was used to produce double-strand RNA (dsRNA) using Megascript T7

transcription kit (Ambion) according to the manufacturer’s instructions. Schneider S2 cells

were grown in Schneider’s Drosophila medium (Gibco BRL) supplemented with 10% fetal calf

serum (FC, Gibco) and 50 µg/ ml streptomycin and penicillin at 25 ºC. To perform RNAi cells

were treated with 15 µg dsRNA as described by Somma et al., 2002. Control cultures were

prepared in the same way with GFP-dsRNA. Both cultures were grown for 144 h at 25ºC and

then processed for biochemical and cytological analysis. For immunofluorescence, cells were

processed as previously described (Adams et al., 2001). Immunostaining of microtubules and

centrosomes was performed with antibodies against α−tubulin and γ-tubulin (Sigma-Aldrich).

Secondary antibodies were FITC-α-rat and Texas red-α-mouse (Jackson ImmunoResearch

Laboratories).

Microscopy images

Larval CNS and S2 cells were examined under phase contrast or fluorescence using a

Zeiss Axioplan II microscope with a 100x/1.30 oil immersion Plan-Neofluar objective. Images

were acquired with a SPOT-2 camera (Diagnostics Instruments, Inc.) and processed with

Adobe Photoshop 6.0 (Adobe System).

RNA Isolation, Target Synthesis and Hybridization to Affymetrix GeneChips

Total RNA was extracted from an appropriate number of brains (2x150 wild type brains

and 2x300 mutant brains) with 71,85 ng/µl (70,2 ng/µl-73 ng/µl) of RNA in average per

sample using the RNeasy Mini Kit (Qiagen, Hilden, Germany). Concentration and purity was

determined by spectrophotometry and integrity was confirmed using an Agilent 2100

Bioanalyzer (Agilent Technologies, Palo Alto, CA).

RNA was processed for use on Affymetrix (Santa Clara, CA, USA) Drosophila Genome

Arrays, according to the manufacturer’s Small Sample Labeling Protocol VII described in the

technical note “GeneChip Eukaryotic Small Sample Target Labeling Assay Version II”

(http://www.affymetrix.com/support/technical/technotes/smallv2_technote.pdf). Briefly, 100

ng of total RNA was used in a reverse transcription reaction (SuperScript II; Invitrogen,

Paisley, UK) to generate first-strand cDNA. After second-strand synthesis, double-strand cDNA

147

Page 172: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

was used in an in vitro transcription (IVT) reaction to generate cRNA (MEGAscript T7 kit;

Ambion, Austin, TX). 400 ng of this cRNA was used for a second cDNA synthesis, followed by a

second IVT reaction to generate biotinylated cRNA (ENZO BioArray HighYield RNA Transcript

Labeling Kit; ENZO Diagnostics, Farmingdale, NY). Size distribution of the cRNA and

fragmented cRNA, respectively, was assessed using an Agilent 2100 Bioanalyzer.

15 µg of cRNA was used in a 300-µl hybridization containing added hybridization

controls. 200 µl of mixture was hybridized on arrays for 16 h at 45°C. Standard post

hybridization wash and double-stain protocols were used on an Affymetrix GeneChip Fluidics

Station 400. Arrays were scanned on an Affymetrix GeneChip scanner 2500.

Data Analysis of the Affymetrix GeneChips

Scanned arrays were analyzed first with Affymetrix MAS 5.0 software to obtain

Absent/Present calls and for subsequent analysis with dChip 1.3 (http://www.dchip.org, Wong

Lab, Harvard). The four arrays used in this study (biological replicates) were normalized

applying an Invariant Set Normalization Method (Li and Wong, 2001b). Then the normalized

probe cell intensities of the 4 arrays were used to obtain model-based gene expression indices

based on a PM-only model (Li and Hung Wong, 2001a). Replicate data for the same sample

were weighted gene-wise by using inverse squared standard error as weights. Only genes

called Present in at least 1 of the 4 arrays and within replicate arrays called Present within a

variation of 0 < Median (Standard Deviation/Mean) < 0.5 were kept for downstream analysis

(7099 genes). Thus, genes called absent in all arrays and genes with inconsistent expression

levels within replicate arrays were excluded. Finally, all genes compared were considered to be

differentially expressed if they were called Present in at least one of the arrays and if the 90%

lower confidence bound of the fold change between experiment and baseline was above 1.2.

The lower confidence bound criterion means that we can be 90% confident that the fold

change is a value between the lower confidence bound and a variable upper confidence bound.

Li and Wong (2001b) have shown that the lower confidence bound is a conservative estimate

of the fold change and therefore more reliable as a ranking statistic for changes in gene

expression. This criterion has been used in other gene expression studies (Becker et al., 2003;

Ramalho-Santos et al., 2002). A LCB threshold of 1.2 seems to be justified, because

comparisons with real-time PCR data showed that Affymetrix Genechips analyses are reliable

and underestimate differences in gene expression (Yuen et al., 2002).

Annotations for the ~13.500 genes represented on the Drosophila Genome Array were

obtained from the NetAffx database (www.affymetrix.com) as of June 2003.

148

Page 173: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Results

1-Molecular analysis of the tsh mutant alleles

The tshNG1 allele was selected amongst a collection of second chromosome late larval

lethals after nitrosoguanidine mutagenesis (Materials and Methods) and a non-complementing

allele (tshA3-2-66) was further identified as a P{lacW} insertion within the promoter of the tsh

gene. A 2 kb EcoRI genomic fragment adjacent to the P{lacW}A3-2-66 insertion was cloned by

plasmid rescue and used as a probe to confirm by in situ hybridisation its localisation to 40A on

wild-type polytene chromosomes (data not shown). DNA sequencing revealed that the

P{lacW}A3-2-66 element is inserted 439 bp upstream of the putative transcription start site

(GTCAGTCG), i.e., 1652bp upstream the translation start (Fig. 1). DNA sequencing of PCR-

generated fragments of tshNG1 allele revealed a deletion of 7 bp at {-18, -26bp} from the

larger exon, which deletes the putative branch site consensus and affects the splicing (Fig. 1).

Northern blot analysis in wild-type third instar larvae confirmed the expression of tsh

transcripts of two size classes: a predominant class of transcripts of 5.4 kb and a minor class

of 8.5 kb transcripts corresponding to the previously reported 5-5.4 kb and 8.1-8.5 kb tsh

transcripts expressed throughout development (Fig. 2A, lane 5;Fasano et al., 1991). In tshA3-2-

66 homozygotes the 8.5 kb transcripts are missing and the 5.4 kb are scarcely expressed,

suggesting that the reduction in the tsh transcripts is associated with the mutant phenotype

(Fig. 2A, lane 2). Transcript expression was restored in those lines showing full reversion to

the wild-type, and in which remobilization of P{lacW}A3-3-66 with the ∆2-3 transposase source

(supplementary material) resulted in its precise excision (Fig. 2A, lane 3). In larvae

homozygous for tshNG1 only an abnormal 7Kb transcript is observed (Fig. 2A, lane 1).

Presumably the absence of the normal transcripts of 5,4 Kb and 8,5 Kb and, the presence of

these abnormal tsh mRNA of 7kb, are responsible for the defects observed in tshNG1 mutants.

To confirm that the tsh transcription unit was responsible for the tsh phenotype, we undertook

to rescue the mutant phenotype by expressing the tsh cDNA using the hs-Gal4/UAS system

(Brand and Perrimon, 1993) in the mutant backgrounds of tshNG1 and tshA3-2-66 (see materials

and methods and Fig. 2A, lane 4). We found that a 30-min heat-shock at 37ºC twice a day

gave the rescue of all mitotic defects observed in both tsh mutant homozygotes and they

reached the adult stage. The lethality was shifted from larval to adult stage.

Expression of Tsh in mutant larval central nervous system (CNS) was also assessed by

Western blotting (Fig. 2B) using a previously described anti-Tsh antibody (Gallet et al., 1998).

On Western-blots this antibody detected a 116 kDa isoform of Tsh in wild-type third instar

larval CNS (Fig. 2B, lane 1). In contrast, the amount of Tsh protein is undetectable in the

tshNG1 and tshA3-2-66 mutants (Fig. 2B, lanes 2 and 3 and Supplementary fig.1). Thus, tshNG1

and tshA3-2-66 mutants appear to be protein nulls or severe hypomorphic alleles. The same blot

was re-probed with anti-Fizzy as a loading control (Fig. 2B, bottom line).

149

Page 174: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Figure 1- Molecular organisation in tshNG1 and tshA3-2-66 mutants.

(A) Schematic overview of the tsh region. Sites of P-element insertion causing tshA3-2-66 mutation and genomic deletion

associated with the lethal tshNG1 allele are explicitly indicated. The tsh transcription unit (open and black boxes) is

interrupted by one intron (kinked lines). Non-coding 5’ and 3’ regions of the transcript are shown in open boxes. The

tshA3-2-66 insertion site maps at –439 nucleotides upstream the transcription start site i.e., 1652bp upstream the

translation start, possibly within the promoter region. The tshNG1 allele deletes 7 bp at {–18, -26 bp} upstream the

second exon. (B) Excerpts from the DNA sequence AE003781 (GenBank) showing relevant segments of the tsh gene,

the tshA3-2-66 insertion site (indicated by a triangle) and the deleted nucleotides in tshNG1 (in bold). The start site of tsh

transcription is in bold and indicated by an arrow.

150

Page 175: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Figure 2- Expression analysis of Tsh.

(A) Northern analysis of tsh expression in tshNG1 and tshA3-2-66 mutants. Lanes 1-5 were loaded with 20 µg of poly(A)+

RNA extracted from homozygous third instar larvae of tshNG1, tshA3-2-66, tshRev1 , tshhs-GAL4:UAS-Tsh and wild-type,

respectively. At the bottom is shown a loading control of the same filter with the NUCB1 probe. (B) Western analysis

of Tsh and Fizzy in CNS cells from tshNG1 and tshA3-2-66 homozygotes. Lanes 1-3 were loaded with proteins extracted

from 12 wild-type third instar larvae, 24 tshNG1 third instar larvae and 18 tshA3-2-66 third instar larvae respectively. The

116 kDa Tsh protein is not detectable in tsh alleles. The same blot probed with anti-Cdc20 (Fizzy) shows equal

amounts of this protein in tsh mutants by comparison with the wild-type.

2- tshNG1 and tshA3-2-66 mutations cause a metaphase-like arrest

tshNG1 homozygotes die as third instar larvae with abnormal trunk. Dissection of these

larvae revealed that imaginal discs are absent or very small and the size of the CNS is also

reduced. This phenotype suggests that tsh gene activity is essential for cell proliferation.

Observations of squashed preparations of the larval CNS from tshNG1 homozygotes revealed

several mitotic defects (Fig.3C, D, G and H). In contrast to wild-type cells (Fig. 3A, B and G),

where hyperploids are never seen, 85% of total mitotic tshNG1 cells (n=1000) contained more

than a diploid complement of chromosomes (Fig. 3C and D). The overall mitotic index in tshNG1

larval CNS was almost three times higher than wild-type (Fig. 3G) and the frequency of

anaphases is very low (0.2%, Fig. 3H), indicative of a metaphase-like arrest. Moreover, in

most mitotic cells, chromosomes are much more highly condensed than that seen in wild-type,

indicating that cells have been delayed in mitosis for some time. A proportion of cells show a

high ploidy number (15-20%, Fig. 3D), suggesting that some metaphase-arrested cells are

capable of reverting to interphase and undergo DNA replication in the absence of chromosome

segregation or cytokinesis.

151

Page 176: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Figure 3- Mitotic phenotypes in tsh

mutant larval brains.

Wild-type control (Oregon-R) metaphase (A)

and anaphase (B) figures. (C) Aneuploid

metaphase figure with hypercondensed

chromosomes from a tshNG1 homozygote. (D)

Aneuploid overcondensed mitotic figure from a

tshNG1 homozygote. (E) Hypercondensed

metaphase figure from a tshA3-2-66 larval

neuroblast. (F) Anaphase figure with lagging

chromatids (arrows) from a tshNG1/tshA3-2-66

trans-heterozygote. (G and H) Mitotic

parameters in larval brains. Error bars

represent standard error of the means. At least

500 fields were counted. (G) Mitotic index is

calculated as the number of mitotic cells per

microscope field using a 100 x objective and 10x/25 eyepieces. (H) Percentage of anaphases is calculated as 100x

(number of anaphases)/ (number of mitotic cells). Bar in A represent 10 µm and is the same for all panels from A-F.

tshA3-2-66 homozygotes also die in third instar larvae with very reduced imaginal discs

and a smaller CNS. tshA3-2-66 squashed CNS display mitotic abnormalities such as aneuploidy

and/or hypercondensed chromosomes (Fig. 3E). The proportion of anaphase figures in tshA3-2-

66 is higher (10%) than in tshNG1 (0.2%), but 3.5 times lower than in wild-type (Fig. 3H),

indicative of a significant delay in the metaphase-anaphase transition. Moreover, 49% of tshA3-

2-66 homozygotes anaphases are abnormal: 24% are disorganised, 16% show lagging

chromatids and 9% are overcondensed (n=1000). Paradoxically, the mitotic index of tshA3-2-66

brains is 2.4 times lowers than wild-type, while the tshNG1 allele causes a significant increase

(Fig. 3G). For note, tshA3-2-66 neuroblasts display higher levels of apoptosis, compared to the

wild-type (Supplementary fig.2).Trans-heterozygotes tshNG1/ tshA3-2-66 die as late larvae and

CNS cells show an abnormal phenotype comparable to tshA3-2-66 homozygous (Fig. 3F, G and

H).

To elucidate if the homozygous tshNG1 or tshA3-2-66 larvae were dying earlier, a simple

test for embryonic lethality was carried out (see Materials and Methods). Trans-heterozygotes

tshNG1/tsh8 or tshA3-2-66/tsh8 die at embryogenesis, before hatching, as does the tsh8 null mutant

(Supplementary fig. 3; Fasano et al., 1991; Gallet et al., 1998; Roder et al., 1992). We

examined such eggs but were unable to detect obvious mitotic defects either in syncytial or

cellularized embryos. This may result from the fact that tsh transcripts are initially detected at

the end of 13th nuclear division of the syncytial embryo (Fasano et al., 1991). We conclude

that the mitotic role of tsh occurs later in the development as compared with the homeotic

role, and that tshNG1 and tshA3-2-66 are hypomorphic alleles.

152

Page 177: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Figure 4- Mitotic figures in preparations of larval CNS cells from wild-type and tsh homozygotes.

Microtubules revealed with anti-α-tubulin are shown in green (A-F), centrosomes revealed with anti-γ-tubulin are

shown in green (G-I) and DNA stained with propidium iodide is shown in red. (A and G) Wild-type metaphase. (D)

Wild-type anaphase. Normal bipolar spindles in a tshNG1 polyploid hypercondensed metaphase (B) and in a tshA3-2-66

overcondensed metaphase (C). (E) tshNG1 polyploid cell containing a normal bipolar spindle in an anaphase-like

configuration. (F) Bipolar spindle in an asymmetric anaphase from tshA-3-2-66. Wild-type (G), tshNG1 (H) and tshA-3-2-66 (I)

metaphase cells containing two normal centrosomes. Bar represent 5 µm and is the same for all panels.

3- tsh mutants organise apparently normal spindles

We analysed mitotic spindle structure in tsh mutants by immunostaining whole-mount

preparations of the larval CNS to visualise microtubules and centrosomes. We found that the

majority (80%, n=1000) of mitotic cells observed in tsh mutants develop an apparently

normal bipolar spindle; the aneuploid and polyploid overcondensed metaphases and anaphases

were in association with bipolar focused spindles (Fig. 4B, C, E and F). Moreover, the majority

of the abnormal mitotic figures have a normal number of centrosomes at the two poles (one at

each pole; Fig. 4H and I). Rarely, only 3-5% (n=300) of metaphase-arrested cells of tshNG1

mutant have a monopolar spindle, where the condensed chromosomes are arranged around a

single centrosome (data not shown).

153

Page 178: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Figure 5- Cyclin A and B levels in wild-type and tsh mutant larval CNS cells.

In Cyclin A panel (leftt panel), microtubules are in green, cyclin A is in red and DNA (blue) is stained with Hoechst

33258 in merged (left) images. The monochromatic image on the right is of cyclin A alone, as revealed with the

antibody Rb270. DNA staining alone can be seen in the inserts on bottom right of the merged figures. In cyclin B panel

(revealed with the antibody Rb271; right panel), cyclin B is in green and DNA in red was stained with propidium

iodide, (A and B) Cyclin A in a wild-type (WT) prophase and metaphase cell, respectivaly. (C and D) Absence of cyclin

A staining in tshA3-2-66 and tshNG1 mutant neuroblasts, respectively. (E) Cyclin B in a wild-type (WT) metaphase cell. (F)

Cyclin B in a wild-type (WT) anaphase cell. (G and H) High levels of cyclin B in tshA3-2-66 and tshNG1 mutant condensed

metaphases, respectively. (I and J) Elevated levels of cyclin B in tsh rod and tsh bub1 polyploid metaphases,

respectively.

In Drosophila, degradation of mitotic Cyclins A, B and B3 are required to allow

inactivation of the associated Cdk1, exit from mitosis and restoration of an interphase stage

(Sigrist et al., 1995). Cyclin A and Cyclin B proteins are targeted for destruction during mitosis

by the anaphase-promoting complex (APC/C)-fzy (Charles et al., 1998; Kotani et al., 1998).

We therefore examined whether Cyclin A and B degradation could take place in tshNG1 and

tshA3-2-66 neuroblasts. In wild-type, Cyclin A is abundant in prophase (Fig. 5A) but is degraded

during the prometaphase/ metaphase transition, ahead of Cyclin B, which one is still abundant

154

Page 179: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

at metaphase (Fig. 5D; Parry and O'Farrell, 2001; Sigrist et al., 1995). After the progressive

Cyclin B degradation at various stages of the metaphase-anaphase transition, wild-type

anaphases have low levels of this mitotic Cyclin (Fig. 5E). In metaphase-arrested tshNG1 (Fig.

5C and G) and tshA3-2-66 neuroblasts (Fig. 5B and F) Cyclin A has been normally degraded but

Cyclin B is still detected.

4- Strong tsh mitotic mutations do not disrupt the association of centromere

and kinetochore proteins

Rough Deal (Rod) is a checkpoint protein that is required for the metaphase arrest in

response to spindle damage, and is also required during normal mitosis to delay anaphase

onset until all chromosomes are properly attached to the spindle (Scaerou et al., 2001; Basto

et al., 2000; Scaerou et al., 1999). During mitosis, the Rod protein accumulates on

kinetochores until cells reach a pro-metaphase state. In metaphase, when chromosomes are

aligned at the spindle equator, Rod decorates kinetochore spindle fibers in an irregular pattern

(Fig. 6A) and remains associated with separating kinetochores throughout anaphase (Scaerou

et al., 2001; Basto et al., 2000). We examined the localisation of this checkpoint protein on

tshNG1 neuroblast cells. On all the bipolar spindles, Rod protein was localised as in wild-type,

with prominent kinetochore signal in prometaphases, and distributed along spindle

microtubules in metaphase plates (Fig. 6B and C).

Furthermore, we examined the localisation of the kinetochore-associated kinase Bub1

on tshNG1 neuroblast cells. Bub 1 is a classical checkpoint protein that is required for the

metaphase arrest in response to spindle damage (Taylor & McKeon, 1997, Basu et al., 1999).

In wild-type, the Bub1 protein is associated with kinetochores in metaphase cells from the

larval CNS (Fig. 6D) and is barely detected after anaphase onset. We found that in metaphase-

like arrested tshNG1 cells, Bub1 was present on all chromosomes and stained kinetochores pairs

as intensively as in control metaphase cells (Fig. 6E). Moreover, Bub 1 was also localised on

kinetochores of tshNG1 anaphase-like cells (Fig. 6F) clearly indicating that sister chromatids did

not disjoin. To more accurately determine the state of chromosomes in these cells, we looked

at the distribution of Cid protein that binds to the centromeric regions of chromosomes (Blower

& Karpen, 2001). In control cells, the centromeres were closely paired in metaphase (Fig. 6G)

but well separated in anaphase. In the majority of tshNG1 neuroblasts, all the centromeres are

tightly clustered in a metaphase-like alignment (Fig. 6H). Furthermore, in anaphase-like cells

Cid staining revealed chromosomes aligned as pairs, rather than chromatids, that are being

pulled apart by tension on the spindle (Fig. 6I). Together this results indicate that mutant

tshNG1 cells fail to undergo sister chromatid separation even in cells that displayed an

anaphase-like configuration.chromome

155

Page 180: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Figure 6- Distribution of centromere CID and kinetochore components Bub1 and Rod in wild-type

and tshNG1 neuroblast cells.

In Rod panel, DNA is in blue, Rod isin red and microtubules are in green. In Bub and CID panels, DNA is in red and

Bub1 or Cid are in green. (A) Rod in a wild-type metaphase cell. (B and C) tshNG1 metaphase-like cells where Rod

localises both at kinetochores and is associated with KMTs. (D) Bub1 in a wild-type metaphase cell. (E) tshNG1

metaphase-like cell showing normal Bub1 staining at kinetochores. (F) tshNG1 anaphase-like cell showing strong

abnormal staining at both kinetochores revealing that sister chromatid separation did not occur. (G) Cid in a wild-type

metaphase. (H) tshNG1metaphase-like cell showing normal Cid staining at centromeres. (I) tshNG1 anaphase-like cell

stained to reveal Cid, confirming that sister chromatid did not disjoin.pair in metaphase (Fig. 6G) but w

5- tshNG1 mutant cells are in a state of metaphase arrest even in the absence of

the checkpoint proteins Bub1 and Rod

To analyse whether tshNG1 cells were in a state of checkpoint arrest, we examined the

mitotic progression in the larval CNS of tsh cells, which were also homozygous-mutant for the

rod or bub1 genes.

156

Page 181: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Table 1- Mitotic index (number of mitotic cells per optic field) and percentage of anaphase figures in the

larval CNS of wild-type, tsh, rod x5, bub1, tsh rodx5 and tsh bub1 mutants.

Mitotic Index % Anaphase

Wild-type 1.8 17

teashirt (tshNG1) 5.1 0.2

rodx5 0.9 24

teashirt rod x5 5.5 0.6

bub1 0.7 38

teashirt bub1 3.9 0.4

Double mutants tsh rodx5 display a high mitotic index and a phenotype similar to the

tsh mutation alone (Table 1). Neuroblasts show a metaphase-like arrest with very condensed

chromosomes and detectable levels of Cyclin-B (Fig. 5H). Therefore, even a checkpoint

mutation as rod cannot override the arrest imposed by tsh. Like tsh rodx5, tsh bub1 mutants

show a high mitotic index, detectable Cyclin-B levels and a metaphase-like arrest similar to tsh

mutation alone (Table 1 and Fig. 5I). These results indicate that neither Rod or Bub1 are able

to override the metaphase arrest imposed by tsh. Even after relieving the metaphase

checkpoint in tshNG1 mutants, the anaphase-promoting complex (APC) is unable to promote

anaphase onset in tsh mutants. Together, these data strongly suggest that, in the absence of

tsh activity, cells arrest in metaphase even in the presence of an active checkpoint, essentially

because progression is inhibited.

6- Depletion of Tsh function in S2 cells also affects mitotic exit as seen in

mutants

To determine whether there were similar effects on mitotic progression, we performed

Tsh-dsRNAi in Drosophila S2 cells. The S2 tsh-dsRNA-treated cells will be referred

subsequently as Tsh-depleted cells and the control GFP-dsRNA-treated cells will be referred

subsequently as control cells. To analyse the Tsh depleting effects on mitotic S2 cells, we

stained every 24h Tsh-depleted cells and control cells with antibodies against α−tubulin and

γ−tubulin and the DNA.

157

Page 182: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Figure 7- Time course analysis of Tsh RNAi in Drosophila S2 Cells.

(A) Western Blot showing the levels of Tsh in S2 dsRNAi-GFP(+) or dsRNAi-Tsh treated (-) cells over time. C

represented the untreated S2 cells. These blots were also probed with anti γ-tubulin antibody as loading control. (B)

Mitotic index after Tsh RNAi is represented as the average percentage of mitotic cells (n > 1000) in the total

population. (C and C´) Mitotic parameters quantification during the experiment, in control and in cells after Tsh RNAi

(n>1000). (D and D´) Quantification of the observed mitotic abnomalities in control and RNAi depleted cells (n>1000).

As judged by Western Blotting the Tsh levels decreased significantly (90 %) within 72h

after the addition of dsRNA (Fig. 7A). Compared with control cells, Tsh-depleted cells show an

increased of three-four folds, in the mitotic index by 96 h (the maximum of RNAi mitotic

effects; Fig. 7B). Later the mitotic index, decreased, coincident with a reappearance of Tsh

protein (Fig.7A lane 9 and Fig.7B). Examination of Tsh–depleted mitotic cells revealed a high

frequency of metaphases from 72 to 96 h (66%-76% compared with 44%-58% in control

cells; Fig. 7C' and C) and a concomitant decrease in anaphases and telophases, indicating a

prolonged metaphase arrest as in tsh mutant neuroblasts. Moreover, throughout the course of

158

Page 183: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

the experiment, the mitotic apparatus in most control cells was well organized with a bipolar

spindle with a single centrosome at each pole (Fig. 8A). However, in cells exposed to tsh

dsRNA from 72 to 120 h, there was a significant increase in mitotic abnormalities. The most

significant aberrant phenotype was the observation of mitotic cells aligned at the equator of a

typical metaphase-like spindle with condensed chromosomes (~25% of mitotic cells; Fig. 7D'

and 8C). This phenotype was maximal at 96 h (Fig. 7C'). A second class of defects, increased

during RNAi experiment, was the assembling of monopolar spindles containing a single or

clustered centrosome at only one pole (Fig. 7D' and 8B; ~10-17% compared with 1-3 % in

control mitotic cells). The third category of defects, which increased after 72 h of Tsh-RNAi,

were the anaphase-like configurations exhibiting lagging chromosomes (Fig. 7D-7D’ and 8F ;

~7 % compared with ~1.5 % of control mitotic cells). Thus, the ablation of Tsh protein

produced a similar metaphase arrest phenotype and revealed striking similar defects in mitotic

cells by comparison with those seen in mutants. These results are consistent and further

support the observation that tsh plays a crucial role in mitotic exit.

Figure 8- Abnormal mitotic figures observed in

S2 TSH-RNAi cells.

Cells were stained for α−tubulin (green), DNA (blue) and γ-

tubulin (red). DNA staining alone can be seen in the inserts on

bottom right of the merged figures. (A) Normal metaphase

displaying a bipolar spindle in a control cell. (B) Polyploid

metaphase displaying a monopolar spindle with a large

centrosome, observed 120 h after tshRNAi. (C) Condensed

metaphase displaying a bipolar spindle observed 96 h after

tshRNAi. (D) Monopolar spindle associated with a metaphase-like

plate exhibiting condensed chromosomes observed 96 h after

tsh

RNAi. (E) Normal anaphase in a control cell. (F) Anaphase-like

cell showing abnormal lagging chromosomes (arrow). Bar, 5

µm.

7- Microarrays data points to some ontology clusters and putative targets of

the tsh gene

In order to get a rough estimate of gene expression differences in tsh mutant brains as

compared to wild-type brains, we screened the Drosophila set of Affimetrix chips. We detected

up- or down-regulation in several genes which have been ordered by lower bound of fold

change (LBFC), after using a cut-off of 1.3 (appendix I).

159

Page 184: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Table 2- List of the most significant gene culters altered in tshNG1 homozygous neuroblats microarrays with

the respectived P value and total number of genes include.

Gene cluster P= N°

genes

include

extracellular region" genes 0.000000 34

imaginal disc pattern formation" genes 0.000007 5

"cell fate specification" genes 0.000019 6

"lysozyme activity" genes 0.000021 5

"defense response" genes 0.000087 30

"Notch signaling pathway" genes 0.000165 7

"antimicrobial humoral response (sensu Protostomia)" genes 0.000329 5

"chymotrypsin activity" genes 0.000674 15

"wing disc anterior/posterior pattern formation" genes 0.000780 4

The obtained results are in agreement with previously published results regarding Tsh

function, but suggest some interesting new connections.

In terms of function we found 9 gene ontology clusters with a P-value <0.001

(appendix II and Table 2), namely "extracellular region" genes (P=0.000000; 1); "imaginal

disc pattern formation" genes (P=0.000007; 2); "cell fate specification" genes (P= 0.000019;

3); "lysozyme activity" genes (P=0.000021; 4); "defense response" genes (P=0.000087; 5);

"Notch signaling pathway" genes (P=0.000165; 6); "antimicrobial humoral response (sensu

Protostomia)" genes (P=0.000329; 7); "chymotrypsin activity" genes (P= 0.000674; 8) and

"wing disc anterior/posterior pattern formation" genes (P=0.000780; 9). In brief, ontology

data significantly indicates a role in both developmental signal transduction and immune

response.

In terms of individual genes (Table 3 and Appendix I), we found Immune induced

molecule 1 as the most up-regulated gene (7.0 of LBFC). We were unable to identify any

known mitotic or cell cycle gene amongst the most up-regulated genes.

The most down-regulated gene is Ecdysone-inducible gene E2 (-11.7 of LBFC) whose

function is unknown. The fourth most down-regulated target is the recently characterised

interference–Hedgehog gene (iHog, -3.0 of LBFC).

Regarding cell-cycle genes we were unable to identify anyone with an outstanding

down-regulated value: candidates with stronger values of LBFC are Cbl (-1.66), Cdc25-string

(-1.63) and Polo (-1.63).

160

Page 185: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Table 3- The most up- and down-regulated genes in tshNG1 homozygous neuroblats microarrays, the putative

Tsh target genes are also listed with the respective lower bound of fold change (LBFC), The positive LBFC represent

the up regulated genes while the negative LBFC represent the downregulated genes.

Gene LBFC

Immune induced molecule 1 7,0

Hedgehog -1,3

Cdc25-String -1,6

Polo -1,6

Cbl -1,7

Engrailed -1,7

interference Hedgehog -3,0

Ecdysone-inducible gene E2 -11,7

Having in mind that the depletion of tsh seems to cause problems during mitosis, we

looked at Polo levels by immunofluorescence in tsh mutant brains and in westerns blot of S2

cells after the Tsh-depletion (Supplementary fig. 4 and 5). We found that Polo localisation in

mitotic cells occurs in the right places in the right time; however, several mitotic figures do not

show staining in all the expected places, as if Polo levels were not enough to occupy all the

sub-cellular locations and to the whole tissue. Accordingly, we noticed that Polo levels are

significantly down-regulated in S2 cells exposed to the tsh dsRNA from 72 to 120 h. Curiously

this observation is coincident with the maximum of RNAi mitotic effects observed.

Discussion

In this study we report that tsh is required for cell proliferation. Loss of tsh function in

tshNG1 and tshA3-2-66 alleles leads to lethality at third instar larvae. Examination of neuroblasts

dissected from dying third instar tshNG1 and tshA3-2-66 homozygous larvae revealed that cells

became arrested in a metaphase-like state in mitosis. Such a general role to Tsh is unusual,

considering its localised expression pattern and well-documented functions on the specification

of particular structures. Regarding the localisation of the mitotic defects on the larval CNS, we

were unable to detect any regional specificity. Although tsh is not ubiquitous express in the

CNS (Supplementary fig. 6), the same range of mitotic abnormalities was observed throughout

the brain hemispheres and the trunk CNS. Moreover, the same range of mitotic defects occurs

in the imaginal discs of the mutants and was also detected in S2 Drosophila culture cells after

161

Page 186: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

tsh-RNAi, re-enforcing the idea of a universal mitotic role for Tsh. Such a role could be

mediated by a long-range diffusible factor responding to the localised action of the tsh

transcription factor.

Our detailed characterisation of the mitotic phenotype revealed only a few structural

abnormalities or defects, not enough to cause such a strong mitotic arrest. Probably, these are

secondary effects of cells escaping from a strong restriction to proliferation after a long delay.

The metaphase-like arrest phenotype observed in mutants, or after the knock-down of tsh,

does not seem to result from defects in the integrity of the mitotic organising centres. Both γ-

tubulin and Centrosomin (data not shown) show a normal localisation to the centrosomes in

mitosis. Accordingly, centrosomes seem to nucleate normal microtubules and are capable of

assembling focused bipolar spindles and organize a metaphase-like array of chromosomes.

Checkpoint proteins localise properly in mutants as if cells were time-frozen in metaphase. The

spindle checkpoint seems to be active and, weirdly, bub neither rod checkpoint mutants are

unable to override the block imposed by the tsh mutation in double mutant constructs. We

conclude that tsh-deficient cells are restricted to bypass metaphase either because they lack or

over-express a potent regulator of cell proliferation. This may act independently of the

checkpoint machinery or known effectors required for the metaphase-anaphase transition. In

fact, it should be stressed that APC/C mutants, thus lacking fundamental effectors proteins

required for this transition, are able to override the checkpoint in double mutant constructs

with bub (Donaldson et al., 2001a). Therefore, even though small changes in mitotic

parameters may activate the metaphase checkpoint they do not explain the strong restriction

to enter in anaphase. Quite probably the mitotic phenotype reflects an indirect role of Tsh in

cell proliferation acting through a potent regulator.

An overview of the expression pattern by microarray analysis of mutant brains failed to

detect obvious alterations in cell cycle candidate genes. However, a putative link to cell

proliferation control mediated by Hedgehog signalling was found, as we identified the recently

characterised interference–Hedgehog gene (iHog) to be significantly down-regulated.

Interestingly, we also got evidence that Hedgehog signalling is a tsh target in ovaries while

carrying out the characterisation of female-sterile tsh mutants in a independent work (see

chapter II part II of this manuscript).

The Hedgehog (Hh) pathway regulates proliferation in a variety of tissues, but its

specific effects on the cell cycle are unclear. Its role has been controversial, with studies

variably suggesting a stimulatory or an inhibitory effect on proliferation. Recently, a general

role of Hh in the nervous system and other tissues has been proposed (Agathocleous et al.,

2007): hh may convert quiescent stem cells into fast cycling transient amplifying progenitors

that are closer to cell cycle exit and differentiation. The fast cycling cells in Hh gain of function

experiments are pushed out of the cell cycle prematurely, while slow dividing progenitors, with

blocked Hh signalling, exhibit delayed cell cycle exit.

162

Page 187: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

The mechanism of Hh induced proliferation varies depending on the tissues, but

includes the induction and regulation of cell cycle components, including Cyclins D1, D2, B1

and E, CDC25 and N-Myc (Barnes et al., 2001; Benazeraf et al., 2006; Kenney and Rowitch,

2000; Kenney et al., 2004). In the case of CyclinD1 and N-Myc, this effect may be direct as

the Gli transcription factors have been shown to bind to sequences in the promoters of these

genes (Hu et al., 2006).

Work in the Drosophila eye has implicated Hh in the activation of cyclin-D and cyclin-E

(Heberlein et al. 1995; Duman-Scheel et al. 2002; Vrailas and Moses 2006). Moreover, it was

demonstrated that Ci (the most downstream element of the Hh pathway) specifically binds to

the cyclin-E promoter, directly inducing Cyc-E expression (Duman-Scheel et al. 2002).

A recent study in retinal precursor cells points to a conserved function of Hh in the

control of cell cycle kinetics, through the regulation of G1 and G2 length (Locker et al., 2006).

It was reported that Hh is both necessary and sufficient to induce G1 and G2 acceleration

explained by a decrease in both G1 and G2 duration (Locker et al. 2006). These cell cycle

accelerating effects of Hh may be brought about because the expression of key cell cycle

stimulators is influenced by Hh signaling. Cyclin-D1 regulates G1/S transition, and, consistent

with a slower G1 transition, its expression is decreased upon Hh inhibition, as already reported

in the mouse retina (Wang et al. 2005) and other areas of the vertebrate central nervous

system (Kenney and Rowitch 2000). In the Locker and colleagues study (2006), cyclin-B1 and

cdc25C, crucial regulators of G2/M transition and cyclin A, involved in both S- and G2-phase

progression (Collins and Garrett 2005), were proposed as key candidates for Hh effects on G2

length duration. The related cdc25B phosphatase was recently found to be a target of Hh

signalling in the neural tube (Benazeraf et al. 2006).

Theoretically, the Drosophila CDC25 homologue String could also be an hh target in

Drosophila neuroblasts, but our microarray data indicates just a slight reduction on the mitotic

target String. Furthermore, this would provoke problems at the G2/M transition, which we

were unable to find as we only detected problems at exiting mitosis. Therefore other targets

must be involved. The slight reduction on the mitotic target Polo (microarray and RNAi data) is

more consistent with the timing of the observed metaphase arrest (Donaldson et al., 2001b).

However, reduced levels of Polo are not enough to justify such a strong metaphase arrest, and

Polo has never been implicated in the Hedgehog pathway.

Our results failed to identify an obvious cell-cycle target. However, the significant

down-regulation of iHog strongly suggests a link between the observed mitotic arrest and Hh

signalling. A number of receptors facilitate Hh binding to its main receptor Patched (Ptc),

including members of the cell adhesion molecule–related/down-regulated by oncogenes (Cdo)

family (reviewed in Jacob and Lum, 2007). A targeted mouse mutation in Cdo, a gene

encoding an Ig-family transmembrane protein, causes microform holoprosencephaly, a

condition associated with loss of Hh signalling (Cole and Krauss, 2003). The Drosophila

163

Page 188: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

homolog of this gene, iHog, was identified in an RNAi screen to be a positive regulator of Hh

signalling (Lum et al., 2003a), which is consistent with the mouse mutant phenotype. The

extracellular protein CDO, IHOG and its relate BOC have been found to positively regulate

hedgehog signalling through the binding and sequestration of the Hh ligand Ptc, resulting in

locally enhanced signalling (Zhang et al., 2006; Yao et al, 2006). Cdo and Ihog associate with

Hh through a fibronectin type III (FnIII) repeat, a motif with potential for binding sulfate ions

(reviewed in Jacob and Lum, 2007). Indeed, dimerization of Ihog and its conversion from a

weak to a high-affinity Hh-binding molecule can be induced by heparin, a protein with

sulphated polysaccharide modifications. How Ihog and Cdo proteins promote Hh-mediated

responses in coordination with Ptc and other Hh receptors, particularly the heparan sulfate–

modified Dally-like protein (Dlp), which also contributes to the Hh response, remains to be

addressed.

A plausible explanation for our results is that the Hh pathway controls a strong

regulator of the cell cycle progression acting at the metaphase-anaphase transition. Even

though Polo levels are affected in our mutants, the phenotype is not ameliorated by

metaphase checkpoint mutations, and do not fully explain the strong block to progression. This

potent regulator could confer a cytoplasmic state similar to the CSF-cytostatic activity blocking

mitotic progression. We speculate that tsh is required for the transcription of iHog and that,

this ligand is required to modulate Hh signalling in mitosis progression. In the absence of iHog

cells are unable to receive the Hh signal to enter in anaphase and exit mitosis. Further work

will be needed to support our speculative model of Hh signalling in mitosis progression.

164

Page 189: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Supplementary Material

Reversion analysis and P{lacW} remobilization to generate new tsh alleles

For mobilisation of the P{lacW}A3-3-66 insert, w / w; T(2;3)CyO TM6, Tb / P{lacW}A3-3-

66;+ females were crossed to males carrying P{ry+, ∆2-3} on a Stubble marked third

chromosome (Robertson et al., 1988) of genotype w/ Y; T(2;3)CyO TM6, Tb/ S Sp Bl bwD; ∆2-

3, Sb. All the strains used in the reversion scheme carried X-chromosomes with the eye-

marker white (w), and thus individuals carrying the P{lacW} chromosome had coloured eyes

due to the presence of the extra wild-type copy w+ on the second chromosome, while lines

having lost the P{lacW} showed white eyes due to the white background on the X-

chromosome. The F1 males of genotype w/ Y ; P{lacW}A3-3-66/ S Sp Bl bwD ; +/ ∆2-3, Sb were

crossed to w/ w ; T(2;3)CyO TM6, Tb/ P{lacW}A3-3-66; + females. Ten white-eyed males were

found amongst 1622 F2 males examined. The ten putative revertant chromosomes were then

independently re-balanced over T(2;3)CyO TM6, Tb to establish revertant lines and test for

viability and fertility. Molecular analysis of six fertile excising lines demonstrated precise

excision of P{lacW} (data not shown) indicating that the transposon insertion is responsible for

the teashirt phenotype. The other four excision lines were infertile apparently due to imprecise

excisions of P{lacW} (data not shown).

165

Page 190: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Tsh protein is absent in tsh mutant larval CNS

We also performed immunostaining of third instar larval CNS from tshNG1 and tshA3-2-66

mutants using the anti-Tsh antibody and compared with the staining pattern observed in wild-

type larval CNS. Tsh was detected using rat anti-Tsh (1:50, (Gallet et al., 1998) and Alexa

488-conjugated anti-rat (1:200, Molecular Probes). Although both methods yielded similar

results, immunostaining of squashed CNS (Supplementary Fig. 1) gave clearer expression

profiles as compared to whole-mount staining. In cells of tsh mutant homozygotes, no specific

Tsh staining was observed at any stage of the cell cycle consistent with the fact that these

alleles behave as genetic nulls (Supplementary Fig. 1D-I). Otherwise, in wild-type CNS, Tsh

protein specifically localises in both the nuclear and cytoplasmic compartments throughout

interphase to prophase (Supplementary Fig. 1A-C). With nuclear envelope breakdown at

prometaphase, Tsh appears to spread over the cytoplasm until the end of mitosis

(Supplementary Fig. 1A-C, cell in the upper right corner). Thus, the localisation of Tsh in CNS

is basically the same as that observed in wild-type embryos at stage 8 of development, where

Tsh is detected as a diffuse labelling within the nuclei and cytoplasm of the cells in the region

of the future thoracic and abdominal segments (Alexandre et al., 1996; Gallet et al., 1999).

Supplementary Figure 1.-

Immunolocalisation of the Tsh protein in

larval CNS cells.

Tsh protein revealed with anti-Tsh (Gallet et al., 1998) is

shown in green and DNA stained with propidium iodide is

shown in red in the merged images. In the wild-type, Tsh

localises both in nuclear and cytoplasmic compartments

during interphase (A-C). In tshNG1 (D-F) and tshA3-2-66 (G-I)

mutant cells Tsh staining is below the level of detection.

166

Page 191: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Apoptose levels in tsh mutant larval CNS

Supplementary Figure 2- In situ cell death detection.

Labelling of apoptotic nuclei in third instar larval CNS by TdT-mediated dUTP nick end labelling (TUNEL) with FICT anti-

dUTP conjugates was performed using the In Situ Cell Death Detection Kit (Roche) according to the manufacturer’s

instructions. Brains were mounted in Vectashield (Vector Laboratories).

167

Page 192: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Cuticle phenotypes

For cuticle preparations, eggs were allowed to develop at 25ºC for 48 hours and

mounted as described (Andrew et al., 1994). Cuticle preparations from Trans combinations of

the mitotic alleles showed characteristic phenotype defects of tsh mutants (Supplementary Fig.

3). The alleles tshNG1 and tshA3-2-66 in Trans with tsh8 revealed similar abnormalities to tsh8

homozygotes, such as defects in labial mouth derivatives and disruption and sclerotization of

the anal opening (Supplementary Fig. 3C-H). Ventral trunk segments are disrupted in

tshNG1/tsh8 embryos, which show segment fusion within restricted parts of the abdomen and

disruption of the denticle belt (Supplementary Fig. 3C and D); tshA3-2-66/tsh8 display smaller

trunk segments and fewer denticles in the abdominal segments (Supplementary Fig. 3E and

F).

Supplementary Figure 3- Cuticules phenotype.

A comparison of the embryo cuticle abnormalities of wild-type (A and B), tshNG1/tsh8 trans-heterozygotes (C and D),

tshA3-2-66/tsh8 trans-heterozygotes (E and F) and tsh8 homozygotes (G and H). Panels B, D, F and H are enlarged views

of A, C, E and G, respectively. Bar represents 50 µm.

168

Page 193: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Polo expression in tshNG1 larval CNS and in dsRNAi- Tsh treated cells

Supplementary Figure 4-

Immunolocalisation of Polo in larval CNS cells.

Polo protein revealed with anti-Polo (a gift from A.

Tavares) is shown in green and DNA is shown in red.

(A, C and E) In the wild-type metaphase (A), Polo

localises in the kinetochore, in late metaphase (C)

and anaphase (E) the polo protein is also detected in

the poles of the spindle. (B, D and F) In neuroblasts

from tshNG1 third instar larvae, Polo is localised in the

right places and in the right time; however, more

than 50% of the mitotic figures do not show staining

in all the expected places. In some mitotic figures

Polo is not detectable (B), and in other mitotic

figures that resemble an anaphase-like, Polo is only

detectable in the poles of the spindle (D) or only in

the kinetochores (F).

Supplementary Figure 5- Analisys of Polo expression by western blot.

(A) Levels of Polo in S2 dsRNAi-GFP(+) or dsRNAi- TSH treated (-) cells 96h after transfection. These blots were also

probed with anti γ-tubulin antibody as loading control. (B) Western analysis of Polo and α-tubulin in CNS cells from

wild-type and tshNG1 homozygotes. Lanes 1-2 were loaded with proteins extracted from 9 wild-type third instar larvae

and 24 tshNG1 third instar larvae, respectively. The polo protein display significantly lower levels in dsRNAi-Tsh treated

cells and in tshNG1 CNS compared to the control. Mouse Polo antibody (a gift from A. Tavares) was used at 1:1000.

169

Page 194: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Expressition patterning of the tshlacZ in CNS during larvae development

Supplementary Figure 6- A CNS from L1, L2 and L3 larvae instars expressing tshlacZ stained with

X-Gal.

The central chord is strongly labelled as well some particular cells in each lobe. The L1, L2 and L3 represent the first,

the second and the third larvae instar. All painels are at the same amplification.

170

Page 195: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

CHAPTER III

DISCUSSION AND PERSPECTIVES

Page 196: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal
Page 197: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

DISCUSSION AND PERSPECTIVES

The objective of this PhD was to explain the teashirt (tsh) function during cell cycle

progression. For that, it was studied the requirement for tsh in two independent processes in

Drosophila melanogaster: mitosis of neuroblasts and S2 cells, and Follicle stem cells (FSC)

proliferation during oogenesis. The results obtained in Drosphila oogenesis, larval neuroblasts

and S2 cells are in accordance and suggest a conserved tsh function through Hh signalling

during the progression of the cell cycle.

1-Tsh and its paralog Tio repress each other in the tip of the Drosophila ovary,

and both are able to regulate En expression.

Tsh and Tiptop (tio) are localised in the left arm of chromosome 2 and are transcribed

in the same orientation (5’-3’). These genes are spaced 183 Kb apart and encode a protein

with zinc finger (Znf) motifs of the C2H2 type. The tsh tio gene pair seems to have arisen from

a recent duplication event characteristic of Drosophilidae. It seems that tio is more close to the

ancestral gene (Laugier et al., 2005; Laugier, 2005) and also to the murine tsh genes (Caubit

et al., 2000) than tsh.

We found that tsh and tio are co-repressed in the tip of the wild-type ovaries. In tio473

ovaries, Tsh is detected in all TF, independently of flies age. When tsh activity is specifically

removed, Tio is ectopically detected in the more proximal TF (Tsh normal expression domain),

showing that, in wild-type, Tsh represses the expression of tio in these particular cells.

Curiously, Tio was never detected in CC of ∆126/tshA3-2-66 ovaries, even in the case of

ovarioles, where Tsh is totally missing from the germarium (phenotype III). However, this is

not the ideal condition to verify the repression of Tio by Tsh, since ∆126/tshA3-2-66 is not a null

mutant. Although no Tsh protein has been detected by immunostaining, this does not mean

that no Tsh at all has been produced. We speculate that a small amount of Tsh protein may

repress tio expression. Another possibility is the presence of another unknown protein that

represses tio, but not tsh in CC.

According to our data, besides the mutual repression between the two members of tsh

family in Drosophila, Tio can also partially replace the loss of tsh during oogenesis, although

less efficiently.

Tsh and Tio show similar functional properties, when expressed ectopically.

Furthermore, tio is able to rescue the oogenetic defects of ∆126/tshA3-2-66 mutants, indicating

that both genes are functionally equivalent during oogenesis, similar to the reported during

imaginal development (Bessa et al., 2009). In addition, these genes share the ability of being

able to induce giant stalk-like structures when ectopic express.Thus, we conclude that Tsh and

Tio have a redundant function during oogenesis, as described during embryogenesis and

imaginal development (Laugier et al., 2005; Laugier, 2005; Bessa et al., 2009; Datta et al.,

173

Page 198: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

DISCUSSION AND PERSPECTIVES

2009). In accordance with the reported phenotype during embryogenesis (Laugier et al., 2005;

Laugier, 2005), the loss of tsh seems more dramatic for oogenesis than the lost of tio, thus tsh

is maintaining as the more effective of the two duplicated genes also during oogenesis. In the

future will be interesting investigate the behaviour of Tio in late pupa stage, during ovary

formation. In this stage, Tsh is already present in all TF and in the precursors of CC, preceding

En expression. And Tio? tio is already express in TF at this stage; tio sharing the same

expression domain with Tsh? Or, tio expression only started after the downregulation of tsh in

the anterior TF cells? These are some of the interesting aspects to better understand the

transcriptional regulation of tio/ tsh and explain the maintenance of a apparently dispensable

gene in Drosophila.

2-Tsh regulates the expression of Hh and En at the Tip of the Drosophila

germarium

The results obtained during this PhD work showed that, in ovaries, tsh affects FSC

proliferation by mediating Hh expression in TF cells and CC. A reduction in Tsh activity resulted

in the loss of Hh in TF and CC. We also showed that, in tsh mutant ovary, En was absent from

CC, but maintained in TF cells. In addition, the observed phenotype in tsh ovaries resembled

the phenotype of ovaries in which hh was absent (Forbes et al, 1996a; 1996b). Inversely, the

ectopic expression of tsh induced the ectopic expression of en and mimicked the phenotype

resultant from hh-ectopic expression (Forbes et al, 1996a; 1996b). These results support the

proposition that tsh regulates both en and hh expression, and that the transcriptional

regulatory mechanisms in TF and CC are different. In addition, they are in agreement with the

interaction between Tsh and Hh pathways previously described during the cellular identity and

polarity of the embryonic segments (Angelats et al., 2002; Gallet et al., 2000). These studies

showed the requeriment of Tsh and Ci in a redundant manner for the Hh dependent regulation

of wg before stage 10 of embryogenesis. Additionally, it was shown that Tsh does not regulate

the transcription of ptc (the Hh receptor), but is required for the expression of rho, a Hh target

gene. Moreover, it was identified a complex containing both proteins: Tsh and Ci (Angelats et

al., 2002). It is important to notice that tio473 tshRNAi embryos lost En expression in the

ventral epidermis, from embryonic stage 11 onwards (Laugier et al., 2005). The parallel

between our observation during oogenesis and those reported during embryogenesis,

suggested a conserved function of the tsh genes family in the regulation of hh expression, at

least in some Drosophila tissue. However our results don’t allow us to conclude about the

nature of the regulation.

174

Page 199: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

DISCUSSION AND PERSPECTIVES

3-Tsh mediating Hh pathway is required for FSC proliferation

The results obtained during the characterisation of the ovary phenotype of tsh female

sterile mutants, allow us to conclude that tsh (through Hh) is required for the proper FSC

and/or early FC proliferation. In the absence of tsh, FSC fail to proliferate correctly, giving rise

to the accumulation of germline cells and swollen germarium. In contrast, in a wild-type

background, tsh overexpression stimulated FSC proliferation, resulting in the excess of stalk

cells and the developing of giants stak-like structures.

Figure 1- Tsh signalling to Follicle Stem cells (FSC) proliferation mediating Hh expression (adapted

from Kirilly and Xie, 2007).

This proposed model shows that Teashirt (Tsh) is required upstream of hedgehog (Hh) expression for follicle stem cells

(FSC; violet cells) proliferation. In terminal filament (TF brown cells) and cap cells (CC, green cells), Tsh acts upstream

of Hh pathway and positively regulates both en and hh expression. In TF, Tiptop (Tio) can also positively affect

engrailed (en) expression. Hh together with Wingless (Wg), (both from CC), and Glassbottomed boat (Gbb) produced

by posterior escort cells (EC; dark blue cells), will signal for FSC proliferation and maintenance.

We speculate that Tsh in EC cells could also signal for FSC proliferation and/or maintenance. In escort stem cells (ESC;

light blue cells), the presence of Tsh suggests a possible role during germline cyst migration.

(CC) cap cells, (ESC) escort stem cells, (GSC) germline stem cell, [EC (IGS)] inner germarial sheath renamed by

escort cells), (FSC) Follicle stem cell, (TF) terminal filament cells. Teashirt (Tsh), Tiptop (Tio), Hedgehog (Hh),

Engrailed (En), Wingless (Wg), Glassbottomed boat (Gbb), Anterior is to the left and posterior is to the right.

175

Page 200: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

DISCUSSION AND PERSPECTIVES

We propose a model to explain the effect of Tsh in FSC proliferation (Fig.1). Hh and Wg

proteins are both expressed in the tip of the ovary and then diffuse to signal for the proper

proliferation and maintenance of FSC, together with Gbb (which one is produced by posterior

EC). In our model, Tsh regulates En and Hh expression at the tip of the ovary. In TF, Tio is

able to regulate the expression of en, but apparently not hh. The role of Hh signalling in FSC

proliferation is well documented (Forbes et al., 1996a; 1996b; King et al., 2001; Liu and

Montell, 1999; Zhang and kalderon, 2000; 2001). We speculate that Tsh, in the posterior EC,

could be upstream of a signal emanating from these cells for FSC proliferation and/or

maintenance. Also, a speculative role for Tsh in the migration of the cyst may be suggested by

the presence of Tsh in the ESC and progeny.The clarification of the Tsh role in posterior EC and

ESC needs more specific work.

Our finding opens new insights about Tsh function. In the ectopic eye formation, Tsh

promote cell proliferation (Bessa et al. 2002), but our results show by first time the implication

of Tsh in stem cell proliferation (probably by an indirect way through Hh). In combination with

the interaction of the Tshz3 with β-Catenin, (a gene mutated in the majority of the colon

cancer cases), this new tsh function could help us to understand why Tshz1 have been

identified in a screen for potential genes implicated in colon cancer (Scanlan et al., 1998).

In our model, Tsh is presented as a positive regulator. However, we do not know the

mechanisms by which Tsh positively regulates the expression of En and Hh: is it directly or

indirectly? The mechanisms by which Tsh controls the expression of targets genes (Alexandre

et al., 1996a; Graba et al., 1994; Waltzer et al., 2001; Saller et al., 2002) are still poorly

understood. So far, Tsh has been shown to directly repress the expression of modulo

(Alexandre et al., 1996a) which suggested that Tsh may be a repressor. Recently, Tsh was

implicated in a repressor complex with FE65/SET and HDAC that directly silence the expression

of the primate specific caspase-4 gene, which is associated with the progression of the

Alzheimer disease (Kajiwara et al., 2009). Thus, it is tempting to propose that, in TF cells and

CC, Tsh may repress a repressor of hh and/or en. Thus, the positive regulation of hh by Tsh

could be indirect. However, an activater role for Tsh is not completely exclude.

The implication of Tsh in the proliferation of epithelial cells could be broadened to other

epithelium type cells and not be restrict to only FSC and progeny.

One characteristic effect of tsh overexpression during embryogenesis is the homeotic

transformation of the labial segment (the more posterior segment of the embryo head) in T1

segment, easily identified by the presence of the beard (additional group of denticles, specific

of the T1 segment; de Zulueta et al., 1994). Curiously, this duplicated T1 segment is bigger

when compared to a wild-type labial segment, suggesting that the enlargement results from

an increase in the number, or in the size, of the cells. In contrast, among with other

developmental specific defects, the trunk segments of tsh larvae are shorter than the normal

(Fasano et al 1991). Based on our finding during oogenesis, we question, whether tsh could

176

Page 201: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

DISCUSSION AND PERSPECTIVES

also have a role in the proliferation of epidermal cells during embryogenesis. To answer this

question we need to be able of compare the total number of cells, in the transformed T1

homeotic segment when tsh is ectopically expressed, with the total number of cells in the wild-

type labial segment and in the equivalent segment in tsh embryo. Unfortunately, we do not

have the appropriate tools to perform an accurate counting of the total cell number. However,

the size of the epidermal cells seems to be the same in the wild-type T1 and in the labial

transformed T1 segment (data not shown). This observation make us suspected, that the

homeotic transformation of the labial segment in T1 segment is accomplished by an increse in

the total number of epidermal cells. If we could prove this increase in the labial homeotic

transformed T1 segment and, on the other hand, a decrease in the total number of epidermal

cells in the T1 tsh- segment, we will be able to propose a conserved tsh function in the

proliferation of epithelial cells, (since embryonic epidermis and follicle cells are both of the

epithelium type cells).

4-The metaphase/anaphase transition is compromised in the absence of Tsh

This PhD started with the observation that tsh was required for mitosis progression. The

loss of tsh function in tshNG1 and tshA3-2-66 alleles, leads to lethality at third instar larvae. The L3

neuroblasts from tshNG1 and tshA3-2-66 homozygous larvae revealed that cells became arrested

in a metaphase-like state in mitosis. Such a general role for Tsh is unusual, considering its

localised expression pattern and well-documented functions on the specification of particular

structures. Despite the preferential Tsh expression in the ventral chord, we were unable to

detect any regional specificity of the mitotic defects in the CNS. Moreover, the same range of

mitotic defects occurred in S2 Drosophila culture cells after tsh-RNAi, re-enforcing the idea of a

universal mitotic role for Tsh. Such a role could be mediated by a long-range diffusible factor

responding to the localised action of the Tsh transcription factor. Showing some similarities

with the role of Tsh (expressed in TF cell and CC) through Hh, in the proliferation of the FSC

and progeny in Drosophila ovaries (described above).

We question if Tsh metaphase arrest could be a secondary effect of cells escaping from

a strong restriction for proliferation after a long delay. The few mitotic structural defects

observed in tsh mutants are not sufficient to explain the strong metaphase arrest imposed by

the loss of tsh. We speculate that, tsh-deficient cells are restricted to bypass metaphase,

because they lack or over-express a potent regulator of cell proliferation. This may act

dependently of the checkpoint machinery and/or known effectors, required for the metaphase-

anaphase transition. However, metaphase checkpoint can be activated by small changes in

mitotic parameters, but this does not explain the strong restriction to enter anaphase. In other

hand, in contrast to what we observed in tsh mutants, APC/C mutants that lack fundamental

effectors proteins required for metaphase/anaphase transition are able to override the

177

Page 202: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

DISCUSSION AND PERSPECTIVES

checkpoint in double mutant constructions with bub (Donaldson et al., 2001a; 2001b). Thus,

we speculate that the tsh mitotic phenotype reflects an indirect role of Tsh in cell proliferation

acting through a potent regulator.

The analysis of the expression pattern of the mutant’s brains did not show an obvious

cell cycle target gene. Interestingly, the third most significantly down-regulated gene in our

microarray analysis was interference–Hedgehog (iHog), providing a link to cell proliferation

control mediated by Hh signalling.

The Cyclins D1, D2, B1 and E, Cdc25 and N-Myc are some of the cell cycle components

regulated by Hh (Barnes et al., 2001; Benazeraf et al., 2006; Kenney and Rowitch, 2000;

Kenney et al., 2004). In addition, there is evidence of a direct binding between Hh effectors

and the promoter of these genes (cyclin D1, N-Myc, cyclin-D and cyclin-E; Duman-Scheel et al.

2002; Heberlein et al. 1995; Hu et al., 2006; Vrailas and Moses 2006). Another interesting

work reported the importance of Hh in the cell cycle kinetics in mouse retina, relating that to

the expression of key cell stimulators (cyclin-D1, Cyclin-B1 and Cdc25C; Locker et al., 2006).

We failed to identify an obvious cell-cycle target. The significant down-regulation of

ihog, strongly suggests a link between the Tsh-dependent mitotic arrest and the Hh signalling.

iHog, positively regulates hedgehog signalling, resulting in a locally enhanced signalling (Zhang

et al., 2006; Yao et al, 2006). We speculated that Hedgehog pathway controls a strong

regulator of cell cycle progression acting at the metaphase-anaphase transition or provoking

arrest at that stage. One potential candidate could be Polo but methaphase ckeckpoint

mutations no override the strong metaphase arrest displayed by tsh neuroblasts. In addition,

Polo was never implicated in the Hh signalling. Other possibility is that, Hh regulates another

potent regulator that could confer a cytoplasmic state, similar to the CSF-cytostatic activity,

blocking mitotic progression. The abnormalities observed in tsh mutations are similar to the

morula phenotypes (Reed and Orr-Weaver, 1997), in which the metaphase arrest associated

with the stabilisation of a set of mitotic proteins, including Cyclin B, was explained as a failure

to maintain active APC/C. In addition, Cyclin B doesn’t seem to be normal degraded in tsh

mutant’s neuroblasts. Thus, we are tempting to hypothesie that Tsh protein is may required to

promote same aspects of APC/C activity that mediated cyclin B degradation. Again the protein

kinases related to the Drosophila gene polo have been implicated as positive regulators of the

APC/C in budding yeast (Shirayama et al., 1998) Drosophila (Donaldson et al., 2001a) and

mammals (Kotani et al., 1998), but, double mutants tsh bub1 doesn’t override the metaphase

arrest in contrast to the observed in polo bub1 mutants, as explain above.

In our microaarys, the Cbl gene, which is donwregulated with a LBFC of -1.7, could be

a candidate to interfere with the APC/C activity. In fact the Cbl proteins are a family of proteins

conserved throughout the animal kingdom (Thien and Langdon, 2001; Nau and Lipkowitz,

2003). Cbl proteins function as ubiquitin protein ligases (E3s), which mediate the

ubiquitination of activated tyrosine kinases, such as the EGFR, and target them for degradation

178

Page 203: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

DISCUSSION AND PERSPECTIVES

(Thien and Langdon, 2001). We question, in what whether, the reduction in the Cbl activity

could not avoid the degradation of some unknow (until now) factor that confer a cytoplasmic

state of the tsh neuroblasts? And in that case, cbl is a new target of Hh signalling? Or Cbl is a

Tsh target independently of the effect of Tsh in Hh pathway?

We propose that tsh is required for the transcription of ihog and that, this ligand is

required to modulate Hh signalling during mitosis progression. In the absence of iHog, cells are

less sensible to receive the Hh signal, which by an unknown target, is reflected in the

incapacity in to enter in anaphase and exit mitosis. Further work will be needed to investigate

the Tsh and/or Hh signalling target (or targets) that confere to the mitotic tsh neuroblasts this

metaphase arrest described in this manuscript and similar to the CSF-cytostatic activity.

5- Tsh/Hh signalling from flies to vertebrates.

Several characteristics of normal and deregulated development of vertebrate epidermis

and hair follicles, suggested that Sonic hedgehog (Shh) might also act on stem cells in those

tissues. Shh is required for hair follicle development in mice, and Shh mutant phenotypes

include reduced proliferation of ectodermal stem cell derivatives (Chiang et al, 1999; St

Jacques et al., 1998). Conversely, inappropriate mutational activation of the Shh signal

transduction pathway is universally observed in human familial and sporadic basal-cell

carcinomas (BCC; Dahmane et al., 1997; Gailani et al., 1996; Johnson et al., 1996; Xie et al.,

1998). Several observations proposed that activation of this pathway might be obligatory in

the aetiology of BCC and can suffice to initiate this cancer (Fan et al., 1997; Grachtchouk et

al., 2000; Nilsson et al., 2000; Oro et al., 1997; Xie et al., 1998). Moreover, it was suggested

that proliferation of follicular stem cells might normally require Shh, among other factors, and

that mutational activation of the Shh signalling pathway in BCC acts specifically on these stem

cells to increase their cycling frequency or their cell number, or perhaps even to expand their

niche into or towards the surrounding epithelium (Zhang and Kalderon, 2001). Any of these

changes could produce a sustainable large increase in a population of transiently proliferating

cells, which would constitute the bulk of the carcinoma.

Interestingly, preliminary studies in mouse revealed that Tshz3 is expressed in

mesenchyme cells that contribute to form the hairs, closely interacting with epithelia cells that

express Shh (Caubit, X. and Fasano, L., not published). Thus, Tshz could conserve a function

upstream of Hh signalling related to the one that plays in TF and CC during Drosophila

oogenesis.

Unlike to that observed in flies, in vertebrate epidermis and hair Follicles, tsh does not

co-localise with Hh, but we speculate that Tshz3 in mesenchyme could signal neighbour

epithelial cells to express Hh. A relation between Tshz3 and Shh was also demonstrated in the

urinary tract in mice, however, in this case, Tshz3 acts downstream of Shh and BMP4 (Caubit

179

Page 204: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

DISCUSSION AND PERSPECTIVES

et al., 2008). During the embryonic development of the mouse urinary tract, Tshz3,

(expressed in mesenchymal populations), is required for proximal ureteric smooth muscle cells

(SMC) differentiation. Inactivation of Tshz3 causes hydroureter and SMC malformation (Caubit

et al., 2008). Shh in the urinary tract, has a dual role: a proliferative effect on ureteric

mesenchyme and a BMP4 inducer in peri-urothelial mesenchymal cells for differentiation of

ureteric mesenchyme into SMC (Yu et al., 2002). This exclusive, but close localisation of Tshz

and Shh is also observed in other mouse tissue. In that cases, Tshz1 and Tshz3 are expressed

in the same cells that express Ptc, which contacts Shh cells (Caubit, X., Core, N. and Fasano,

L., not published). According to our findings of iHog expression in neuroblasts, TshZ and Ptc

co-localisation could indicate the needs of Tsh to one accurate Shh response. We question if

Tshz could not contribute to make these cells capable of answering to Shh signal.

According to what we found in Drosophila Tshz upregulation in mouse precedes the

satellite cells proliferation in response to a muscle injury (Faralli H. and Fasano L., not

publish). Shh has never been implicated in the proliferation of satellite cells. In opposite, Tshz

probably interacts with the Pax genes, similarly to what has been described during Drosophila

eye development (Bessa et al., 2002).

Despite the differences in some aspects, the examples describe above reveal a positive

TshZ interaction with Shh pathways and/or stem cell proliferation, suggesting some conserved

function, at this level, from flies to vertebrates.

Concluding Remarks:

The analysis of tsh function during Drosophila oogenesis, as well as in neuroblasts and

S2 cells mitosis, suggests that Tsh is required for proper cell proliferation, potentially through

Hh signalling. tsh loss of function induces a strong delay or arrest in cell proliferation.

Moreover, in neuroblasts and S2 cells, the absence of Tsh prevented the exit of mitosis and

induced a strong metaphase arrest. Additionally, hh (in TF and CC) is downregulated in tsh

ovaries, while ihog is significantly downregulated in tsh neuroblasts. Thus, in the two systems,

the loss of tsh is followed by a decrease in the activity of the Hh signalling pathway, affecting

the expression of the hh itself or of the Hh receptor-like ihog. Based in all these similarities

found in both systems, we propose a speculative model where Tsh, acting upstream of Hh

signalling, could affect the progression of cell proliferation (Fig.2).

We propose a general role for Tsh acting upstream Hh signalling for proper progression

of cell proliferation in Drosophila. We proved that during oogenesis, Tsh controls Hh expression

in the tip of the ovary. Unfortunately, our results do not allow us to conclude much about the

nature of this regulation. Further work is needed to explain whether Tsh affects directly or

indirectly hh expression, and the nature of the regulation (repression or activation).

180

Page 205: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

DISCUSSION AND PERSPECTIVES

Figure 2- Proposed model to explain

Tsh role in cell proliferation, mediating Hh

expression signal in FSC and neuroblasts.

This speculative model proposes a role for Tsh in

cell proliferation both in follicle stem cells (FSC, in

ovaries; left circle gray) and in neuroblasts (in CNS;

right circle gray). In FSC, Tsh at the tip of the

germarium, regulates positively Hh expression,

which in turn will positively affect FSC proliferation

through unknown target(s) gene(s). In neuroblast

cells, Tsh affects cell cycle progression probably

through a diffusible factor. Hh is a potential

candidate, because it was shown that Tsh positively

affects iHog expression. We speculate that Polo

could be a new Hh target.

Although the positive regulation of Hh pathway in FSC proliferation is well documented,

some questions remain: How does Hh signals for FSC proliferation? What are the Hh targets in

FSC?

In neuroblasts, we speculate that Tsh is required for the transcription of iHog that by its

turn, is required to enhance Hh signalling and promote mitosis progression through the

regulation of one or more potent cell cycle regulators. Additionally, we question whether Polo

could be a new, but not the only, mitotic target controlled by Hh signalling, at least in

Drosophila neuroblasts.

The present work has shed some light in the biological Tsh function on cell proliferation,

but opens up several questions: How does Tsh regulates hh (in ovaries) and ihog (in

neuroblasts)? What is the nature of this regulation (direct or indirect)? Is Tsh an activator or a

repressor, or both? Is it polo a new mitotic target gene of Hh? Why do egg chambers stop

exactly at stage S2/S3 in absence of tsh and at stage S5/S6 when rescued by ubiquitously

provided tsh? Do tsh mutants allow to identify two checkpoints in egg chamber development?

In the follicular cells, the endocycles begin at stage S6/S7, when N pathway mediates the

downregulation of cut, string and hh, promoting the M/E switch. Could tsh have a later role in

the regulation of this process? Future works are required to try answer all these questions.

181

Page 206: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal
Page 207: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCE

Page 208: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal
Page 209: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Aberle, H., Bauer, A., Stappert, J., Kispert, A., Kemler, R. (1997). beta-catenin is a target for the ubiquitin-

proteasome pathway. Embo J 16, 3797-804.

Abu-Shaar, M., Mann, R. S. (1998). Generation of multiple antagonistic domains along the proximodistal axis during

Drosophila leg development. Development 125, 3821-30.

Adams, R. R., Maiato, H., Earnshaw, W. C., Carmena, M. (2001). Essential roles of Drosophila inner centromere

protein (INCENP) and aurora B in histone H3 phosphorylation, metaphase chromosome alignment,

kinetochore disjunction, and chromosome segregation. J Cell Biol 153, 865-80.

Adolphe, C., Hetherington, R., Ellis, T., Wainwright, B., (2006). Patched1 Functions as a Gatekeeper by

Promoting Cell Cycle Progression. Cancer Res. 66, 2081-8.

Affolter, M., Basler, K. (2007). The Decapentaplegic morphogen gradient: from pattern formation to growth

regulation. Nat Rev Genet. 8, 663-74 .

Agathocleous, M., Locker A. M, Harris W. A,, Perron, M. (2007) A general role of hedgehog in the regulation of

proliferation. .Cell Cycle 6, 156-9.

Ahonen, L.J., Kallio, M. J., Daum, J. R., Bolton, M., Manke, I. A., Yaffe, M. B., Stukenberg, P. T.,,Gorbsky, G.

J. (2005). Polo-like kinase 1 creates the tension-sensing 3F3/2 phosphoepitope and modulates the

association of spindle-checkpoint proteins at kinetochores. Curr. Biol. 15, 1078–89.

Alberts B., D. Bray, Lewis J., Raff, M., Roberts, K., Watson J. D. (1994). Molecular Biology of the Cell. 3rd

Edition. Garland Publishing Inc.

Alexandre, C., Jacinto, A., Ingham, P.W., (1996a). Transcriptional activation of hedgehog target genes in

Drosophila is mediated directly by the cubitus interruptus protein, a member of the GLI family of zinc finger

DNA-binding proteins. Genes Dev 10, 2003–13.

Alexandre, C., Lecourtois, M., Vincent, J. (1999). Wingless and Hedgehog pattern Drosophila denticle belts by

regulating the production of short-range signals. Development 126, 5689-98.

Alexandre, E., Graba, Y., Fasano, L., Gallet, A., Perrin, L., De Zulueta, P., Pradel, J., Kerridge, S., Jacq, B.

(1996b). The Drosophila teashirt homeotic protein is a DNA-binding protein and modulo, a HOM-C regulated

modifier of variegation, is a likely candidate for being a direct target gene. Mech Dev 59, 191-204.

Alexandru, G., Zachariae, W., Schleiffer, A., Nasmyth, K. (1999). Sister chromatid separation and chromosome

re-duplication are regulated by different mechanisms in response to spindle damage. Embo J 18, 2707-21.

Altschul, S. F., Gish, W., Miller, W., Myers, E. W., Lipman, D. J. (1990). Basic local alignment search tool. J Mol

Biol 215, 403-10

Amon, A. (1999). The spindle checkpoint. Curr Opin Genet Dev 9, 69-75.

Andrew, D. J., Horner, M. A., Petitt, M. G., Smolik, S. M., Scott, M. P. (1994). Setting limits on homeotic gene

function: restraint of Sex combs reduced activity by teashirt and other homeotic genes. Embo J 13, 1132-44

Angelats, C., Gallet, A., Therond, P., Fasano, L., Kerridge, S. (2002). Cubitus interruptus acts to specify naked

cuticle in the trunk of Drosophila embryos. Dev Biol 241, 132-44.

Ashburner, M. (1989). Drosophila: A Laboratory Handbook. Cold Spring Harbor Press, Cold Spring Harbor.

Assa-Kunik, E., Torres, I. L., Schejter, E. D., St Johnston, D., Shilo, B. Z., (2007). Drosophila follicle cells are

patterned by multiple levels of Notch signaling and antagonism between the Notch and JAK/STAT pathways.

Development 134, 1161-1169.

Atherton F. S., Parker L. L., Geahlen R. L., Piwnica W. H. (1993). Mechanisms of p34 cdc2 regulation. Mol Cell Biol 13, 1675-85.

Aza-Blanc, P., Kornberg T. B. (1999). Ci: a complex transducer of the hedgehog signal. Trends Genet. 15, 458-62.

Aza-Blanc, P., Ramirez-Weber, F. A., Laget, M. P., Schwartz, C., Kornberg, T. B. (1997). Proteolysis that is

inhibited by hedgehog targets Cubitus interruptus protein to the nucleus and converts it to a repressor. Cell

89, 1043–53.

Azpiazu, N., Morata, G. (2002). Distinct functions of homothorax in leg development in Drosophila. Mech Dev 119,

55-67.

Bai, J., Montell, D. (2002) Eyes Absent, a key repressor of polar cell fate during Drosophila oogenesis. Development

129,5377-88.

Page 210: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Baker, N. (1988). Localization of transcripts from the wingless gene in whole Drosophila embryos. Development 1988,

103, 289-98.

Baonza, A., Freeman, M., (2005). Control of cell proliferation in the Drosophila eye by Notch signaling. Dev Cell. 8,

529-39.

Barker, D. D., Wang C., Moore, J., Dickinson L. K., Lehmann R. (1992). Pumilio is essential for function but not

for distribution of the Drosophila abdominal determinant Nanos. Genes Dev 6, 2312–26.

Barnes, E. A., Kong, M., Ollendorff, V., Donoghue, D. J., (2001). Patched1 interacts with cyclin B1 to regulate cell

cycle progression. EMBO J. 20, 2214–23.

Basto, R., Gomes, R., Karess, R. E. (2000). Rough deal and Zw10 are required for metaphase checkpoint in

Drosophila. Nat Cell Biol 2, 939-43.

Basu, J., Bousbaa, H., Logarinho, E., Li, Z., Williams, B. C., Lopes, C., Sunkel, C. E., Goldberg, M. L. (1999).

Mutations in the essential spindle checkpoint gene bub1 cause chromosomes missegregation and fail to block

apoptosis in Drosophila. J Cell Biol 146, 13-28.

Baumann, C., Korner, R., Hofmann, K., Nigg, E. A. (2007). PICH, a centromere-associated SNF2 family ATPase, is

regulated by Plk1 and required for the spindle checkpoint. Cell 128, 101–14.

Becker, J. D., Boavida, L. C., Carneiro, J., Haury, M., Feijó, J. A. (2003). Transcriptional profiling of Arabidopsis

tissues reveals the unique characteristics of the pollen transcriptome. Plant Physiol 133, 713-25.

Behrens, J., von Kies, J., Kühl, M., Wedlich, D., Grosschedl, R., Birchmeier, W. (1996). Functional interaction of

b-catenin with the transcription factor LEF-1. Nature 382:638-42.

Bejsovec, A., Martinez-Arias, A. (1991). Roles of wingless in patterning the larval epidermis of Drosophila.

Development 113, 471-85.

Benazeraf, B., Chen, Q., Peco, E., Lobjois, V., Medevielle, F., Ducommun, B., Pituello, F., (2006). Identification

of an unexpected link between the Shh pathway and a G2/M regulator, the phosphatase CDC25B. Dev. Biol.

294, 133–47.

Bergeron, S. A., Milla, L. A., Villegas, R., Shen, M., Burgess, S. M., Allende, M. L., Karlstrom, R. O., Palma, V.

(2008). Expression profiling identifies novel Hh/Gli regulated genes in developing zebrafish embryos.

Genomics 91, 165-77.

Bessa, J., Carmona, L., Casares, F. (2009). Zinc-finger Paralogues tsh and tio Are Functionally Equivalent During

Imaginal Development in Drosophila and Maintain Their Expression Levels Through Auto- and Cross-Negative

Feedback Loops. Dev. Dynamics 238, 19–28.

Bessa, J., Casares, F. (2005). Restricted teashirt expression confers eye-specific responsiveness to Dpp and Wg

signals during eye specification in Drosophila. Development 132, 5011–20.

Bessa, J., Gebelein, B., Pichaud, F., Casares, F., Mann, R. S. (2002). Combinatorial control of Drosophila eye

development by eyeless, homothorax, and teashirt. Genes Dev 16, 2415-27.

Besse, F., Pret, A. M., (2003). Apoptosis-mediated cell death within the ovarian polar cell lineage of Drosophila

melanogaster. Development 130, 1017-27.

Bhalla, K. N. (2003). Microtubule-targeted anticancer agents and apoptosis. Oncogene 22, 9075–86.

Bharadwaj, R., Yu, H. (2004). The spindle checkpoint, aneuploidy, and cancer. Oncogene 23, 2016–27.

Bhat, K., Schedl, P. (1997). Establishment of stem cell identity in the Drosophila germline. Dev. Dyn. 210, 371-82.

Bhojwani, J., Shashidhara, L. S., Sinha, P. (1997). Requirement of teashirt (tsh) during cell fate specification in

developing head structures in Drosophila. Dev. Genes Evol. 207, 137-46.

Bienz, M. (1997). Endoderm induction in Drosophila: The nuclear targets of the inducing signals. Curr. Opin. Genet.

Dev.7, 683–88.

Bienz, M., Tremml, G. (1988). Domain of Ultrabithorax expression in Drosophila visceral mesoderm from

autoregulation and exclusion. Nature 33, 576–8.

Bjerknes, M., Cheng, H. (1999). Clonal analysis of mouse intestinal epithelial progenitors. Gastroenterology 1999;

116:7-14.

186

Page 211: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Blasina, A., de Weyer, I. V., Laus, M. C., Luyten, W. H., Parker, A. E., McGowan, C. H. (1999). A human

homologue of the checkpoint kinase Cds1 directly inhibits Cdc25 phosphatase. Curr Biol 9, 1-10.

Blower, M. D., Karpen, G. H. (2001). The role of Drosophila CID in kinetochore formation, cell-cycle progression and

heterochromatin interactions. Nat Cell Biol. 3, 730-9.

Bohni, R., Riesgo-Escovar, J., Oldham, S., Brogiolo, W., Stocker, H., Andruss, B. F., Beckingham, K., Hafen,

E. (1999). Autonomous control of cell and organ size by CHICO, a Drosophila homolog of vertebrate IRS1-4.

Cell.97, 865-75.

Bopp, D., Horabin, J. I., Lersch, R. A. , Cline, T. W., Schedl, P. (1993). Expression of the Sex-lethal gene is

controlled at multiple levels during Drosophila oogenesis. Development 118, 797–812.

Bornens, M. (2002). Centrosome composition and microtubule anchoring mechanisms. Curr. Opin. Cell Biol. 14, 25–

34.

Bownes, M., Blair, M. (1986). The effects of a sugar diet and hormones on the expression of the Drosophila yolk-

protein genes. J. Insect Physiol. 32, 493– 501.

Brand, A. H., Perrimon, N. (1993). Targeted gene expression as a means of altering cell fates and generating

dominant phenotypes. Development 118, 401-15.

Brandeis, M., Rosewell, I., Carrington, M., Crompton, T., Jacobs, M. A., Kirk, J., Gannon, J., Hunt, T. (1998).

Cyclin-B2 null mice develop normally and are fertile whereas cyclin B1-null mice die in utero. Proc Natl Acad

Sci USA, 95:4344-49.

Brawley, C., Matunis, E. (2004). Regeneration of male germline stem cells by spermatogonial dedifferentiation in

vivo. Science 304, 1331–34.

Brunner, E., Peter, O., Schweizer, L., Basler, K. (1997). pangolin encodes a Lef-1 homologue that acts

downstream of Armadillo to transduce the Wingless signal in Drosophila. Nature 385, 829-33.

Burton, J. L., Solomon, M. J. (2001). D box and KEN box motifs in buddun yeast Hssl1p are required APC-mediated

degradation and direct biding to Cdc20p and Cdh1p. Genes Dev 15, 2381-95.

Calvi, B. R., Lilly, M. A., Spradling, A.C. (1998). Cell cycle control of chorion gene amplification. Genes Dev.12,

734–44.

Campos-Ortega, J. A., Hartenstein, V. (1985). The Embryonic Development of Drosophila melanogaster. New York:

Springer Verlag.

Casanueva, M. O., Ferguson, E. L. (2004). Germline stem cell number in the Drosophila ovary is regulated by

redundant mechanisms that control Dpp signalling. Development 131, 1881–90.

Casares, F., Mann, R. S. (2000). A dual role for homothorax in inhibiting wing blade development and specifying

proximal wing identities in Drosophila. Development 127, 1499-508.

Casares, F., Mann, R. S. (2000). A dual role for homothorax in inhibiting wing blade development and specifying

proximal wing identities in Drosophila. Development 127, 1499-508.

Caubit, X., Core, N., Boned, A., Kerridge, S., Djabali, M., Fasano, L. (2000). Vertebrate orthologues of the

Drosophila region-specific patterning gene teashirt. Mech Dev 91, 445-8.

Caubit X, Lye C. M., Martin E., Coré N., Long D. A., Vola, C., Jenkins, D., Garratt, A. N., Skaer, H., Woolf, A.

S, Fasano, L. (2008). Teashirt 3 is necessary for ureteral smooth muscle differentiation downstream of SHH

and BMP4.Development 135, 3301-10.

Caubit, X., Tiveron, M.C., Cremer, H., Fasano, L., (2005). Expression patterns of the three Teashirt-related genes

define specific boundaries in the developing and postnatal mouse forebrain. J. Comp. Neurol. 486, 76–88.

Cavodeassi, F., Rodriguez, I., Modolell, J. (2002). Dpp signalling is a key effector of the wing-body wall subdivision

of the Drosophila mesothorax. Development 129, 3815-23.

Chan, G. K., Liu, S. T., Yen, T. J. (2005). Kinetochore structure and function. Trends Cell Biol. 15, 589–98.

Charles, J. F., Jaspersen, S. L., Tinker-Kulberg, R. L., Hwang, L., Szidon, A., Morgan, D. O. (1998). The Polo-

related kinase Cdc5 activates and is destroyed by the mitotic cyclin destruction machinery in S. cerevisiae.

Curr Biol 8, 497-507.

187

Page 212: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Cheeseman, I. M., Chappie, J. S., Wilson-Kubalek, E. M., Desai, A. (2006). The conserved KMN network

constitutes the core microtubule-binding site of the kinetochore. Cell 127, 983–97.

Cheeseman, I. M., Niessen, S., Anderson, S., Hyndman, F., Yates, J.R. 3rd, Oegema, K., Desai, A. (2004). A

conserved protein network controls assembly of the outer kinetochore and its ability to sustain tension. Genes

Dev. 18, 2255–68.

Chen, C., Jack, J., Garofalo, R. S. (1996a). The Drosophila insulin receptor is required for normal growth.

Endocrinology 137, 846–56.

Chen, D., McKearin, D. (2003). Dpp signalling silences bam transcription directly to establish asymmetric divisions of

germline stem cells. Curr Biol 13, 1786–91.

Chen, D., McKearin, D. (2005). Gene circuitry controlling a stem cell niche. Curr Bio.15,179–84.

Chen, J., Godt, D., Gunsalus, K., Kiss, I., Goldberg, M., Laski, F. (2001). Cofilin/ADF is required for cell motility

during Drosophila ovary development and oogenesis. Nat. Cell Biol. 3, 204-09.

Chen, R. H. (2002). BubR1 is essential for kinetochore localization of other spindle checkpoint proteins and its

phosphorylation requires Mad1. J. Cell Biol. 158, 487–96.

Chen, R. H. (2004). Phosphorylation and activation of Bub1 on unattached chromosomes facilitate the spindle

checkpoint. EMBO J. 23, 3113–21.

Chen, R. H., Waters, J. C., Salmon, E. D., Murray, A. W. (1996b). Association of spindle assembly checkpoint

component XMAD2 with unattached kinetochores. Science 274, 242–6.

Chen, Y., Struhl, G. (1996). Dual roles for patched in sequestering and transducing Hedgehog. Cell 87, 553-63.

Chiang, C., Swan, R. Z., Grachtchouk, M., Bolinger, M., Litingtung, Y., Robertson, E. K., Cooper, M. K.,

Gaffield, W., Westphal, H., Beachy, P. A., Dlugosz, A. A. (1999). Essential role for Sonic hedgehog

during hair follicle morphogenesis. Dev Bio 205, 1-9.

Christerson, L. B., McKearin, D. M. (1994). orb is required for anteroposterior and dorsoventral patterning during

Drosophila oogenesis. Genes Dev 8, 614–28.

Chung, E., Chen, R. H. (2003). Phosphorylation of Cdc20 is required for its inhibition by the spindle checkpoint. Nat.

Cell Biol. 5, 748–53.

Cifuentes, F. J., Garcia-Bellido, A. (1997). Proximo-distal specification in the wing disc of Drosophila by the nubbin

gene. Proceedings of the National Academy of Sciences of the United States of America 94, 11405–10.

Cimini, D., Degrassi, F. (2005) Aneuploidy: a matter of bad connections. Trends Cell Biol. 15, 442–51.

Clarke, M. F., Fuller, M. (2006). Stem cells and cancer: two faces of eve. Cell 124, 1111-5.

Cleard and Spierer, 2001

Clemens, J. C., Worby, C. A., Simonson-Leff, N., Muda, M., Maehama, T., Hemmings, B. A., Dixon, J. E.

(2000). Use of double-stranded RNA interference in Drosophila cells lines to dissect signal transduction

pathways. Proc Natl Acd Sci USA 97, 6499-503.

Cleveland, D. W., Mao, Y., Sullivan, K. F. (2003). Centromeres and kinetochores: from epigenetics to mitotic

checkpoint signaling. Cell 112, 407–21.

Clute, P., Pines, J. (1999). Temporal and spatial control of cyclin B1 destruction in metaphase. Nature Cell Biol.1,

82–7.

Cody, J. D., Ghidoni, P. D., DuPont, B. R., Hale, D. E., Hilsenbeck, S. G., Stratton, R. F., Hoffman, D. S.,

Muller, S., Schaub, R. L., Leach, R. J., Kaye, C. I., (1999). Congenital anomalies and anthropometry of 42

individuals with deletions of chromosome 18q. Am J Med Gene. 85, 455–62.

Cohen, S. M., (1990). Specification of limb development in the Drosophila embryo by positional cues from

segmentation genes. Nature 343: 173–7.

Cole, F., Krauss, R. S. (2003) Microform holoprosencephaly in mice that lack the Ig superfamily member Cdon. Curr

Biol. 13, 411-5.

Collins, I., Garrett, M. D. (2005) Targeting the cell division cycle in cancer: CDK and cell cycle checkpoint kinase

inhibitors. Curr Opin Pharmacol. 5, 366-73.

188

Page 213: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Coré, N., Caubit, X., Metchat, A., Boned, A., Djabali, M., Fasano, F. (2007). Tshz1 is required for axial skeleton,

soft palate and middle ear development in mice. Dev Biol 308, 407–20.

Core, N., Charroux, B., McCormick, A., Vola, C., Fasano, L., Scott, M. P., Kerridge, S. (1997). Transcriptional

regulation of the Drosophila homeotic gene teashirt by the homeodomain protein Fushi tarazu. Mech Dev 68,

157-72.

Corona, D. F., Langst, G., Clapier, C. R., Bonte, E. J., Ferrari, S., Tamkun, J. W., Becker, P. B. (1999). ISWI is

an ATP-dependent nucleosome remodeling factor. Mol Cell 3, 239-45.

Cox, D. N., Chao, A., Baker, J., Chang, L., Qiao, D., Lin, H. (1998). A novel class of evolutionarily conserved genes

defined by piwi are essential for stem cell self-renewal. Genes Dev 12, 3715–27.

Cox, D. N., Chao, A., Lin, H. (2000). piwi encodes a nucleoplasmic factor whose activity modulates the number and

division rate of germline stem cells. Development 127, 503–14.

Cremers, C. W., Teunissen, E., Marres, E. H., (1988). Classification of congenital aural atresia and results of

reconstructive surgery. Adv. Otorhinolaryngol 40, 9–14.

Crosby, M. E., Jacobberger, J., Gupta, D., Macklis, R. M., Almasan, A. (2007). E2F4 regulates a stable G2 arrest

response to genotoxic stress in prostate carcinoma. Oncogene 26, 1897-909.

Curtiss, J., Mlodzik, M. (2000). Morphogenetic furrow initiation and progression during eye development in

Drosophila: The roles of decapentaplegic, hedgehog and eyes absent. Development 127: 1325–36.

Czerny, T., Halder, G., Kloter, U., Souabni, A., Gehring, W. J., Busslinger, M. (1999). twin of eyeless, a second

Pax-6 gene of Drosophila, acts upstream of eyeless in the control of eye development. Mol Cell 3, 297-307.

D’Angiolella, V., Mari, C., Nocera, D., Rametti, L., Grieco, D. (2003). The spindle checkpoint requires cyclin-

dependent kinase activity. Genes Dev. 17, 2520–5.

Dahmane, N., Lee, J., Robins, P., Heller, P., Ruiz, I., Altaba, A. (1997). Activation of the transcription factor Gli1

and the Sonic hedgehog signalling pathway in skin tumours. Nature. 389, 876-81 Erratum in:

Dahmann, C.,Futcher, B. (1995) Specialization of B-cyclins for mitosis or meiosis in S. cerevisae. Genetics, 140,

957-63.

Datta, R. R., Lurye, J. M., Kumar, J. P. (2009). Restriction of Ectopic Eye Formation by Drosophila Teashirt and

Tiptop to the Developing Antenna. Dev Dyn. 238, 2202–2210.

Dawson, I. A., Roth, S., Artavanis-Tsakonas, S. (1995). The Drosophila cell cycle gene fizzy is required for normal

degradation of cyclin A and B during mitosis and has homology to the CDC20 gene of Saccharomyces

cerevisiae. J Cell Biol 129, 725-37.

de Celis, J. F., Baonza, A., Garcia-Bellido, A. (1995). Behavior of extramacrochaetae mutant cells in the

morphogenesis of the Drosophila wing. Mech Dev 53, 209–21.

de Cuevas, M., Lee, J. K., Spradling A. C. (1996). alpha-spectrin is required for germline cell division and

differentiation in the Drosophila ovary. Development 122, 3959-68.

de Zulueta, P., Alexandre, E., Jacq, B., Kerridge, S. (1994). Homeotic complex and teashirt genes co-operate to

establish trunk segmental identities in Drosophila. Development 120, 2287-96.

Decotto, E., Spradling, A. C. (2005). The Drosophila ovarian and testis stem cell niches: similar somatic stem cells

and signals. Dev Cell. 9, 501-10.

DeLuca, J. G., Gall, W. E, Ciferri, C., Cimini, D., Musacchio, A., Salmon, E. D. (2006). Kinetochore microtubule

dynamics and attachment stability are regulated by hec1. Cell 127, 969–82.

Deluca, J. G., Moree, B., Hickey, J. M., Kilmartin, J. V., Salmon, E. D. (2002). hNuf2 inhibition blocks stable

kinetochore-microtubule attachment and induces mitotic cell death in HeLa cells. J. Cell Biol. 159, 549–55.

DeLuca, J. G., Salmon, E. D. (2004). Kinetochores: if you build it, they will come. Curr. Biol. 14, R921–3.

Deming, P. B., Flores, K. G., Downes, C. S., Paules, R. S., Kaufmann, W. K. (2002). ATR enforces the

topoisomerase II-dependent G2 checkpoint through inhibition of PLk1 kinase. J Biol Chem 277, 36832-8.

Den Elzen, N., Pines, J. (2001) Cyclin A is destroyed in prometaphase and can delayed chromosomes alignment and

anaphase. J Cell Biol 153, 121-36.

189

Page 214: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Denholm, B., Sudarsan, V., Pasalodos-Sanchez, S., Artero, R., Lawrence, P., Maddrell, S., Baylies, M.,

Skaer, H. (2003). Dual origin of the renal tubules in Drosophila: mesodermal cells integrate and polarize to

establish secretory function. Curr Biol 13, 1052-7.

Desplan, C. (1997). Eye development: governed by a dictator or a junta? Cell 91, 861-4.

Diaz-Benjumea, F. J., Cohen, B., Cohen, S. M. (1994). Cell interaction between compartments establishes the

proximal-distal axis of Drosophila legs. Nature 372, 175-79.

DiNardo, S., Sher, E., Heemskerk-Jongens, J., Kassis, J. A., O'Farrell, P. H. (1988). Two-tiered regulation of

spatially patterned engrailed gene expression during Drosophila embryogenesis. Nature 332, 604-9.

Dominguez, M. (1999). Dual role for Hedgehog in the regulation of the proneural gene atonal during ommatidia

development. Development 126, 2345-53.

Dominguez, M., Brunner, M., Hafen, E., Basler, K. (1996). Sending and receiving the hedgehog signal: control by

the Drosophila Gli protein Cubitus interruptus. Science. 14, 1621-25.

Dominguez, M., Casares, F. (2005). Organ specification-growth control connection: New in-sights from the

Drosophila eye-antennal disc. Dev. Dyn.232, 673-84.

Dominguez, M., Hafen, E. (1997). Hedgehog directly controls initiation and propagation of retinal differentiation in

the Drosophila eye. Genes Dev.11, 3254-64.

Donaldson M. M., Tavares A. A., Ohkura H., Deak P., Glover D. M. (2001 b) Metaphase arrest with centromere

separation in polo mutants of Drosophila. J Cell Biol. 153, 663-76.

Donaldson, M. M., Tavares, A. A., Hagan, I. M., Nigg, E. A., Glover, D. M. (2001a) The mitotic roles of Polo-like

Kinase. J Cell Sci. 114, 2357-8.

Dougan, S., DiNardo, S. (1992). Drosophila wingless generates cell type diversity among engrailed expressing cells.

Nature 360, 347-50.

Draviam, V. M., Stegmeier, F., Nalepa, G., Sowa, M. E., Chen, J., Liang, A., Hannon, G. J., Sorger, P. K.,

Harper, J. W., Elledge, S. J. (2007). A functional genomic screen identifies a role for TAO1 kinase in

spindle-checkpoint signalling. Nat. Cell Biol. 9, 556–64.

Drummond-Barbosa D., Spradling, A. C. (2004). Alpha-endosulfine, a potential regulator of insulin secretion, is

required for adult tissue growth control in Drosophila. DevBiol 266, 310-21.

Drummond-Barbosa, D., Spradling, A. C. (2001). Stem cells and their progeny respond to nutritional changes

during Drosophila oogenesis. Dev. Biol. 231, 265–78.

Du, Q., Stukenberg, P. T., Macara, I. G. (2001). A mammalian Partnerof inscuteable binds NuMA and regulates

mitotic spindle organization. Nat. Cell Biol. 3, 1069–75.

Duilio, A., Zambrano, N., Mogavero, A. R., Ammendola, R., Cimino, F., Russo, T. (1991). A rat brain mRNA

encoding a transcriptional activator homologous to the DNA binding domain of retroviral integrases. Nucleic

Acids Res 19, 5269–5274.

Duman-Scheel, M., Weng, L., Xin, S., Du, W., (2002). Hedgehog regulates cell growth and proliferation by inducing

Cyclin D and Cyclin E. Nature 417, 299–304.

Dworkin, I., Gibson, G. (2006). Epidermal Growth Factor Receptor and Transforming Growth Factor-b Signaling

Contributes to Variation for Wing Shape in Drosophila melanogaster. Genetics 173, 1417–1431.

Echeverri, C. J., Paschal, B. M., Vaughan, K. T., Vallee, R. B. (1996). Molecularcharacterization of the 50-kD

subunit of dynactin reveals function for the complex in chromosome alignment and spindle organization

during mitosis. J Cell Biol 132, 617-33.

Elledge, S. J. (1996). Cell cycle checkpoints: preventing a identity crisis. Science 274, 1664-72.

Elzen, N., Pines, J. (2001) Cyclin A is destroyed in prometaphase and can delay chromosome alignment and

anaphase. J Cell Biol, 153, 121-35.

Emanuele, M. J., McCleland, M. L., Satinover, D. L., Stukenberg, P. T. (2005). Measuring the stoichiometry and

physical interactions between components elucidates the architecture of the vertebrate kinetochore. Mol. Biol.

Cell 16, 4882–92.

190

Page 215: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Emerald, B. S., Curtiss, J., Mlodzik, M., Cohen, S. M. (2003). Distal antenna and distal antenna related encode

nuclear proteins containing pipsqueak motifs involved in antenna development in Drosophila. Development.

130, 1171-80.

Erkner, A., Gallet, A., Angelats, C., Fasano, L., Kerridge, S. (1999). The role of Teashirt in proximal leg

development in Drosophila: ectopic Teashirt expression reveals different cell behaviours in ventral and dorsal

domains. Dev Biol 215, 221-32.

Erkner, A., Roure, A., Charroux, B., Delaage, M., Holway, N., Core, N., Vola, C., Angelats, C., Pages, F.,

Fasano, L. , Kerridge, S. (2002). Grunge, related to human Atrophin-like proteins, has multiple functions in

Drosophila development. Development 129, 1119-29.

Evans, T., Rosenthal, E. T., Youngblom, J., Distel, D., T. Hunt (1983). Cyclin: a protein specified by maternal

mRNA in sea urchin eggs that is destroyed at each cleavage division. Cell 33, 389-96.

Fan, H., Oro, A. E., Scott, M. P., Khavari, P. A. (1997). Induction of basal cell carcinoma features in transgenic

human skin expressing Sonic Hedgehog..Nat. Med. 3, 788–92.

Fang, G., Yu, H., Kirschner, M. W. (1998). The checkpoint protein MAD2 and the mitotic regulator CDC20 form a

ternary complex with the anaphase-promoting complex to control anaphase initiation. Genes Dev. 12, 1871–

83.

Fang, G., Yu, H., Kirschner, M. W. (1999). Control of mitotic transition by the anaphase-promoting complex. Philos

Trans R Soc Lond B Biol Sci 354, 1583-90.

Fasano, L., Kerridge, S., (1988). Monitoring positional information during oogenesis in adult Drosophila.

Development 104, 245-53.

Fasano, L., Roder, L., Core, N., Alexandre, E., Vola, C., Jacq, B., Kerridge, S. (1991). The gene teashirt is

required for the development of Drosophila embryonic trunk segments and encodes a protein with widely

spaced zinc finger motifs. Cell 64, 63-79.

Fesquet, D., Labbé, J. C., Derancourt, J., Capony, J. P., Galas, S., Girard, F., Lorca, T., Shuttleworth, J.,

Dorée, M., Cavadore, J. C. (1993). The MO15 gene encodes the catalytic subunit of a protein kinase that

activates cdc2 and other cyclin-dependent kinases (CDKs) through phosphorylation of Thr161 and its

homologues. EMBO J 12, 3111–21.

Firth, L. C., Baker, N. E., (2005). Extracellular signals responsible for spatially regulated proliferation in the

differentiating Drosophila eye. Dev Cell. 8, 541-551. Erratum in: Dev Cell. 8, 972.

Flemming, W. (1965) Contributions to the knowledge of cell and its vital processes. Part II. J Cell Biol, 25, 3-69.

Forbes, A. J., Lin, H., Ingham, P. W., Spradling, A. C. (1996a). Hedgehog is required for the proliferation and

specification of ovarian somatic cells prior to egg chamber formation in Drosophila. Development 122, 1125-

35.

Forbes, A. J., Spradling, A. C., Ingham, P. W., Lin, H. (1996b). The role of segment polarity genes during early

oogenesis in Drosophila. Development 122, 3283-94.

Forbes, A., Lehmann, R. (1998). Nanos and Pumilio have critical roles in the development and function of Drosophila

germline stem cells. Development 125, 679–90.

Förstemann, K., Tomari, Y., Du, T., Vagin, V. V., Denli, A. M., Bratu, D. P., Klattenhoff, C., Theurkauf, W. E.,

Zamore, P. D. (2005). Normal microRNA maturation and germ-line stem cell maintenance requires

Loquacious, a double-stranded RNA-binding domain protein. PLoS Biol 3, 1187-201.

Fraser, S., Keynes, R., Lumsden, A. (1990). Segmentation in the chick embryo hindbrain is defined by cell lineage

restrictions. Nature 344, 431–5.

Fuller, M. T., Spradling, A. C. (2007). Male and Female Drosophila Germline Stem Cells: Two Versions of

Immortality. Science 316, 402-4.

Furnari, B., Blasina, A., Boddy, M. N., McGowan, C. H., Russell, P. (1999). Cdc25 inhibited in vivo and in vitro by

checkpoint kinases Cds1 and Chk1. Mol Biol Cell 10, 833-45.

Furnari, B., Rhind, N., Russel, P. (1997). Cdc25 mitotic inducer target by Chk1 DNA damage checkpoint kinase.

Science 277, 1495-7.

191

Page 216: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Furuno, N., Elzen, N., Pines, J. (1999) Human cyclin A is required for mitosis until mid prophase. J Cell Biol. 147,

295-306.

Gabrielli, B. G., De Souza C. P., Tonks, I. D., Clark, J. M., Hayward, N. K., Ellem, K. A. (1996). Cytoplasmic

accumulation of cdc25B phosphatase in mitosis triggers centrossmal microtubule nucleation in HeLa cells.

JCell Sci. 109, 1081-93.

Gaglio, T., Saredi, A., Bingham, J. B., Hasbani, M. J., Gill, S. R., Schroer, T. A., Compton, D. A. (1996).

Opposing motor activities are required for the organization of the mammalian mitotic spindle pole. J. Cell Biol.

135, 399–414.

Gailani, M. R., Stahle-Bäckdahl, M., Leffell, D. J., Glynn, M., Zaphiropoulos, P. G., Pressman ,C., Undén, A.

B., Dean, M., Brash, D.E., Bale, A. E., Toftgard, R. (1996). The role of the human homologue of

Drosophila patched in sporadic basal cell carcinomas. Nat Genet. 1, 78-81.

Gallet, A., Angelats, C., Erkner, A., Charroux, B., Fasano, L., Kerridge, S. (1999). The C-terminal domain of

armadillo binds to hypophosphorylated teashirt to modulate wingless signalling in Drosophila. Embo J. 18,

2208-17.

Gallet, A., Angelats, C., Kerridge, S., Therond, P. P. (2000). Cubitus interruptus-independent transduction of the

Hedgehog signal in Drosophila. Development 127, 5509-22.

Gallet, A., Erkner, A., Charroux, B., Fasano, L., Kerridge, S. (1998). Trunk-specific modulation of wingless

signalling in Drosophila by teashirt binding to armadillo. Curr Biol. 8, 893-902.

Gateff E., Kurzik-Dumke U., Wismar J., Löffler T., Habtemichael N., Konrad L., Dreschers S., Kaiser S.,

Protin U. (1996). Drosophila differentiation genes instrumental in tumor suppression. Int. J Dev Biol. 40,

149–56.

Gehring, W. J. (2002). The genetic control of eye development and its implications for the evolution of the various

eye-types. Int. J. Dev. Biol. 46, 65-73.

Geley, S., Kramer, E., Gieffers, C., Gannon, J., Peters, J. M., Hunt, T. (2001). Anaphase-promoting

complex/cyclosome-dependent proteolysis of human cyclin A starts at the beginning of mitosis and is not

subject to the spindle assembly checkpoint. J. Cell Biol. 153, 137–148.

Gibson, G., Gehring, A. W. J., (1988). Head and thoracic transformations caused by ectopic expression of

antennapedia during Drosophila development. Development 102, 657–75.

Gibson, G., Schier, A., LeMotte, P., Gehring, W. J. (1990). The specificities of Sex combs reduced and

Antennapedia are defined by a distinct portion of each protein that includes the homeodomain. Cell 62, 1087-

103.

Gilboa, L., Forbes, A., Tazuke, S. I., Fuller, M. T., Lehmann, R. (2003). Germ line stem cell differentiation in

Drosophila requires gap junctions and proceeds via an intermediate state. Development 130, 6625–34.

Gilboa, L., Lehmann, R. (2004). How different is Venus from Mars? The genetics of germline stem cells in Drosophila

females and males. Development 131, 4895–905.

Girard, F., Fernandez, A., N. Lamb (1995) Delayed Cyclin A and B1 degradation in non-transformed mammalian

cels. J Cell Sci, 108, 2599-5.

Glotzer, M., Murray, A. W., Kirschner, M. W. (1991). Cyclin is degraded by the ubiquitin pathway. Nature 349,

132-8.

Glover, D. M., Hagan, I. M., Tavares, A. A. (1998) Polo-like kinases: a team that plays throughout mitosis. Genes

Dev. 12, 3777-87.

Godt, D., Laski, F. A. (1995). Mechanisms of cell rearrangement and cell recruitment in Drosophila ovary

morphogenesis and the requirement of bric à brac. Development. 121, 173-87.

Gonzalez, C., Glover, D. M. (1993). Techniques for studying mitosis in Drosophila. In The Cell Cycle: A Practical

Approach., (ed. P. Fantes and R. Brooks), pp. 145-175. Oxford: IRL Press.

Gonzalez-Crespo, S., Morata, G., (1996). Genetic subdivision of the arthropod limb into coxopodite and telopodite.

Development 122, 3921–28.

192

Page 217: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Gonzalez-Gaitan, M., Rothe, M., Wimmer, E. A., Taubert, H., Jackle, H. (1994). Redundant functions of the

genes knirps and knirps-related for the establishment of anterior Drosophila head structures. Proc Natl Acad

Sci USA 91, 8567-71.

Gonzalez-Reyes, A. (2003) Stem Cells, niche and Cadherins: a view from Drosophila. J Cell Science 116, 949-54.

Gorbsky, G. J., Ricketts, W. A. (1993). Differential expression of a phosphoepitope at the kinetochores of moving

chromosomes. J. Cell Biol. 122, 1311–21.

Gordon, M. B., Howard, L., Compton, D. A. (2001). Chromosome movement in mitosis requires microtubule

anchorage at spindle poles. J. Cell Biol. 152, 425–34.

Goto, H., Kiyono, T., Tomono, Y., Kawajiri, A., Urano, T., Furukawa, K., Nigg, E. A., Inagaki, M. (2006).

Complex formation of Plk1 and INCENP required for metaphase-anaphase transition. Nat. Cell Biol. 8, 180–7.

Graba, Y., Laurenti, P., Perrin, L., Aragnol, D., Pradel, J., (1994). The modifier of variegation modulo gene acts

downstream of dorsoventral and HOM-C genes and is required for morphogenesis in Drosophila. Dev Biol

166, 704–15.

Grachtchouk, M., Mo, R., Yu, S., Zhang, X., Sasaki, H., Hui , C. C., Dlugosz, A. A. (2000). Basal cell carcinomas

in mice overexpressing Gli2 in skin. Nat Genet. 24, 216-7.

Greenwood, S., Struhl, G. (1999). Progression of the morphogenetic furrow in the Drosophila eye: The roles of

Hedgehog, Decapentaplegic and the Raf pathway. Development 126, 5795–5808.

Gritzan, U., Hatini, V., DiNardo, S. (1999). Mutual antagonism between signals secreted by adjacent wingless and

engrailed cells leads to specification of complementary regions of the Drosophila parasegment. Development

126, 4107-15.

Grivna S. T., Beyret E., Wang Z., Lin H. (2006a). A novel class of small RNAs in mouse spermatogenic cells. Genes

Dev 20,1709-14.

Grivna S. T., Pyhtila B., Lin H. (2006b). MIWI associates with translational machinery and PIWI-interacting RNAs

(piRNAs) in regulating spermatogenesis. Proc Natl Acad Sci USA 103, 13415-20.

Grossniklaus, U., Pearson, R. K., Gehring, W. J. (1992). The Drosophila sloppy paired locus encodes two proteins

involved in segmentation that show homology to mammalian transcription factors. Genes Dev 6, 1030-51.

Gupta, A., Tsai, L. H., Wynshaw-Boris, A. (2002). Life is a journey: a genetic look at neocortical development. Nat

Rev Genet 3, 342-55.

Gustavson, E., Goldsborough, A. S., Ali, Z., Kornberg, T. B. (1996). The Drosophila engrailed and invected genes:

partners in regulation, expression and function. Genetics 142, 893-906.

Haerry, T. E., Khalsa, O., O’Connor, M. B., Wharton, K. A. (1998). Synergistic signalling by two BMP ligands

through the SAX and TKV receptors controls wing growth and patterning in Drosophila. Development 125,

3977–87.

Hagting, A., Den Elzen, N., Vodermaier, H. C., Waizenegger, I. C., Peters, J. M., Pines, J. (2002). Human

securin proteolysis is controlled by the spindle checkpoint and reveals when the APC/C switches from

activation by Cdc20 to Cdh1. J. Cell Biol. 157, 1125–37.

Hagting, A., Karlsson, C., Clute, P., Jackman, M., Pines, J. (1998) MPF localization is controlled by nuclear export.

EMBO J, 17, 4127-38.

Halder, G., Callaerts, P., Gehring, W. J. (1995). Induction of ectopic eyes by targeted expression of the eyeless

gene in Drosophila. Science 267, 1788-92.

Harlow, E., Lane, D. (1988). Antibodies : a laboratory manual. Cold Spring Harbor, NY: Cold Spring Harbor

Laboratory

Hashimoto, N., Kishimoto, T. (1988). Regulation of meiotic metaphase by a cytoplasmic maturation-promoting

factor during mouse oocyte maturation. Dev Biol. 126, 242-52.

Hatfield S. D., Shcherbata H. R., Fischer K. A., Nakahara K., Carthew R. W., Ruohola-Baker H. (2005). Stem

cell division is regulated by the microRNA pathway. Nature 435, 974-78.

193

Page 218: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Hauf, S., Cole, R. W., LaTerra, S., Zimmer, C., Schnapp, G., Walter, R., Heckel, A., van Meel, J., Rieder, C. L.,

Peters, J. M. (2003). The small molecule Hesperadin reveals a role for Aurora B in correcting kinetochore–

microtubule attachment and in maintaining the spindle assembly checkpoint. J. Cell Biol. 161, 281–94.

Hayes, M. J., Kimata, Y., Wattam, S. L., Lindon, C., Mao, G., Yamano, H., Fry, A. M. (2006). Early mitotic

degradation of Nek2A depends on Cdc20-independent interaction with the APC/C. Nat. Cell Biol. 8, 607–14.

He, X. C., Zhang, J., Tong, W. G., Tawfik, O., Ross, J., Scoville, D. H., Tian, Q., Zeng, X., He, X., Wiedemann,

L. M., Mishina Y. Li L. (2004). BMP signalling inhibits intestinal stem cell self-renewal through suppression of

Wnt-β-catenin signaling. Nat. Genet. 36, 1117–21.

He, X., Rines, D. R., Espelin, C. W., Sorger, P. K. (2001). Molecular analysis of kinetochore-microtubule

attachment in budding yeast. Cell 106, 195–206.

Heald, R., McLoughlin, M., McKeon, F. (1993) Human Wee1 maintains mitotic timing by protecting the nucleus

from cytoplasmically activated Cdc2 Kinase. Cell 74, 463-74.

Heald, R., Tournebize, R., Habermann, A., Karsenti, E., Hyman, A. (1997). Spindle assembly in Xenopus egg

extracts: respective roles of centrosomes and microtubule self-organization. J. Cell Biol. 138, 615–28.

Heberlein, U., Singh, C. M., Luk, A. Y. Donohoe, T. J. (1995). Growth and differentiation in the Drosophila eye

coordinated by hedgehog. Nature 373, 709–11.

Heberlein, U., Singh, C. M., Luk, A. Y., Donohoe, T. J. (1995). Growth and differentiation in the Drosophila eye

coordinated by hedgehog. Nature 373, 709-11.

Heberlein, U., Wolff, T., Rubin, G. M. (1993). The TGF beta homolog dpp and the segment polarity gene hedgehog

are required for propagation of a morphogenetic wave in the Drosophila retina. Cell 75, 913-26.

Heemskerk, J., DiNardo, S., Kostriken, R., O'Farrell, P. H. (1991). Multiple modes of engrailed regulation in the

progression towards cell fate determination. Nature 352, 404-10.

Held, L. I. J., Heup, M. A., Sappington, J. M., Peters, S. D. (1994).Interactions of decapentaplegic, wingless and

Distal-less in the Drosophila leg. Roux’ Arch. Dev. Biol. 203, 310-19.

Hepker, J., Wang, Q. T., Motzny, C. K., Holmgren, R., Orenic, T. V. (1997). Drosophila cubitus interruptus forms

a negative feedback loop with patched and regulates expression of Hedgehog target genes. Development

124, 549-58.

Herke, S. W., Serio, N. V., Rogers, B. T. (2005). Functional analyses of tiptop and Antennapedia in the embryonic

development of Oncopeltus fasciatus suggests an evolutionary pathway from ground state to insect legs.

Development 132, 27-34.

Hershko, A. (1999). Mechanisms and regulation of the degradation of the cyclin B. Philos Trans R Soc Lond B Biol Sci

354, 1571-6.

Hidalgo, A., Ingham, P. (1990). Cell patterning in the Drosophila segment: spatial regulation of the segment polarity

gene patched. Development 110, 291-301.

Hoodless, P. A., Wrana, J. L. (1998). Mechanism and function of signaling by the TGF beta superfamily. Curr. Top.

Microbiol. Immunol. 228, 235-72.

Hooper, J. E., Scott, A. M. P. (1989). The drosophila patched gene encodes a putative membrane protein required

for segmental patterning. Cell 59, 751-65.

Howell, B. J., McEwen, B. F., Canman, J. C., Hoffman, D. B., Farrar, E. M., Rieder, C. L., Salmon, E. D. (2001).

Cytoplasmic dynein/dynactin drives kinetochore protein transport to the spindle poles and has a role in

mitotic spindle checkpoint inactivation. J. Cell Biol. 155, 1159–72.

Howell, B. J., Moree, B., Farrar, E. M., Stewart, S., Fang, G., Salmon, E. D. (2004). Spindle checkpoint protein

dynamics at kinetochores in living cells. Curr. Biol. 14, 953–64.

Hoyt, M. A. (2001) A new view of the spindle checkpoint. J Cell Biol. 154, 909-11.

Hoyt, M. A., Totis, L., Roberts, B. T. (1991). S. cerevisiae genes required for cell cycle arrest in response to loss of

microtubule function. Cell 66, 507–17.

194

Page 219: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Hu, M. C., Mo, R., Bhella, S., Wilson, C. W., Chuang, P. T., Hui, C. C., Rosenblum, N. D. (2006) GLI3-dependent

transcriptional repression of Gli1, Gli2 and kidney patterning genes disrupts renal morphogenesis.

Development 133, 569-78.

Huang, J. Y., Raff, J. W. (2002). The dynamic localisation of the Drosophila APC/C: evidence for the existence of

multiple complexes that perform distinct functions and are differentially localised. J. Cell Sci. 115, 2847-56.

Huang, J., J. Raff (1999) The disappearance of cyclin B at the end of mitosis is regulated spatially in Drosophila cells.

EMBO J. 18, 2184-95.

Huber, O., Korn, R., McLaughlin, J., Ohsugi, M., Herrmenn, B., Kemler, R. (1996). Nuclear localization of b-

catenin by interaction with transcription factor LEF-1. Mech. Dev. 59, 3-10.

Hunt, T. (1991) Cyclins and their partners: from a single idea to a complicated reality. Semin. Cell Biol. 2, 213-22.

Hwang, L. H., Lau, L. F., Smith, D. L., Mistrot, C. A., Hardwick, K. G., Hwang, E. S., Amon, A., Murray, A. W.

(1998). Budding yeast Cdc20: a target of the spindle checkpoint. Science 279, 1041–4.

Hyman, A. A., Mitchison, T. J. (1991). Two different microtubule based motor activities with opposite polarities in

kinetochores. Nature 351, 206–11.

Iliakis, G., Wang, Y., Guan, J. Wang, H. (2003). DNA damage checkpoint control in cells exposed to ionizing

radiation. Oncogene 22, 5834-47.

Immerglück, K., P. A. Lawrence M. Bienz (1990). Induction across germ layers in Drosophila mediated by a

genetic cascade. Cell 62, 261-8.

Ingham, P. (1998a). The patched gene in development and cancer. Curr Opin. Genet. Dev. 8, 88-94.

Ingham, P. W. (1993). Localized hedgehog activity controls spatial limits of wingless transcription in the Drosophila

embryo. Nature 366, 560-2.

Ingham, P. W. (1998b). Transducing Hedgehog: the story so far. EMBO J. 17, 3505-11.

Ingham, P. W., Feitz, M. (1995). Quantitative effects of hedgehog and decapentaplegic activity on the patterning of

the Drosophila wing. Curr.Biol. 5, 432-40.

Ingham, P. Wand Hidalgo, A. (1993). Regulation of wingless transcription in the Drosophila embryo. Development

117, 283–91.

Izumi, T., Walker, D. H., Maller, J. L. (1992). Periodic changes in phosphorylation of the Xenopus cdc25

phosphatase regulate its activity. Mol. Biol. Cell 3, 927-39.

Jackman, M., Firth, M., Pines, J. (1995) Human cyclin B1 and B2 are localized to strikingly different structures: B1

to microtubules, B2 primarily to the Golgi apparatus. EMBO J. 14, 1646-54.

Jacob, L., Lum, L. (2007) Deconstructing the hedgehog pathway in development and disease. Science. 2007 Oct

5;318(5847):66-8.

Jacobs, H. W., Keidel, E. and Lehner, C. F. (2001). A complex degradation signal in Cyclin A required for G1 arrest,

and a C-terminal region for mitosis. Embo J 20, 2376-86.

Jacobs, H. W., Knoblich, J. A., Lehner, C. F. (1998) Drosophila Cyclin B3 is required for female fertility and is

dispensable for mitosis like Cyclin B. Genes Dev. 12, 3741-51.

Jan, Y. N., Jan L. Y. (2001). Asymmetric cell division in the Drosophila nervous system. Nat. Rev. Neurosci. 2, 772-9.

Jeannotte, L., Lemieux, M., Charron, J., Poirier, F., Robertson, E. J., (1993). Specification of axial identity in the

mouse: role of the Hoxa-5 (Hox1.3) gene. Genes Dev. 7, 2085–96.

Jiang F., Ye X., Liu X., Fincher L., McKearin D., Liu Q. (2005). Dicer-1 and R3D1-L catalyze microRNA maturation

in Drosophila. Genes Dev 19, 1674-79.

Jiang, J., Hui, C. C. (2008). Hedgehog signaling in development and cancer. Dev Cell 15, 801-2.

Johnson, D. G., Walker C. L. (1999) Cyclins and cell cycle checkpoints. Annu. Rev. Pharmacol. Toxico.l 39, 295-312.

Johnson, R. L., Rothman, A. L., Xie, J., Goodrich, L. V., Bare, J. W., Bonifas, J. M., Quinn, A. G., Myers, R. M.,

Cox, D. R., Epstein, E. H., Jr., Scott, M. P. (1996). Human homolog of patched, a candidate gene for the

basal cell nevus syndrome. Science 272, 1668–71.

Johnson, V. L., Scott, M. I., Holt, S. V., Hussein, D. Taylor, S. S. (2004). Bub1 is required for kinetochore

localization of BubR1, Cenp-E, Cenp-F and Mad2, and chromosome congression. J. Cell Sci. 117, 1577–89.

195

Page 220: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Jordan, M. A., Wilson, L. (2004). Microtubules as a target for anticancer drugs. Nat. Rev. Cancer 4, 253–65.

Jürgens, G. (1988). Head and tail development of the Drosophila embryo involves spalt, a novel homeotic gene.

EMBO J. 7, 189-96.

Jürgens, G., Weigel, D. (1988). Terminal versus segmental development in the Drosophila embryo: the role of the

homeotic gene fork head. Roux’s Arch. Dev. Biol. 197, 345-54.

Kai, T., Spradling, A. (2003). An empty Drosophila stem cell niche reactivates the proliferation of ectopic cells. Pro

Natl Acad Sci USA 100, 4633–38.

Kai, T., Spradling, A. (2004). Differentiating germ cells can revert into functional stem cells in Drosophila

melanogaster ovaries. Nature 428, 564–9.

Kai, T., Wiliams, D., Spradling A. C. (2005). The expression profile of purified Drosophila germline stem cells. Dev

Biol. 283, 486-502.

Kajiwara, Y., Akram, A., Katsel, P., Haroutunian, V., Schmeidler, J., Beecham, G., Haines, J. L., Pericak-

Vance, M. A., Buxbaum, J. D. (2009). FE65 binds Teashirt, inhibiting expression of the primate-specific

caspase-4. PLoS One. 4, e5071.

Kallio, M., Weinstein, J., Daum, J. R., Burke, D. J., Gorbsky, G. J. (1998). Mammalian p55CDC mediates

association of the spindle checkpoint protein Mad2 with the cyclosome/anaphase-promoting complex, and is

involved in regulating anaphase onset and late mitotic events. J Cell Biol 141, 1393-406.

Kapoor, T. M., Compton, D. A. (2002). Searching for the middle ground:mechanisms of chromosome alignment

during mitosis. J. Cell Biol. 157, 551-6.

Karess, R. (2005). Rod-Zw10-Zwilch: a key player in the spindle checkpoint.Trends Cell Biol. 15, 386-92.

Karess, R. E., Glover, D. M. (1989). rough deal: a gene required for proper mitotic segregation in Drosophila. J Cell

Biol 109, 2951-61.

Karki, S., Holzbaur, E. L. (1999). Cytoplasmic dynein and dynactin in celldivision and intracellular transport. Curr.

Opin. Cell Biol. 11, 45-53.

Kastan, M. B., Bartek, J. (2004). Cell-cycle checkpoints and cancer. Nature 432, 316-23.

Kawase, E.,Wong, M. D., Ding, B.C., Xie, T. (2004). Gbb/Bmp signalling is essential for maintaining germline stem

cells and for repressing bam transcription in the Drosophila testis. Development 131, 1365–75.

Kenney, A. M., Rowitch, D. H. (2000) Sonic hedgehog promotes G(1) cyclin expression and sustained cell cycle

progression in mammalian neuronal precursors. Mol Cell Biol. 20, 9055-67.

Kenney, A. M., Widlund, H. R., Rowitch, D. H. (2004) Hedgehog and PI-3 kinase signaling converge on Nmyc1 to

promote cell cycle progression in cerebellar neuronal precursors. Development 131, 217-28. Erratum in:

Development. 2004 131, 489.

Kenyon, K. L., Ranade, S. S., Curtiss, J., Mlodzik, M., Pignoni, F. (2003). Coordinating proliferation and tissue

specification to promote regional identity in the Drosophila head. Dev. Cell 5, 403-14.

Keppler-Noreuil, K. M., Carroll, A. J., Finley, S. C., Descartes, M., Cody, J. D., DuPont, B. R., Gay, C. T.,

Leach, R. J. (1998). Chromosome 18q paracentric inversion in a family with mental retardation and hearing

loss. Am. J. Med. Genet. 76, 372–8.

Khalsa, O., Yoon, J. W., Torres-Schumann, S., Wharton, K. A. (1998). TGF-beta/BMP superfamily members, Gbb-

60A and Dpp, cooperate to provide pattern information and establish cell identity in the Drosophila wing.

Development 125, 2723–34.

Kiecker C, Lumsden A. (2005). Compartments and their boundaries in vertebrate brain development. Nature

Reviews Neuroscience 6, 553–64.

Kim, S. H., Lin, D. P., Matsumoto, S., Kitazono, A., Matsumoto, T. (1998). Fission yeast Slp1: an effector of the

Mad2-dependent spindle checkpoint. Science 279, 1045–7.

King, F. J., Lin, H. (1999). Somatic signaling mediated by fs(1)Yb is essential for germline stem cell maintenance

during Drosophila oogenesis. Development 126, 1833–44.

King, F. J., Szakmary, A., Cox, D. N., Lin, H. (2001). Yb modulates the divisions of both germline and somatic stem

cells through piwiand hh-mediated mechanisms in the Drosophila ovary. Mol. Cell 7, 497–508.

196

Page 221: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

King, J. M., Hays, T. S., Nicklas, R. B. (2000). Dynein is a transient kinetochore component whose binding is

regulated by microtubule attachment, not tension. J Cell Biol 151, 739-48.

King, R. C. (1970). Ovarian development in Drosophila melanogaster. Academic Press: New York.

King, R. W., Deshaies, R. J., Peters, J. M., Kirshner, M. W. (1996). How proteolysis drives the cell cycle. Science

274, 1652- 9.

King, R. W., Peters, J. M., Tugendreich, S., Rolfe, M., Hieter, P., Kirschner, M. W. (1995). A 20S complex

containing CDC27 and CDC16 catalyzes the mitosis-specific conjugation of ubiquitin to cyclin B. Cell 81, 279-

88.

Kirilly, D., Spana, E. P., Perrimon, N., Padgett, R. W., Xie, T. (2005) BMP signalling is required for controlling

somatic stem cell self-renewal in the Drosophila ovary. Dev. Cell 9, 651-62.

Kirilly, D., Xie, T., (2007). The Drosophila ovary: an active stem cell community. Cell Res. 17, 15-25.

Kishimoto, T., Kanatani, H. (1976). Cytoplasmatic factor responsible for germinal vesicle breakdown and meiotic

maturation in starfish oocyte. Nature 260, 321-2.

Kline, A. D., White, M. E., Wapner, R., Rojas, K., Biesecker, L. G., Kamholz, J., Zackai, E. H., Muenke, M.,

Scott Jr., C. I., Overhauser, J. (1993). Molecular analysis of the 18q-syndrome—and correlation with

phenotype. Am. J. Hum. Genet. 52, 895–906.

Kline, S. L., Cheeseman, I. M., Hori, T., Fukagawa, T., Desai, A. (2006). The human Mis12 complex is required

for kinetochore assembly and proper chromosome segregation. J. Cell Biol. 173, 9–17.

Klingensmith, J., Nusse, R. (1994). Signaling by wingless in Drosophila. Dev Biol 166, 396-414.

Koebernick, K., Kashef, J., Pieler, T., Wedlich, D. (2006). Xenopus Teashirt1 regulates posterior identity in brain

and cranial neural crest. Developmental Bio. 298, 312–26.

Kops, G. J., Weaver, B. A., Cleveland, D. W. (2005). On the road to cancer: aneuploidy and the mitotic checkpoint.

Nat. Rev. Cancer 5, 773–85.

Korinek, V., Barker, N., Moerer, P., van Donselaar, E., Huls, G., Peters, P.J., Clevers, H. (1998). Depletion of

epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat. Genet. 19, 379–83.

Kotaja, N., Lin, H., Parvinen, M., Sassone-Corsi, P. (2006). Interplay of PIWI/ Argonaute protein MIWI and kinesin

KIF17b in chromatoid bodies of male germ cells. J. Cell Sci. 119, 2819-25.

Kotani, S., Tugendreich, S., Fujii, M., Jorgensen, P. M., Watanabe, N., Hoog, C., Hieter, P. and Todokoro, K.

(1998). PKA and MPF-activated polo-like kinase regulate anaphase-promoting complex activity and mitosis

progression. Mol Cell 1, 371-80.

Kotani, S., Tugendreich, S., Fujii, M., Jorgensen, P. M., Watanabe, N., Hoog, C., Hieter, P., Todokoro, K.

(1998). PKA and MPF-activated polo-like kinase regulate anaphase-promoting complex activity and mitosis

progression. Mol Cell 1, 371-80.

Koumenis, C., Giaccia, A. (1997). Transformed cells require continuous activity of RNA polymerase II to resist

oncogene-induced apoptosis. Mol. Cell Biol. 17, 7306–16.

Kühl, M., Wedlich, D. (1997). Wnt signalling goes nuclear. Bioessays 19, 101-4.

Kumar, J. P., Moses, K. (2001). EGF receptor and Notch signaling act upstream of Eyeless/Pax6 to control eye

specification. Cell 104, 687-97.

Labbe, J. C., Lee, M. G., Nurse, P., Picard, A., Doree, M. (1988). Activation at M-phase of a protein kinase

encoded by a starfish homologue of the cell cycle control gene cdc2+. Nature 335, 251-4.

LaFever, L., Drummond-Barbosa, D. (2005). Direct control of germline stem cell division and cyst growth by neural

insulin in Drosophila. Science 309, 1071-73.

Lampson, M. A., Renduchitala, K., Khodjakov, A., Kapoor, T. M. (2004). Correcting improper chromosomespindle

attachments during cell division. Nature Cell Biol. 6, 232–7.

Lantz, V., Chang, J. S., Horabin, J. I., Bopp, D., Schedl, P. (1994). The Drosophila orb RNA-binding protein is

required for the formation of the egg chamber and establishment of polarity. Genes Dev. 8, 598–613.

197

Page 222: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Laugier, E. (2005). Etude fonctionnelle du gene tiptop, un paralogue de teashirt chez Drosophila melanogaster. PhD

thesis. l’Université de la Méditerranée.

Laugier, E., Yang, Z., Fasano, L., Kerridge, L., Vola, C. (2005). A critical role of teashirt for patterning the ventral

epidermis is masked by ectopic expression of tiptop, a paralog of teashirt in Drosophila. Dev Biol 283, 446-

58.

Lavoie, C. A., Ohlstein, B., McKearin, D. M. (1999). Localization and function of Bam protein require the benign

gonial cell neoplasm gene product. Dev. Biol. 212, 405–13.

Lecuit, T., Cohen, S. M. (1997). Proximal-distal axis formation in the Drosophila leg. Nature 388, 139-45.

Lehner, C. F., O’Farrell, P. H. (1989) Expression and function of Drosophila cyclin A during embryonic cell cycle

progression. Cell 56, 957-68.

Lehner, C. F., O’Farrell, P. H. (1990) The role of Drosophila Cyclins A and B in mitotic control. Cell 61, 535-47.

Leismann, O., Herzig, A., Heidmann, S. Lehner, C. F. (2000). Degradation of Drosophila PIM regulates sister

chromatid separation during mitosis. Genes Dev 14, 2192-205.

Lepage, T., Cohen, S. M., Diaz-Benjumea, F. J., Parkhurst, S. M. (1995) Signal transduction by cAMP-dependent

protein kinase A in Drosophila limb patterning. Nature 373, 711-15.

Levine, K., Kiang, L., Jacobso, M., Fisher, R. Cross, F. (1999). Directed evolution to bypass cyclin requirements

for the cdc28p cyclin-dependent Kinase. Molecular cell 4, 353-63.

Li, C., Wong, W. H. (2001a). Model-based analysis of oligonucleotide arrays: model validation, design issues and

standard error application. Genome Biol 2, RESEARCH0032.

Li, C., Wong, W. H. (2001b). Model-based analysis of oligonucleotide arrays: expression index computation and

outlier detection. Proc Natl Acad Sci U S A 98, 31-6.

Li, J., Meyer, A. N. Donoghue, D. J. (1997) Nuclear localization of cyclin B1 mediates its biological activity and is

regulated by phosphorylation. Proc. Natl. Acad. Sci. USA, 94, 502-7.

Li, L., Xie, T. (2005). Stem cell niche: structure and function. Annu. Rev. Cell Dev. Biol. 21, 605-31.

Li, R., Murray, A. (1991) Feedback control of mitosis in budding yeast. Cell 66, 519–31.

Li, W., Ohlmeyer, J. T., Lane, M. E., Kalderon, D. (1995) Function of protein kinase A in hedgehog signal

transduction and Drosophila imaginaldisc development. Cell 80, 553-62.

Li, X., Nicklas, R. B. (1995). Mitotic forces control a cell-cycle checkpoint. Nature 373, 630-2.

Li, X., Nicklas, R. B. (1997). Tension-sensitive kinetochore phosphorylation and the chromosome distribution

checkpoint in praying mantid spermatocytes. J. Cell Sci. 110, 537-45.

Lilly, M. A., Spradling, A. C. (1996). The Drosophila endocycle is controlled by Cyclin E and lacks a checkpoint

ensuring S-phase completion. Genes Dev. 10, 2514–26.

Lin H., Spradling, A. C. (1993). Germline stem cell division and egg chamber development in transplanted

Drosophila germaria. Dev. Biol. 159, 140-52.

Lin, H. (2002). The stem-cell niche theory: lessons from flies. Nat. Rev. Genet. 3, 931–40.

Lin, H., Spradling, A. (1997). A novel group of pumilio mutations affects the asymmetric division of germline stem

cells in the Drosophila ovary. Development 124, 2463-76.

Lin, H., Yue, L., Spradling, A. C. (1994). The Drosophila fusome, a germline-specific organelle, contains membrane

skeletal proteins and functions in cyst formation. Development 120, 947–56.

Lynch, M., Force, A. (2000). The probability of duplicate gene preservation by subfunctionalization. Genetics 154,

459–473.

Lindsley, D. L., Zimm, G. G. (1992). The genome of Drosophila melanogaster. San Diego: Academic Press.

Liu X., Grammont M., Irvine K. D. (2000) Roles for scalloped and vestigial in regulating cell affinity and interactions

between the wing blade and the wing hinge. Dev. Biol. 228, 287–303.

Liu, S. T., Rattner, J. B., Jablonski, S. A., Yen, T. J. (2006). Mapping the assembly pathways that specify

formation of the trilaminar kinetochore plates in human cells. J. Cell Biol. 175, 41–53.

Liu, Y., Montell, D. J. (1999). Identification of mutations that cause cell migration defects in mosaic clones.

Development 126, 1869-78.

198

Page 223: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Llamazares, S., Moreira, A., Tavares, A., Girdham, C., Spruce, B. A., Gonzalez C., Karess, R. E., Glover, D.

M., Sunkel, C. E. (1991). polo encodes a protein kinase homolog required for mitosis in Drosophila. Genes

Dev 5, 2153-65.

Locker, M., Agathocleous, M., Amato, M. A., Parain, K., Harris, W. A., Perron, M., 2006. Hedgehog signaling

and the retina: insights into the mechanisms controlling the proliferative properties of neural precursors.

Genes Dev. 20, 3036-48.

Lohka, M. J., Hayes, M. K., Maller, J. L. (1988). Purification of maturation-promoting factor, an intracellular

regulator of early mitotic events. Proc. Natl. Acad. Sci. USA 85, 3009-13.

Lopez-Schier, H., St. Johnston, D. (2001) Delta signalling from the germ line controls the proliferation and

differentiation of the somatic follicle cells during Drosophila oogenesis. Genes Dev. 15, 1393-405.

Lou, Y., Yao, J., Zereshki, A., Dou, Z., Ahmed, K., Wang, H., Hu, J., Wang, Y., Yao, X. (2004). NEK2A interacts

with MAD1 and possibly functions as a novel integrator of the spindle checkpoint signaling. J. Biol. Chem.

279, 20049–57.

Lowe, M., Rabouille, C., Nakamura, N., Watson, R., Jackman, M., Jamsa, E., Rahman, D., Pappin, D. J.,

Warren, G. (1998) Cdc2 kinase directly phosphorylates the cis-Golgi matrix protein G130 and is required for

Golgi fragmentation in mitosis. Cell 94, 783-93.

Lum, L., Zhang, C., Oh, S., Mann, R. K., von Kessler, D. P., Taipale, J., Weis-Garcia, F., Gong, R., Wang, B.,

Beachy, P. A. (2003) Hedgehog signal transduction via Smoothened association with a cytoplasmic complex

scaffolded by the atypical kinesin, Costal-2. Mol Cell.12,1261-74.

Mahaffey, J. W., Griswold, C. M., Cao, Q. M. (2001). The Drosophila genes disconnected and disco-related are

redundant with respect to larval head development and accumulation of mRNAs from deformed target genes.

Genetics 157, 225-36.

Maiato, H., DeLuca, J., Salmon, E.D., Earnshaw, W.C. (2004). The dynamic kinetochore-microtubule interface. J.

Cell Sci. 117, 5461–77.

Manfroid, I., Caubit, X., Kerridge, S. Fasano, L. (2004). Three putative murine Teashirt orthologues specify trunk

structures in Drosophila in the same way as the Drosophila teashirt gene. Development 131, 1065-73.

Margolis, J., Spradling, A. (1995). Identification and behavior of epithelial stem cells in the Drosophila ovary.

Development 121, 3797-807.

Margolis, S. S., Perry, J. A., Forester, C. M., Nutt, L. K., Guo, Y., Jardim, M. J., Thomenius, M. J., Freel, C.D.,

Darbandi, R., Ahn, J. H., Arroyo, J. D., Wang, X. F., Shenolikar, S., Nairn, A. C., Dunphy, W.G., Hahn,

W. C., Virshup, D. M., Kornbluth, S. (2006). Role for the PP2A/B56 delta phosphatase in regulating 14-3-3

release from Cdc25 to control mitosis. Cell 127, 759-73.

Martinez-Arias, A., Baker, N. E., Ingham, P. W. (1988). Role of segment polarity genes in the definition and

maintenance of cells states in the Drosophila embryo. Development 103, 157-70.

Masui, Y. and Market, C. (1971). Cytoplasmic control of nuclear behaviour during meiotic maturation of frog

oocytes. J. Exp. Zool. 177, 129-45.

Mathies, L. D., Kerridge, S., Scott, M. P. (1994). Role of the teashirt gene in Drosophila midgut morphogenesis:

secreted proteins mediate the action of homeotic genes. Development 120, 2799-809.

McCleland, M. L., Gardner, R. D., Kallio, M. J., Daum, J. R., Gorbsky, G. J., Burke, D. J., Stukenberg, P. T.

(2003). The highly conserved Ndc80 complex is required for kinetochore assembly, chromosome congression,

and spindle checkpoint activity. Genes Dev. 17, 101–14.

McCleland, M. L., Kallio, M. J., Barrett-Wilt, G. A., Kestner, C. A., Shabanowitz, J., Hunt, D. F., Gorbsky, G.

J., Stukenberg, P. T. (2004). The vertebrate Ndc80 complex contains Spc24 and Spc25 homologs, which are

required to establish and maintain kinetochore-microtubule attachment. Curr. Biol. 14, 131–7.

McCormick, A., Core, N., Kerridge, S., Scott, M. P. (1995). Homeotic response elements are tightly linked to

tissue-specific elements in a transcriptional enhancer of the teashirt gene. Development 121, 2799-812.

McKearin, D. M., Spradling, A. C. (1990). bag-of-marbles: a Drosophila gene required to initiate both male and

female gametogenesis. Genes Dev. 4, 2242–51.

199

Page 224: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

McKearin, D., Ohlstein, B. (1995). A role for the Drosophila bag-of-marbles protein in the differentiation of

cystoblasts from germline stem cells. Development 121, 2937–47.

Megosh H. B., Cox D. N., Campbell C., Lin H. (2006). The role of PIWI and the miRNA machinery in Drosophila

germline determination. Curr. Biol. 16, 1884-94.

Mendenhal, M. D., Hodge, A. E. (1998) Regulation of Cdc28 cyclin-dependent protein kinase activity during the cell

cycle of the yeast Saccharomyces cerevisae. Microbiol. Mol. Biol. Rev. 62, 1191-243.

Meraldi, P., Draviam, V. M., Sorger, P. K. (2004). Timing and checkpoints in the regulation of mitotic progression.

Dev. Cell 7, 45–60.

Meraldi, P., McAinsh, A. D., Rheinbay, E., Sorger, P. K. (2006). Phylogenetic and structural analysis of

centromeric DNA and kinetochore proteins. Genome Biol. 7, R23.

Merdes, A., Ramyar, K., Vechio, J. D., Cleveland, D.W. (1996). A complex of NuMA and cytoplasmic dynein is

essential for mitotic spindle assembly. Cell 87, 447–58.

Méthot, N., Basler, K. (1999). Hedgehog controls limb development by regulating the activities of distinct

transcriptional activator and repressor forms of Cubitus interruptus. Cell 96, 819–31.

Méthot, N., Basler, K. (2001). An absolute requirement for Cubitus interruptus in Hedgehog signaling. Development

128, 733–42.

Millband, D. N., Hardwick, K. G. (2002). Fission yeast Mad3p is required for Mad2p to inhibit the anaphase-

promoting complex and localizes to kinetochores in a Bub1p-, Bub3p-, and Mph1p-dependent manner. Mol.

Cell. Biol. 22, 2728–42.

Miller, J. R., Moon, R. T. (1996). Signal transduction through b-catenin and specification of cell fate during

embryogenesis. Genes Dev. 10, 2527-39.

Milyavsky, M., Shats, I., Cholostoy, A., Brosh, R., Buganim, Y., Weisz, L., Kogan, I., Cohen, M., Shatz, M.,

Madar, S., Kalo, E., Goldfinger, N., Yuan, J., Ron, S., MacKenzie, K., Eden, A., Rotter, V. (2007).

Inactivation of myocardin and p16 during malignant transformation contributes to a differentiation defect.

Cancer Cell. 11, 133-46.

Mitchison, T. J., Kirschner, M. W. (1984). Dynamic instability of microtubule growth. Nature 312, 237-41.

Mohler, J., Mahaffey, J. W., Deutsch, E., Vani, K. (1995). Control of Drosophila head segment identity by the bZIP

homeotic gene cnc. Development 121, 237-47.

Montagne, J., Stewart, M. J., Stocker, H., Hafen, E., Kozma, S. C., Thomas, G. (1999). Drosophila S6 kinase: a

regulator of cell size. Science 285, 2126–9.

Moore, J. D., Yang, J., Truant, R., Kornbluth, S. (1999) Nuclear import of Cdk/cyclin complexes: Identification of

distinct mechanisms for import of Cdk2/ cyclinE and Cdc2/ cyclin B1. J. Cell Biol. 144, 213-24.

Morgan, D. O. (1999). Regulation of the APC and the exit from mitosis. Nat Cell Biol 1, E47-53.

Moritz, M., Braunfeld, M. B., Guenebaut, V., Heuser, J., Agard, D.A. (2000). Structure of the gamma-tubulin ring

complex: a template for microtubule nucleation. Nat. Cell Biol. 2, 365–70.

Murone, M., Rosenthal, A., de Sauvage, F. J. (1999). Hedgehog signal transduction: from flies to vertebrates. Exp.

Cell Res. 253, 25-33.

Musacchio, A., Hardwick, K. G. (2002). The spindle checkpoint: structural insights into dynamic signalling. Nature

Rev. Mol. Cell Biol. 3, 731–41.

Musacchio, A., Salmon, E. D. (2007). The spindle-assembly checkpoint in space and time. Nat Rev Mol Cell Biol. 8,

379-93.

Nakano, Y., Guerrero, I., Hidalgo, A., Taylor, A., Whittle, J. R. S., Ingham, P. W. (1989). A protein with several

possible membrane-spanning domains encoded by the Drosophila segment polarity gene patched. Nature

341, 508-13.

Narbonne-Reveau, K., Besse, F., Lamour-Isnard, C., Busson, D., Prêt, A. (2006). fused regulates germline cyst

mitosis and differentiation during Drosophila oogenesis. Mech. Dev. 123, 197-209.

Nasmyth, K. (1996) Viewpoint: putting the cell cycle in order. Science 274, 1643-5.

Nasmyth, K., Hunt, T. (1993) Cell cycle: dams and sluices. Nature 366, 634-5.

200

Page 225: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Nau, M. M., Lipkowitz, S. (2003). Comparative genomic organization of the cbl genes. Gene 308 : 103-13.

Ng, M., Diaz-Benjumea, F. J., Cohen, S. M. (1995). Nubbin encodes a POU-domain protein required for

proximaldistal patterning in the Drosophila wing. Development 121, 589–99.

Ng, M., Diaz-Benjumea, F. J., Vincent, J. P., Wu, J., Cohen, S. M. (1996). Specification of the wing by localized

expression of wingless protein. Nature 381, 316–8.

Nicklas, R. B. (1997). How cells get the right chromosomes. Science 275, 632–7.

Nicklas, R. B., Ward, S. C., Gorbsky, G. J. (1995). Kinetochore chemistry is sensitive to tension and may link

mitotic forces to a cell cycle checkpoint. J. Cell Biol. 130, 929–39.

Nicklas, R. B., Waters, J. C., Salmon, E. D., Ward, S. C. (2001). Checkpoint signals in grasshopper meiosis are

sensitive to microtubule attachment, but tension is still essential. J. Cell Sci. 114, 4173–83.

Nigg, E. A. (1993). Cellular substrates of p34(cdc2) and its companion cyclin-dependent kinases. Trends Cell Biol. 3,

296-301.

Nigg, E. A. (1998). Polo-like kinases: positive regulators of cell division from start to finish. Curr Opin Cell Biol. 6,

776-83.

Nigg, E. A. (2001). Mitotic kinases as a regulators of cell division and its checkpoints. Nat. Rev. Mol. Cell Biol. 2, 21-

32.

Nilsson, M., Undèn, A. B., Krause, D., Malmqwist, U., Raza, K., Zaphiropoulos, P. G., Toftgard, R. (2000).

Induction of basal cell carcinomas and trichoepitheliomas in mice overexpressing GLI-1. Proc Natl Acad Sci U

S A. 97, 3438-43.

Nonet, G. H., Stampfer, M. R., Chin, K., Gray, J. W., Collins, C. C., Yaswen, P. (2001). The ZNF217 gene

amplified in breast cancers promotes immortalization of human mammary epithelial cells. Cancer Res. 61,

1250-4.

Noordermeer, J., Johnston, P., Rijsewijk, F., Nusse, R., Lawrence, P. A. (1992). The consequences of

ubiquitous expression of the wingless gene in the Drosophila embryo. Development 116, 711–19.

Nurse, P. (1990) Universal control mechanism regulating onset of M-phase. Nature 344, 503-8.

Nystul, T., Spradling, A., (2010). Regulation of epithelial stem cell replacement and follicle formation in the

Drosophila ovary. Genetics 184, 503-515.

Oelgeschlager, T. (2002). Regulation of RNA polymerase II activity by CTD phosphorylation and cell cycle control. J.

Cell Physiol. 190, 160–9.

O'Hare, K., Rubin, G. M. (1983). Structures of P transposable elements and their sites of insertion and excision in

the Drosophila melanogaster genome. Cell 34, 25-35.

Ohi, R., Gould, K. L. (1999). Regulating the onset of mitosis. Curr. Opin. Cell Biol. 11, 267-73.

Ohlmeyer, J. T., Kalderon, D. (1998). Hedgehog stimulates maturation of Cubitus interruptus into a labile

transcriptional activator. Nature 396, 749–53.

Ohlstein, B., Lavoie, C. A., Vef, O., Gateff, E., McKearin, D. M. (2000). The Drosophila cystoblast differentiation

factor, benign gonial cell neoplasm, is related to DExH-box proteins and interacts genetically with bag-of-

marbles. Genetics 155, 1809–19.

Ohlstein, B., McKearin, D. (1997). Ectopic expression of the Drosophila Bam protein eliminates oogenic germline

stem cells. Development 124, 3651–62.

O'Keefe, L., Dougan, S. T., Gabay, L., Raz, E., Shilo, B. Z., DiNardo, S. (1997). Spitz and Wingless, emanating

from distinct borders, cooperate to establish cell fate across the Engrailed domain in the Drosophila

epidermis. Development 124, 4837-45.

Onai, T., Matsuo-Takasaki, M., Incomata, H., Aramaki, T., Matsumura, M., Yakura, R., Sasai, N., Sasai, Y.,

(2007). XTsh3 is an essential enhancing factor of canonical Wnt signaling in Xenopus axial determination.

Embo J. 26, 2350-60.

Ookata, K., Hisanaga, S., Okano, T., Tachibana, K., Kishimoto, T. (1992) Relocation and distinct subcellular

localization of p34cdc2-cyclin B complex at meiosis reinitiation in starfish oocytes. EMBO J. 11, 1763-72.

201

Page 226: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Oro, A. E., Higgins, K. M., Hu, Z., Bonifas, J. M., Epstein, E. H., Jr., Scott, M. P. (1997). Basal cell carcinomas in

mice overexpressing sonic hedgehog. Science 276, 817–21.

Orsulic, S., Peifer, M. (1996). An in vivo structure function study of Armadillo, the b-catenin homologue, reveals

both separate and overlapping regions of the protein required for cell adhesion and for Wingless signaling. J.

Cell Biol. 134, 1283-300.

Otte, S., Barnikol-Watanabe, S., Vorbruggen, G., Hilschmann, N. (1999). NUCB1, the Drosophila melanogaster

homolog of the mammalian EF-hand proteins NEFA and nucleobindin. Mech Dev 86, 155-8.

Ouyang, B., Li, W., Pan, H., Meadows, J., Hoffmann, I., Dai, W. (1999). The physical association and

phosphorylation of Cdc25C protein phosphatase by Prk. Oncogene 18, 6029–36.

Pagano, M., Pepperkork, R., Verde, F., Ansorge, W., G. Draetta (1992) Cyclin A is required for two points in the

human cell cycle. EMBO J. 11, 961-71.

Pai, L. M., Orsulic, S., Bejsovec, A., Peifer, M. (1997). Negative regulation of Armadillo, a Wingless effector in

Drosophila. Development 124, 2255-66.

Pan, D., Rubin, G. M. (1995) cAMP-dependent protein kinase and hedgehog act antagonistically in regulating

decapentaplegic transcription in Drosophila imaginal disks. Cell 80, 543-52.

Pan, D., Rubin, G. M. (1998). Targeted expression of teashirt induces ectopic eyes in Drosophila. Proc. Natl. Acad.

Sci. U S A 95, 15508-12.

Pan, J., Chen, R. H. (2004). Spindle checkpoint regulates Cdc20p stability in Saccharomyces cerevisiae. Genes Dev.

18, 1439–51.

Panganiban, G. E. F., R. Reuter, M. P. Scott, and F. M. Hoffmann (1990). A Drosophila growth factor homolog,

decapentaplegic, regulates homeotic gene expression within and across germ layers during midgut

morphogenesis. Development 110, 1041-50.

Parker, L. L., Piwnica-Worms, H. (1992). Inactivation of the p34cdc2-cyclin B complex by the human WEE1

tyrosine kinase. Science 257, 1955-7.

Parry, D. H., O'Farrell, P. H. (2001). The schedule of destruction of three mitotic cyclins can dictate the timing of

events during exit from mitosis. Curr Biol 11, 671-83.

Peifer M, Wieschaus E. (1990). The segment polarity gene armadillo encodes a functionally modular protein that is

the Drosophila homolog of human plakaglobin. Cell 63, 1167-76.

Peifer, M. (1995). Cell adhesion and signal transduction: the Armadillo connection. Trends Cell Biol. 5, 224-9.

Peifer, M., Polakis, P. (2000). Wnt signaling in oncogenesis and embryogenesis – a look outside the nucleus. Science

287, 1606-09.

Peifer, M., Rauskolb, C., Williams, M., Riggleman, B., Wieschaus, E. (1991). The segment polarity gene

armadillo interacts with the wingless signaling pathway in both embryonic and adult pattern formation.

Development 111, 1029–43.

Peng, C. Y., Graves, P. R., Thoma, R. S., Wu, Z., Shaw, A. S., Piwnica-Worms, H. (1997). Mitotic and G2

checkpoint control: Regulation of 14-3-3 protein binding by phosphorylation of Cdc25C on serine-216. Science

277, 1501-5.

Perrimon, N. (1994). The genetic basis of patterned baldness in Drosophila. Cell 76, 781-4.

Peter, J. M. (1999). Subunits and substrates of the anaphase promoting complex. Exp. Cell Res. 248, 339-49.

Peters, J. M. (2006). The anaphase promoting complex/ cyclosome: a machine designed to destroy. Nature Rev. Mol.

Cell Biol. 7, 644–56.

Pfleger, C. M., Kirschner, M. W. (2000). The KEN box: an APC recognition signal distinct from the D box target by

Cdh1. Genes Dev. 14, 655-65.

Pfleger, C. M., Lee, E. Kirschner, M.W. (2001a). Substrate recognition by the Cdc20 and Cdh1 components of the

anaphase-promoting complex. Genes Dev. 15, 2396-407.

Pfleger, C. M., Salic, A., Lee, E. Kirschner, M. W. (2001b). Inhibition of Cdh1-APC by the MAD2-related protein

MAD2L2: a novel mechanism for regulating Cdh1. Genes Dev. 15, 1759–64.

202

Page 227: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Pichaud, F., Treisman, J. Desplan, C. (2001). Reinventing a common strategy for patterning the eye. Cell 105, 9-

12.

Pines, J. (1999) Four-dimensional control of the cell cycle. Nature Cell Biol. 1, E-73-9.

Pines, J., Hunter, T. (1991) Human cyclin A and B1 are differentially located in the cell and undergo cell cycle-

dependent nuclear transport. J. Cell Bio. 115, 1-17.

Pines, J., Hunter, T. (1994) The differential localization of human cyclins A and B is due to a cytoplasmic retention

signal in cyclin B. EMBO J. 13, 3772-81.

Pinsky, B. A., Biggins, S. (2005).The spindle checkpoint: tension versus attachment. Trends Cell Biol. 15, 486-93.

Plesca, D., Crosby, M. E., Gupta, D., Almasan, A. (2007). E2F4 function in G2: maintaining G2-arrest to prevent

mitotic entry with damaged DNA. Cell Cycle 6, 1147-52.

Poole, S. J., Kauvar, L. M., Drees, B. Kornberg, T. (1985). The engrailed locus of Drosophila: structural analysis of

an embryonic transcript. Cell 40, 37-43.

Porter, L. A., Donoghue, D. J. (2003). Cyclin B1 and CDK1: nuclear localization and upstream regulators. Prog. Cell

Cycle Res. 5, 335–47.

Qi, W., Tang, Z., Yu, H. (2006). Phosphorylation- and polo-box dependent binding of Plk1 to Bub1 is required for the

kinetochore localization of Plk1. Mol. Biol. Cell 17, 3705–16.

Quiring, R., Walldorf, U., Kloter, U., Gehring, W. J. (1994). Homology of the eyeless gene of Drosophila to the

Small eye gene in mice and Aniridia in humans. Science 265, 785-9.

Raff, J. W., Jeffers, K., Huang, J. Y. (2002). The role of Fzy/Cdc20 and Frzr/Cdh1 in regulating the destruction of

cyclin B in space and time. J. Cell Biol. 157, 1139-49.

Ramalho-Santos, M., Yoon, S., Matsuzaki, Y., Mulligan, R. C., Melton, D. A. (2002). "Stemness": transcriptional

profiling of embryonic and adult stem cells. Science 298, 597-600

Rawlings, J. S., Rosler, K. M., Harrison, D. A. (2004). The JAK/STAT signaling pathway. J. Cell Sci. 117, 1281–83.

Ray, S., Shyam, S., Fraizer, G., Almasan, A. (2007). S-phase checkpoints regulate Apo2 Ligand/Tumor Necrosis

Factor-related Apoptosis-inducing Ligand and CPT-11-induced apoptosis of prostate cancer cells. Mol Cancer

Ther. 6, 1368-78.

Reddy, S. K., Rape, M., Margansky, W. A., Kirschner, M. W. (2007). Ubiquitination by the anaphase-promoting

complex drives spindle checkpoint inactivation. Nature 446, 921–5.

Reed, B. H. and Orr-Weaver, T. L. (1997). The Drosophila gene morula inhibits mitotic functions in the endo cell

cycle and the mitotic cell cycle. Development 124, 3543-53.

Resnitzky, D., Hengst, L., Reed, S. (1995) Cyclin A-associated kinase activity is the rate limiting for entrance into S

phase and it is negatively regulated in G1 by p27 kip1. Mol. Biol. Cell 15, 4347-52.

Reuter, R., Panganiban, G. E. F., Hoffmann, F. M. Scott, M. P. (1990). Homeotic genes regulate the spatial

expression of putative growth factors in the visceral mesoderm of Drosophila embryos. Development 110,

1031-40.

Reuter, R., Panganiban, G. E., Hoffmann, F. M., Scott, M. P. (1990). Homeotic genes regulate the spatial

expression of putative growth factors in the visceral mesoderm of Drosophila embryos. Development. 110,

1031-40.

Reuter, R., Scott, M. P. (1990). Expression and functions of the homoerotic genes Antennapedia and Sex combs

reduced in the embryonic midgut of Drosophila. Development 109, 289-303.

Rieder, C. L., Cole, R. W., Khodjakov, A., Sluder, G. (1995). The checkpoint delaying anaphase in response to

chromosomes monoorientation is mediated by an inhibitory signal produced by unattached kinetochores. J.

Cell Biol. 130, 941-8.

Rieder, C. L., Maiato, H. (2004). Stuck in division or passing through: what happens when cells cannot satisfy the

spindle assembly checkpoint. Dev. Cell 7, 637–51.

Rieder, C. L., Salmon, E. D. (1998). The vertebrate cell kinetochore and itsroles during mitosis. Trends Cell Biol. 8,

310-8.

203

Page 228: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Riese, J., Xiang, Y., Munnerlyn, A., Eresh, S., Hsu, S. C., Grosschedl, R., Bienz, M. (1997). LEF-1, a nuclear

factor coordinating signaling inputs from wingless and decapentaplegic. Cell 88, 777-87.

Riggleman, B., Schedl, P., Wieschaus, E. (1990). Spatial expression of the Drosophila segment polarity gene

armadillo is posttranscriptionally regulated by wingless. Cell 63, 549–60.

Robbins, D. J., Nybakken, K. E., Kobayashi, R., Sisson, J. C., Bishop, J. M., The´rond, P. P. (1997) Hedgehog

elicits signal transduction by means of a large complex containing the kinesin-related protein Costal2.Cell 90,

225–34.

Roberts, D. B. (1998). Drosophila : a practical approach. In Practical approach series, (ed. . Oxford ; New York: IRL

Press at Oxford University Press.

Robertson, H. M., Preston, C. R., Phillis, R. W., Johnson-Schlitz, D. M., Benz, W. K., Engels, W. R. (1988). A

stable genomic source of P element transposase in Drosophila melanogaster. Genetics 118, 461-70.

Robertson, H. M., Preston, C. R., Phillis, R. W., Johnson-Schlitz, D. M., Benz, W. K., Engels, W. R. (1988). A

stable genomic source of P element transposase in Drosophila melanogaster. Genetics 118, 461-70.

Robertson, L. K., Bowling, D. B., Mahaffey, J. P., Imiolczyk, B., Mahaffey, J. W. (2004). An interactive network

of zinc-finger proteins contributes to regionalization of the Drosophila embryo and establishes the domains of

HOM-C protein function. Development 131, 2781-9.

Rochat, A., Kobayashi, K., Barrandon, Y. (1994). Location of stem cells of human hair follicles by clonal analysis.

Cell 76, 1063-73.

Roder, L., Vola, C., Kerridge, S. (1992). The role of the teashirt gene in trunk segmental identity in Drosophila.

Development 115, 1017-33.

Rodriguez, A., Terriente, J., Galindo, M. I., Couso, J. P., Diaz-Benjumea, F. J. (2002). Different mechanisms

initiate and maintain wingless expression in the Drosophila wing hinge. Development 129, 3995–4004.

Roshak, A. K., Capper, E. A., Imburgia, C., Fornwald, J., Scott, G., Marshall, L. A. (2000). The human polo-like

kinase, PLK, regulates cdc2/cyclin B through phosphorylation and activation of the cdc25C phosphatase. Cell

Signal 12, 405–11.

Rothe, M., Nauber, U., Jackle, H. (1989). Three hormone receptor-like Drosophila genes encode an identical DNA-

binding finger. Embo J. 8, 3087-94.

Rudner, A. D., Murray, A. W. (2000). Phosphorylation by Cdc28 activates the Cdc20-dependent activity of the

anaphase-promoting complex. J Cell Biol 149, 1377-90.

Ruhf, M. L., Braun, A., Papoulas, O., Tamkun, J. W., Randsholt, N., Meister, M. (2001). The domino gene of

Drosophila encodes novel members of the SWI2/SNF2 family of DNA-dependent ATPases, which contribute to

the silencing of homeotic genes. Development. 128, 1429-41.

Ruohola, H., Bremer, K., Baker, D., Swedlow, J., Jan, L., Jan, Y., (1991). Role of neurogenic genes in

establishment of follicle cell fate and oocyte polarity during oogenesis in Drosophila. Cell 66, 433-49.

Rushlow, C., Doyle, H., Hoey, T., Levine, M. (1987). Molecular characterization of the zerknullt region of the

Antennapedia gene complex in Drosophila. Genes Dev 1, 1268-79.

Rushlow, C., Frasch, M., Doyle, H., Levine, M. (1987b). Maternal regulation of zerknullt: a homoeobox gene

controlling differentiation of dorsal tissues in Drosophila. Nature 330, 583-6.

Salazar, I., Gomes, R. (1999). teashirt is required for the metaphase-to-anaphase transition in Drosophila

neuroblasts Europ. Dros. Res. Conf. 16, 159.

Saller, E., Kelley, A., Bienz, M. (2002). The transcriptional repressor Brinker antagonizes Wingless signaling. Genes

Dev. 16, 1828-38.

Sambrook, J., Fritsch, E. F., Maniatis, T. (1989). Molecular cloning : a laboratory manual. Cold Spring Harbor,

N.Y.: Cold Spring Harbor Laboratory.

Sanchez, Y., Wong, C., Thoma, R. S., Richman, R., Wu, Z., Piwnica-Worms, H., Elledge, S. J. (1997).

Conservation of the Chk1 checkpoint pathway in mammals: Linkage of DNA damage to Cdk regulation

through Cdc25. Science 277, 1497-501.

204

Page 229: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Sanson, B., Alexandre, C., Fascetti, N., Vincent, J. P. (1999). Engrailed and hedgehog make the range of Wingless

asymmetric in Drosophila embryos. Cell 98, 207-16.

Scaerou, F., Aguilera, I., Saunders, R., Kane, N., Blottiere, L., Karess, R. (1999). The rough deal protein is a

new kinetochore component required for accurate chromosomes segregation in Drosophila. J Cell Sci 112,

3757-68.

Scaerou, F., Starr, D. A., Piano, F., Papoulas, O., Karess, R. E., Goldberg, M. L. (2001). The ZW10 and Rough

Deal checkpoint proteins function together in a large, evolutionarily conserved complex target to the

kinetochore. J Cell Sci 114, 3103-14.

Scanlan, M. J., Chen, Y. T., Williamson, B., Gure, A. O., Stockert, E., Gordan, J. D., Tureci, O., Sahin, U.,

Pfreundschuh, M., Old, L. J. (1998). Characterization of human colon cancer antigens recognized by

autologous antibodies. Int. J. Cancer 76, 652-8.

Schmit , T. L. and Ahmad, N. (2007) Regulation of mitosis via mitotic kinases: new opportunities for cancer

management, Mol. Cancer Ther. 6, 1920-31.

Schulz, C., Kiger, A. A., Tazuke, S. I., Yamashita, Y. M., Pantalena-Filho, L. C., Jones, D. L., Wood, C.G.,

Fuller, M. T. (2004). A misexpression screen reveals effects of bag-of-marbles and TGF beta class signaling

on the Drosophila male germ-line stem cell lineage. Genetics 167, 707–23.

Seo, G. J., Kim, S. E., Lee, Y. M., Lee, J. R., Hahn, M. J., Kim, S. T. (2003). Determination of substrate specificity

and putative substrates of ChK2 kinase. Biochem. Res. Commun. 304, 339-43.

Shah, J. V., Botvinick, E., Bonday, Z., Furnari, F., Berns, M., Cleveland, D. W. (2004). Dynamics of centromere

and kinetochore proteins; implications for checkpoint signaling and silencing. Curr. Biol. 14, 942–52.

Shah, J., Cleveland, D. W. (2000). Waiting for anaphase: Mad2 and the spindle assembly checkpoint. Cell 103, 997-

1000.

Shannon, K. B., Canman, J. C., Salmon, E. D. (2002). Mad2 and BubR1 function in a single checkpoint pathway

that responds to a loss of tension. Mol. Biol. Cell 13, 3706–19.

Shcherbata, H. R., Althauser, C., Findley, S. D., Ruohola-Baker, H. (2004). The mitotic-to-endocycle switch in

Drosophilafollicle cells is executed by Notch-dependent regulation of G1/S, G2/M and M/G1 cell-cycle

transitions. Development 131, 3169-81.

Shippy, T. D., Tomoyasu, Y., Nie, W., Brown, S. J., Denell, R. E. (2008). Do teashirt family genes specify trunk

identity? Insights from the single tiptop/teashirt homolog of Tribolium castaneum. Dev Genes Evol 218, 141–

152.

Shirayama, M., Zachariae, W., Ciosk, R. and Nasmyth, K. (1998). The Polo-like kinase Cdc5p and the WD-repeat

protein Cdc20p/fizzy are regulators and substrates of the anaphase promoting complex in Saccharomyces

cerevisiae. Embo J 17, 1336-49.

Shivdasani, A. A., Ingham, P. W. (2003). Regulation of stem cell maintenance and transit amplifying cell

proliferation by tgf-beta signalling in Drosophila spermatogenesis. Curr. Biol. 13, 2065–72.

Shreeram, S., Demidov, O. N., Hee, W. K., Yamaguchi, H., Onishi, N., Kek, C., Timofeev, O. N., Dudgeon, C.,

Fornace, A. J., Anderson, C. W., Minami Y., Appella, E., Bulavin, D. V. (2006) Wip1 phosphatase

modulates ATM-dependent signaling pathways. Mol. Cell 23, 757-64.

Sigrist, S. J., Lehner, C. F. (1997). Drosophila fizzy-related down-regulates mitotic cyclins and is required for cell

proliferation arrest and entry into endocycles. Cell 90, 671-81

Sigrist, S., Jacobs, H., Stratmann, R., Lehner, C. F. (1995). Exit from mitosis is regulated by Drosophila fizzy and

the sequential destruction of cyclins A, B and B3. Embo J 14, 4827-38

Singh, A., Kango-Singh, M., Choi, K. W., Sun, Y. H. (2004). Dorso-ventral asymmetric functions of teashirt in

Drosophila eye development depend on spatial cues provided by early DV patterning genes. Mech. Dev. 121,

365-70.

Singh, A., Kango-Singh, M., Sun, Y. H. (2002). Eye suppression, a novel function of teashirt, requires Wingless

signaling. Development 129, 4271-80.

205

Page 230: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Sisson, J. C., Ho, K. S., Suyama, K., Scott, M. P. (1997). Costal2, a novel kinesin-related protein in the hedgehog

signaling pathway. Cell 90, 235-45.

Skaer, H. (1993). The alimentary canal. In The development of Drosophila melanogaster (ed. A. Martinez Arias and

M. Bate), pp. 941-1012. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY.

Skoufias, D. A., Andreassen, P. R., Lacroix, F. B., Wilson, L., Margolis, R. L. (2001). Mammalian mad2 and

bub1/bubR1 recognize distinct spindle-attachment and kinetochore-tension checkpoints. Proc. Natl. Acad.Sci.

USA 98, 4492–97.

Smith, L., Ecker, R. (1971) The interaction of steroids with Rana pipiens oocytes in the induction of maturation. Dev.

Biol. 25, 232-47.

Smits, V. A., Klompmaker, R., Arnaud, L., Rijksen, G., Nigg, E. A., Medema, R. H. (2000). Polo-like kinase-1 is a

target of the DNA damage checkpoint. Nat. Cell Biol. 2, 672-6.

Soanes, K. H., Bell, J. B. (2001). The drosophila aeroplane mutant is caused by an I-element insertion into a tissue-

specific teashirt enhancer motif. Genome 44, 919-28.

Soanes, K. H., MacKay, J. O., Core, N., Heslip, T., Kerridge, S., Bell, J. B. (2001). Identification of a regulatory

allele of teashirt (tsh) in Drosophila melanogaster that affects wing hinge development. An adult-specific tsh

enhancer in Drosophila. Mech. Dev. 105, 145-51.

Soanes, K., Bell, J.B., (1999). Rediscovery and further characterization of the aeroplane (ae) wing posture mutation

in Drosophila melanogaster. Genome 42, 403-11.

Solomon, M. J., Glotzer, M., Lee, T. H., Philippe, M., Kirschner, M. W. (1990). Cyclin activation of p34cdc2. Cell

63, 1013-24.

Somma, M. P., Fasulo, B., Cenci, G., Cundari, E., Gatti, M. (2002). Molecular dissection of cytokinesis by RNA

interference in Drosophila cultured cells. Mol Biol Cell 13, 2448-60.

Song X., Xie T. (2002). DE-cadherin-mediated cell adhesion is essential for maintaining somatic stem cells in the

Drosophila ovary. Proc. Natl. Acad. Sci. USA 99, 14813-18.

Song X., Xie T. (2003). Wingless signaling regulates the maintenance of ovarian somatic stem cells in Drosophila.

Development 130, 3259-68.

Song, X., Wong, M. D., Kawase, E., Xi, R., Ding, B. C., McCarthy J. J., Xie T. (2004). Bmp signals from niche

cells directly repress transcription of a differentiation-promoting gene, bag of marbles, in germline stem cells

in the Drosophila ovary. Development 131, 1353–64.

Song, X., Zhu, C. H., Doan, C., Xie, T. (2002). Germline stem cells anchored by adherens junctions in the Drosophila

ovary niches. Science 296, 1855–57.

Starr, D. A., Williams, B. C., Hays, T. S., Goldberg, M.L. (1998). ZW10 helpsrecruit dynactin and dynein to the

kinetochore. J. Cell Biol. 142, 763-74.

Starz-Gaiano, M., Lehmann, R. (2000). Moving towards the next generation. Mech. Dev. 105, 5-18.

Stegmeier, F., Rape, M., Draviam, V. M., Nalepa, G., Sowa, M. E., Ang, X. L., McDonald, E. R. 3rd, Li, M. Z.,

Hannon, G. J., Sorger, P. K., Kirschner, M. W., Harper, J. W., Elledge, S. J. (2007). Anaphase initiation

is regulated by antagonistic ubiquitination and deubiquitination activities. Nature 446, 921–25.

Stiffler, L. A., Ji J. Y., Trautmann, S., Trusty, C., G. Schubiger (1999) Cyclin A and B functions in the early

Drosophila embryos. Development 126, 5505-13.

St-Jacques, B., Dassule, H. R., Karavanova, I., Botchkarev, V. A., Li, J., Danielian, P. S., McMahon, J.A.,

Lewis, P. M., Paus, R., McMahon, A. P. (1998). Sonic hedgehog signaling is essential for hair development.

Curr Biol. 8, 1058-68.

Strathdee, G., Zackai, E.H., Shapiro, R., Kamholz, J., Overhauser, J., (1995). Analysis of clinical variation seen

in patients with 18q terminal deletions. Am. J. Med. Genet. 59, 476–83.

Struhl, G. (1983). Role of the esc+ gene product in ensuring the selective expression of segment-specific homeotic

genes in Drosophila. J. Embryol. Exp. Morphol. 76, 297-331.

Styhler, S., Nakamura, A., Swan, A., Suter, B., Lasko, P. (1998). vasa is required for GURKEN accumulation in the

oocyte, and is involved in oocyte differentiation and germline cyst development. Development 125, 1569–78.

206

Page 231: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Sudakin, V., Chan, G. K., Yen, T. J. (2001). Checkpoint inhibition of the APC/C in HeLa cells is mediated by a

complex of BUBR1, BUB3, CDC20, and MAD2. J. Cell Biol. 154, 925–36.

Sun, Y. H., Tsai, C. J., Green, M. M., Chao, J. L., Yu, C. T., Jaw, T. J., Yeh, J. Y., Bolshakov, V. N, (1995). white

as a reporter gene to detect transcriptional silencers specifying position-specific gene expression during

Drosophila melanogaster eye development. Genetics 141:1075-1086.

Szakmary, A., Cox, D. N., Wang, Z., Lin, H. (2005). Regulatory Relationship among piwi, pumilio, and bag-of-

marbles in Drosophila germline stem cell self-renewal and differentiation. Curr. Biol. 15, 171–8.

Szuts, D., Freeman, M., Bienz, M. (1997). Antagonism between EGFR and Wingless signalling in the larval cuticle of

Drosophila. Development 124, 3209-19.

Tabata, T., Kornberg, T. B. (1994). Hedgehog is a signaling protein with a key role in patterning Drosophila imaginal

discs. Cell 76, 89-102.

Taghli-Lamallem, O., Gallet, A., Lerou, F., Mallapert, P., Vola, C., Kerridge, S., Fasano, L. (2007) Direct

interection between Teashirt and Sex combs reduced proteins, via Tsh’s acidic domain, is essential for

specifying the identity of the prothorax in Drosophila. Dev Biol 307, 142-51.

Tanaka, T. U., Rachidi, N., Janke, C., Pereira, G., Galova, M., Schiebel, E., Stark, M. J., Nasmyth, K. (2002).

Evidence that the Ipl1–Sli15 (Aurora kinase–INCENP) complex promotes chromosome bi-orientation by

altering kinetochore spindle pole connections. Cell 108, 317–29.

Tang, R. H., Zheng, X. L., Callis, T. E., Stansfield, W. E., He, J, Baldwin, A. S., Wang, D. Z., Selzman, C. H.

(2008). Myocardin inhibits cellular proliferation by inhibiting NF-kappaB(p65)-dependent cell cycle

progression. Proc Natl Acad Sci U S A. 105, 3362-7.

Tang, Z., Bharadwaj, R., Li, B., Yu, H. (2001). Mad2-independent inhibition of APCCdc20 by the mitotic checkpoint

protein BubR1. Dev. Cell 1, 227–37.

Tang, Z., Shu, H., Oncel, D., Chen, S., Yu, H. (2004). Phosphorylation of Cdc20 by Bub1 provides a catalytic

mechanism for APC/C inhibition by the spindle checkpoint. Mol. Cell 16, 387–97.

Taylor, S. S., McKeon, F. (1997) Kinetochore localization of murine Bub1 is required for normal mitotic timing and

checkpoint response to spindle damage. Cell .89, 727-35.

Taylor, S. S., Scott, M. I., Holland, A. J. (2004). The spindle checkpoint: a quality control echanism which ensures

accurate chromosome segregation. Chromosome Res. 12, 599–616.

Tazuke, S. I., Schulz, C., Gilboa, L., Fogarty, M., Mahowald, A. P., Guichet A., Ephrussi A., Wood C. G.,

Lehmann R., Fuller M. T. (2002). A germline-specific gap junction protein required for survival of

differentiating early germ cells. Development 129, 2529–39.

Thien, C. B., Langdon, W. Y. (2001). Cbl : many adaptations to regulate protein tyrosine Kinases. Nat. Rev. Cell

Biol. 2, 294-307.

Toyoshima, F., Moriguchi, T., Wada, A., Fukuda, M., Nishida, E. (1998) Nuclear export of cyclin B1 and its

possible role in the DNA damage-induced G2 checkpoint. EMBO J. 17, 2728-35.

Treisman, J. E., Heberlein, U. (1998). Eye development in Drosophila: Formation of the eye field and control of

differentiation. Curr. Top. Dev. Biol. 39, 119–58.

Tremml, G, Bienz, M. (1989). Homeotic gene expression in the visceral mesoderm of Drosophila embryos. EMBO J.

9, 2677-85.

Tremml, G., Bienz, M. (1989). Homeotic gene expression in the visceral mesoderm of Drosophila embryos. EMBO J.

8, 2677-85.

Tremml, G., Bienz, M. (1992). Induction of labial expression in the Drosophila endoderm: Response elements for dpp

signalling and for autoregulation. Development 116, 447-56.

Tsukiyama, T., Daniel, C., Tamkun, J., Wu, C. (1995). ISWI, a member of the SWI2/SNF2 ATPase family, encodes

the 140 kDa subunit of the nucleosome remodeling factor. Cell 83, 1021-26.

Tulina, N., Matunis, E. (2001). Control of stem cell self-renewal in Drosophila spermatogenesis by JAK- STAT

signaling. Science 294, 2546–49.

207

Page 232: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Tworoger, M., Larkin, M. K., Bryant, Z., Ruohola-Baker, H. (1999) Mosaic analysis in the Drosophila ovary

reveals a common Hedgehog inducible precursor stage for stalk and polar cells. Genetics 151, 739-48.

Van de Wetering, M., Cavallo, R., Dooijes, D., van Beest, M., van Es, J., Loureiro, J., Ypma, A., Hursh, D.,

Jones, T., Bejsovec, A., Peifer, M., Mortin, M., Clevers, H. (1997). Armadillo coactivates transcription

driven by the product of the Drosophila segment polarity gene dTCF. Cell 88, 789–99.

Van den Heuvel, M., Nusse, R., Johnston, P., Lawrence, P. A. (1989). Distribution of the wingless gene product in

Drosophila embryos: a protein involved in cell-cell communication. Cell 59, 739-49.

Van Vugt, M. A., Smits, V. A., Klompmaker, R., Medema, R. H. (2001). Inhibition of Polo-like Kinase-1 by DNA

damage occurs in an ATM- or ATR-dependent fashion. J Biol Chem 276, 41656-60.

Vigneron, S., Prieto, S., Bernis, C., Labbe, J. C., Castro, A., Lorca, T. (2004). Kinetochore localization of spindle

checkpoint proteins: who controls whom? Mol. Biol. Cell 15, 4584–96.

Von, Ohlen, T., Lessing, D., Nusse, R. Hooper, J. E. (1997). Hedgehog signaling regulates transcription through

cubitus interruptus, a sequence-specific DNA binding protein. Proc. Natl. Acad. Sci. U S A. 94, 2404-9.

Vrailas, A. D., Moses, K. (2006). Smoothened, thickveins and the genetic control of cell cycle and cell fate in the

developing Drosophila eye. Mech. Dev. 123, 151–65.

Wahl, G. M., Carr, A. M. (2001). The evolution of diverse biological responses to DNA damage: Insights from yeast

and p53. Nat. Cell Biol. 3, E277-86.

Wakefield, J., Huang, J., Raff, J. (2000) Centrosomes have a role in regulating the destruction of cyclin B in early

drosophila embryos. Curr. Biol. 10, 1367-70.

Waltzer, L., Vandel, L., Bienz, M. (2001). Teashirt is required for transcriptional repression mediated by high

Wingless levels. Embo J. 20, 137-45.

Wang, Q. T., Holmgren, R. A. (1999). The subcellular localization and activity of Drosophila cubitus interruptus are

regulated at multiple levels. Development 126, 5097–06.

Wang, Y., Dakubo, G. D., Thurig, S., Mazerolle, C. J., Wallace, V. A. (2005) Retinal ganglion cell-derived sonic

hedgehog locally controls proliferation and the timing of RGC development in the embryonic mouse retina.

Development 132, 5103-13.

Wang, Z, Lin, H. (2005). The division of Drosophila germline stem cells and their precursors requires a specific cyclin.

Curr. Biol. 15, 328–33.

Wang, Z., Lin, H. (2004). Nanos maintains germline stem cell self-renewal by preventing differentiation. Science

303, 2016–19.

Waters, J. C., Chen, R. H., Murray, A. W., Salmon, E. D. (1998). Localization of Mad2 to kinetochores depends on

microtubule attachment, not tension. J. Cell Biol. 141, 1181–91.

Watkins, D. N., Berman, D. M., Burkholder, S. G., Wang, B., Beachy, P. A., Baylin, S. B. (2003). Hedgehog

signalling within airway epithelial progenitors and in small-cell lung cancer. Nature 422, 313–17.

Weinert, T. A., Hartwell, L. H. (1988). The RAD9 gene controls the cell cycle reponse to DNA damage in

Saccharomyces cerevisiae. Science 241, 317-22.

Whitfield, W. G., Gonzalez, C., Maldonado-Colina, G., Glover, D. M. (1990). The A- and B-type cyclins of

Drosophila are accumulated and destroyed in temporally distinct events that define separable phases of the

G2-M transition. EMBO J. 9, 2563-72.

Wiellette, E. L., Harding, K. W., Mace, K. A., Ronshaugen, M. R., Wang, F. Y., McGinnis, W. (1999). spen

encodes an RNP motif protein that interacts with Hox pathways to repress the development of head-like

sclerites in the Drosophila trunk. Development 126, 5373-85.

Wieschaus, E., Szabad, J. (1979). The development and function of the female germ line in Drosophila

melanogaster: a cell lineage study. Dev. Biol. 68, 29–46.

Wiese, C., Zheng, Y. (2000). A new function for the gamma-tubulin ring complex as a microtubule minus-end cap.

Nat. Cell Biol. 2, 358–64.

Wigge, P. A., Kilmartin, J. V. (2001). The Ndc80p complex from Saccharomyces cerevisiae contains conserved

centromere components and has a function in chromosome segregation. J. Cell Biol. 152, 349–60.

208

Page 233: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Wilkinson, K. D. (1999). Ubiquitin-dependent signalling: the role of ubiquitination in the response of cells to their

environment. J. Nutr. 129, 1933-6.

Willert, K., Nusse, R. (1998). Beta-catenin: a key mediator of Wnt signaling. Curr. Opin. Genet. Dev. 8, 95-102.

Wojcik, E., Basto, R., Serr, M., Scaërou, F., Karess, R., Hays, T. (2001). Kinetochore dynein: its dynamics and

role in the transport of the Rough deal checkpoint protein. Nature Cell Biol. 3, 1001–07.

Wong, O. K., Fang, G. (2005). Plx1 is the 3F3/2 kinase responsible for targeting spindle checkpoint proteins to

kinetochores. J. Cell Biol. 170, 709–19.

Wood, K. W., Sakowicz, R., Goldstein, L. S., Cleveland, D. W. (1997). CENP-E is a plus end-directed kinetochore

motor required for metaphase chromosome alignment. Cell 91, 357–66.

Wu, J., Cohen, S. M. (2000). Proximal distal axis formation in the Drosophila leg: distinct functions of teashirt and

homothorax in the proximal leg. Mech. Dev. 94, 47-56.

Wu, J., Cohen, S.M. (2002). Repression of Teashirt marks the initiation of wing development. Development 129,

2411-8.

Xi, R., Doan C., Liu D., Xie T. (2005). Pelota controls self-renewal of germline stem cells by repressing a Bam-

independent differentiation pathway. Development 132, 5365-74.

Xi, R., Xie T. (2005) Stem cell self-renewal controlled by chromatin remodelling factors. Science 310, 1487-9.

Xia, G., Luo, X., Habu, T., Rizo, J., Matsumoto, T., Yu, H. (2004). Conformation-specific binding of p31comet

antagonizes the function of Mad2 in the spindle checkpoint. EMBO J. 23, 3133–43.

Xie, J., Murone, M., Luoh, S. M., Ryan, A., Gu, Q., Zhang, C., Bonifas, J. M., Lam, C. W., Hynes, M., Goddard,

A., Rosenthal, A, Epstein, E. H. Jr., de Sauvage, F. J. (1998). Activating Smoothened mutations in

sporadic basal-cell carcinoma. Nature. 391, 90-2.

Xie, T., Kawase, E., Kirilly, D., Wong, M. D. (2005). Intimate relationships with their neighbors: Tales of stem cells

in Drosophila reproductive systems. Dev. Dyn. 232, 775–90.

Xie, T., Spradling, A. C. (1998). decapentaplegic is essential for the maintenance and division of germline stem cells

in the Drosophila ovary. Cell 94, 251–60.

Xie, T., Spradling, A. C. (2000). A niche maintaining germ line stem cells in the Drosophila ovary. Science 290, 328–

30.

Xu, T., Caron, L. A., Fehon, R. G., Artavanis-Tsakonas, S., (1992). The involvement of the Notch locus in

Drosophila oogenesis. Development 115, 913-22.

Yamashita, Y. M., Jones, D. L., Fuller, M. T. (2003). Orientation of asymmetric stem cell division by the APC tumor

suppressor and centrosome. Science 301, 1547-50.

Yang, J., Bardes, E. S., Moore, J. D., Brennan, J., Powers, M. A., Kornbluth, S. (1998) Control of cyclin B1

localization through regulated biding of the nuclear export factor CRM1. Genes Dev. 12, 2131-43.

Yao, S., Lum, L., Beachy, P. (2006). The Ihog Cell-Surface Proteins Bind Hedgehog and Mediate Pathway Activation.

Cell 125, 343–57.

Ying, Q. L., Nichols, J., Chambers, I., Smith, A. (2003). BMP induction of Id proteins suppresses differentiation and

sustains embryonic stem cell self-renewal in collaboration with STAT3. Cell 115, 281–92.

Yu, H. (2006). Structural activation of Mad2 in the mitotic spindle checkpoint: the two-state Mad2 model versus the

Mad2 template model. J. Cell Biol. 173, 153–7.

Yu, H. (2007). Cdc20:a WD40 activator for a cell cycle degradation machine. Mol. Cell 27, 3-16.

Yu, J., Carroll, T. J., McMahon, A. P. (2002). Sonic hedgehog regulates proliferation and differentiation of

mesenchymal cells in the mouse metanephric kidney. Development. 129, 5301-12.

Yuen, T., Wurmbach, E., Pfeffer, R. L., Ebersole, B. J., Sealfon, S. C. (2002). Accuracy and calibration of

commercial oligonucleotide and custom cDNA microarrays. Nucleic Acids Research 30, art. no.-e48.

Zachariae, W., Nasmyth, K. (1999). Whose end is destruction: cell division and the anaphase-promoting complex.

Genes Dev 13, 2039-58

209

Page 234: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

REFERENCES

Zhang, J., Niu, C., Ye, L., Huang, H., He, X., Tong, W. G., Ross, J., Haug, J., Johnson, T., Feng, J. Q., Harris,

S., Wiedemann, L. M., Mishina, Y., Li, L. (2003). Identification of the haematopoietic stem cell niche and

control of the niche size. Nature 425, 836-41.

Zhang, W., Kang, J., Cole, F., Yi, M., Krauss, R. S., (2006). Cdo Functions at Multiple Points in the Sonic Hedgehog

Pathway, and Cdo-Deficient Mice Accurately Model Human Holoprosencephaly. Developmental Cell 10, 657–

65.

Zhang, Y., Kalderon, D., (2000). Regulation of cell proliferation and patterning in Drosophila oogenesis by Hedgehog

signaling. Development 127, 2165-76.

Zhang, Y., Kalderon, D. (2001). Hedgehog acts as a somatic stem cell factor in the Drosophila ovary. Nature 410,

599–604.

Zhao, J. J., Lazzarini, R. A., Pick, L., (1993). The mouse Hox-1.3 gene is functionally equivalent to the Drosophila

Sex combs reduced gene. Genes Dev. 7, 343–54.

Zheng, Y., Wong, M. L., Alberts, B., Mitchison, T. (1995). Nucleation of microtubule assembly by a gamma-

tubulin-containing ring complex. Nature 378, 578–83.

Zhou, J., Yao, J., Joshi, H. C. (2002). Attachment and tension in the spindle assembly checkpoint. J. Cell Sci. 115,

3547–55.

Zhu C. H., Xie T. (2003). Clonal expansion of ovarian germline stem cells during niche formation in Drosophila.

Development 130, 2579-88.

Zimmerman, W., Doxsey, S. J. (2000). Construction of centrosomes and spindle poles by molecular motor-driven

assembly of protein particles. Traffic 1, 927–34.

Zirin, J. D., Mann, R. S. (2004). Differing strategies for the establishment and maintenance of teashirt and

homothorax repression in the Drosophila wing. Development 131, 5683-93.

Zirin, J. D., Mann, R. S. (2007). Nubbin and Teashirt mark barriers to clonal growth along the proximal-distal axis of

the Drosophila wing. Dev. Biol. 304, 745–58.

210

Page 235: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX

Page 236: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal
Page 237: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

Appendix I- Microarays Results for expression patterning of tshNG1 neuroblasts expression comparing to wild-type (ordered

by lower bound of fold change (LBFC), after using a cut-off of 1.3)

Table I- Results of the up- or down-regulation genes in tshNG1 homozygous neuroblats which have been ordered by lower bound of

fold change (LBFC), after using a cut-off of 1.3. The positive LBFC represent the up regulated genes while the negative LBFC represent

the downregulated genes. Only the genes with the absolute value of LBFC upper than 1.3 are represented.

Gene LBFC

Immune induced molecule 1 7,0

FB:FBgn0034330 /sym=CG18107 /name= /prod= /func= /map=55C9-55C9 /transc=CT40695 /len=248 /GB:AE003799 5,5

FB:FBgn0036833 /sym=CG3819 /name= /prod= /func= /map=75E4-75E4 /transc=CT12789 /len=1272 /GB:AE003518 4,4

FB:FBgn0034294 /sym=CG5765 /name= /prod= /func= /map=55B1-55B1 /transc=CT18116 /len=1528 /GB:AE003801 4,0

FB:FBgn0040735 /sym=CG16836 /name= /prod= /func= /map=55C9-55C9 /transc=CT37415 /len=298 /GB:AE003799 3,3

Drosomycin 3,3

FB:FBgn0031246 /sym=CG11592 /name= /prod= /func=structural protein /map=21B6-21B6 /transc=CT33137 /len=1668 /GB:AE003590 3,3

FB:FBgn0030072 /sym=CG7090 /name= /prod= /func=enzyme /map=8C3-8C3 /transc=CT21911 /len=763 /GB:AE003445 3,2

FB:FBgn0040734 /sym=CG15065 /name= /prod= /func= /map=55C9-55C9 /transc=CT34936 /len=123 /GB:AE003799 3,2

FB:FBgn0036393 /sym=CG17362 /name= /prod= /func= /map=70C9-70C9 /transc=CT33207 /len=552 /GB:AE003536 3,2

Lysozyme A /// Lysozyme C 3,1

FB:FBgn0034900 /sym=CG12491 /name= /prod= /func= /map=59F7-59F7 /transc=CT32938 /len=474 /GB:AE003461 3,1

Attacin-A 3,0

Odorant-binding protein 56a 3,0

FB:FBgn0034296 /sym=CG10912 /name= /prod= /func= /map=55B2-55B2 /transc=CT30553 /len=875 /GB:AE003801 /note=3prime sequence from clone BDGP:GH07575.3prime-hit

3,0

FB:FBgn0038074 /sym=CG6188 /name= /prod= /func= /map=87C7-87C7 /transc=CT19392 /len=941 /GB:AE003696 3,0

Esterase 6 2,9

FB:FBgn0028920 /sym=BG:DS00941.12 /name= /prod= /func= /map=34D4-34D4 /transc=CT25774 /len=826 /GB:AE003641 2,8

FB:FBgn0034512 /sym=CG18067 /name= /prod= /func= /map=57A7-57A7 /transc=CT40493 /len=773 /GB:AE003791 2,8

FB:FBgn0028534 /sym=BG:DS00941.13 /name= /prod= /func= /map=34D4-34D4 /transc=CT23894 /len=864 /GB:AE003641 2,7

FB:FBgn0039419 /sym=CG12290 /name= /prod=G-protein coupled receptor-like /func=G protein linked receptor /map=97A6-97A6 /transc=CT19320 /len=3056 /GB:AE003755 /note=3prime sequence from clone BDGP:GH12381.3prime-hit

2,7

Drosocin 2,7

Page 238: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

FB:FBgn0038241 /sym=CG8087 /name= /prod= /func= /map=88C7-88C7 /transc=CT24214 /len=461 /GB:AE003705 2,6

FB:FBgn0039316 /sym=CG11893 /name= /prod= /func= /map=96C7-96C7 /transc=CT33087 /len=1301 /GB:AE003751 2,6

FB:FBgn0031560 /sym=CG16713 /name= /prod= /func= /map=24A4-24A4 /transc=CT37195 /len=298 /GB:AE003579 2,5

Immune induced molecule 23 2,5

FB:FBgn0030262 /sym=CG2081 /name= /prod= /func= /map=10A3-10A3 /transc=CT6756 /len=712 /GB:AE003485 2,5

FB:FBgn0028533 /sym=BG:DS00941.14 /name= /prod= /func= /map=34D4-34D4 /transc=CT23898 /len=913 /GB:AE003641 2,5

FB:FBgn0031561 /sym=CG16712 /name= /prod= /func= /map=24A4-24A4 /transc=CT37183 /len=336 /GB:AE003579 2,5

FB:FBgn0032209 /sym=CG18144 /name= /prod= /func= /map=31D4-31D4 /transc=CT40888 /len=365 /GB:AE003628 2,5

FB:FBgn0035611 /sym=CG13285 /name= /prod= /func= /map=64E1-64E2 /transc=CT32571 /len=597 /GB:AE003565 2,5

FB:FBgn0037181 /sym=CG11370 /name= /prod= /func= /map=79F2-79F2 /transc=CT21093 /len=1173 /GB:AE003598 2,4

Trypsin 2,4

Henna 2,4

Glutathione S transferase E6 2,3

Ecdysone-dependent gene 91 2,2

FB:FBgn0032282 /sym=CG7299 /name= /prod= /func= /map=32A1-32A1 /transc=CT22515 /len=534 /GB:AE003629 2,2

FB:FBgn0036419 /sym=CG13482 /name= /prod= /func= /map=70D5-70D5 /transc=CT32848 /len=309 /GB:AE003535 2,2

FB:FBgn0028921 /sym=BG:DS00941.11 /name= /prod= /func= /map=34D4-34D4 /transc=CT37476 /len=1217 /GB:AE003641 /note=3prime sequence from clone BDGP:GH07914.3prime-hit

2,2

FB:FBgn0031908 /sym=CG5177 /name= /prod= /func=enzyme /map=27F6-27F6 /transc=CT16575 /len=980 /GB:AE003617 /note=3prime sequence from clone BDGP:LD18740.3prime-hit

2,2

Juvenile hormone-inducible protein 26 2,2

FB:FBgn0033820 /sym=CG4716 /name= /prod= /func= /map=50A1-50A1 /transc=CT15231 /len=783 /GB:AE003819 /note=3prime sequence from clone BDGP:GH06388.3prime-hit

2,2

Alcohol dehydrogenase 2,2

FB:FBgn0038783 /sym=CG4367 /name= /prod= /func= /map=92D9-92D9 /transc=CT14252 /len=642 /GB:AE003730 2,1

upheld 2,1

Cytochrome b5-related 2,1

FB:FBgn0028542 /sym=BG:DS00180.8 /name= /prod= /func=cell adhesion /map=34E1-34E1 /transc=CT37470 /len=3394 /GB:AE003642 2,1

Troponin C at 73F 2,1

FB:FBgn0031562 /sym=CG3604 /name= /prod=trypsin inhibitor-like /func=enzyme inhibitor /map=24A4-24A4 /transc=CT11958 /len=536 /GB:AE003579 2,1

FB:FBgn0034203 /sym=CG17288 /name= /prod= /func= /map=54A3-54A3 /transc=CT35735 /len=996 /GB:AE003804 2,1

FB:FBgn0028510 /sym=BG:DS07851.3 /name= /prod= /func= /map=35C4-35C4 /transc=CT35205 /len=502 /GB:AE003647 2,1

FB:FBgn0034899 /sym=CG13560 /name= /prod= /func= /map=59F7-59F7 /transc=CT32937 /len=546 /GB:AE003461 2,1

FB:FBgn0033589 /sym=CG13227 /name= /prod= /func= /map=47D8-47D8 /transc=CT32471 /len=363 /GB:AE003826 2,0

FB:FBgn0033593 /sym=CG9080 /name= /prod= /func= /map=47E1-47E1 /transc=CT26066 /len=435 /GB:AE003826 2,0

FB:FBgn0036181 /sym=CG18331 /name= /prod= /func= /map=68C9-68C9 /transc=CT41625 /len=360 /GB:AE003544 2,0

Jonah 74E 2,0

214

Page 239: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

Rhythmically expressed gene 3 2,0

FB:FBgn0034364 /sym=CG5493 /name= /prod= /func= /map=55E4-55E4 /transc=CT17422 /len=967 /GB:AE003799 2,0

FB:FBgn0039896 /sym=CG1629 /name= /prod= /func= /map=102A3-102A3 /transc=CT4279 /len=1158 /GB:AE003844 2,0

FB:FBgn0038420 /sym=CG10311 /name= /prod= /func= /map=89B22-89B22 /transc=CT28967 /len=719 /GB:AE003713 2,0

Misexpression suppressor of ras 4 1,9

FB:FBgn0040733 /sym=CG15068 /name= /prod= /func= /map=55C9-55C9 /transc=CT34939 /len=189 /GB:AE003799 1,9

Lysozyme A /// Lysozyme D 1,9

/// 1,9

Lysozyme E 1,9

FB:FBgn0033928 /sym=CG13941 /name= /prod= /func=signal transduction /map=50F6-50F6 /transc=CT33487 /len=582 /GB:AE003815 1,9

Tissue inhibitor of metalloproteases 1,9

FB:FBgn0032726 /sym=CG10621 /name= /prod= /func= /map=37B8-37B8 /transc=CT29750 /len=1470 /GB:AE003661 1,9

Pherokine 3 1,9

FB:FBgn0030138 /sym=CG2996 /name= /prod= /func=enzyme /map=8E9-8E10 /transc=CT7882 /len=12908 /GB:AE003447 /note=3prime sequence from clone BDGP:SD01877.3prime-hit

1,9

Odorant-binding protein 18a 1,9

Tetraspanin 42El 1,9

FB:FBgn0031558 /sym=CG16704 /name= /prod= /func= /map=24A4-24A4 /transc=CT37177 /len=318 /GB:AE003579 1,9

FB:FBgn0035743 /sym=CG15829 /name= /prod=diazepam-binding inhibitor-like protein /func=enzyme inhibitor /map=65E5-65E5 /transc=CT39291 /len=249 /GB:AE003560

1,9

BDGP:GH07188.complete-hit /ESTpos=maps 3prime of FB:FBgn0039613 1,8

Punch 1,8

FB:FBgn0032785 /sym=CG10026 /name= /prod=alpha-tocopherol transfer relative protein /func=transcription factor /map=37E3-37E3 /transc=CT28189 /len=1143 /GB:AE003663 /note=3prime sequence from clone BDGP:SD01558.3prime-hit

1,8

FB:FBgn0038243 /sym=CG8066 /name= /prod=cystatin-like /func=cysteine protease inhibitor /map=88C7-88C7 /transc=CT24182 /len=418 /GB:AE003705 1,8

Glutathione S transferase E7 1,8

Troponin C at 47D 1,8

Esterase-7 1,8

FB:FBgn0031220 /sym=CG4822 /name= /prod=ATP-binding cassette transporter-like /func=enzyme /map=21B1-21B1 /transc=CT41972 /len=2894 /GB:AE003590

1,8

FB:FBgn0035742 /sym=CG8629 /name= /prod=diazepam-binding inhibitor-like protein /func=enzyme inhibitor /map=65E5-65E5 /transc=CT14596 /len=255 /GB:AE003560

1,8

Larval cuticle protein 3 1,8

FB:FBgn0036616 /sym=CG4893 /name= /prod= /func= /map=72F1-72F1 /transc=CT15728 /len=716 /GB:AE003527 1,8

Elastin-like 1,8

FB:FBgn0036996 /sym=CG5932 /name= /prod=triacylglycerol lipase-like /func=enzyme /map=77C1-77C1 /transc=CT18587 /len=1461 /GB:AE003591 /note=3prime sequence from clone BDGP:LP10120.3prime-hit

1,8

FB:FBgn0034289 /sym=CG10910 /name= /prod= /func=structural protein /map=55B1-55B1 /transc=CT30545 /len=1407 /GB:AE003801 1,8

215

Page 240: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

FB:FBgn0032427 /sym=CG5453 /name= /prod= /func= /map=33D4-33D4 /transc=CT17294 /len=819 /GB:AE003636 /note=3prime sequence from clone BDGP:GH02216.3prime-hit

1,8

FB:FBgn0034583 /sym=CG10527 /name= /prod= /func= /map=57B20-57B20 /transc=CT29543 /len=1133 /GB:AE003452 /note=3prime sequence from clone BDGP:LD46156.3prime-hit

1,8

FB:FBgn0038242 /sym=CG14852 /name= /prod= /func= /map=88C7-88C7 /transc=CT34668 /len=525 /GB:AE003705 1,8

Myosin light chain 2 1,8

FB:FBgn0031692 /sym=CG6514 /name= /prod= /func=ligand binding or carrier /map=25C10-25C10 /transc=CT20253 /len=536 /GB:AE003609 1,8

FB:FBgn0038113 /sym=CG11668 /name= /prod=serine protease /func=endopeptidase /map=87D11-87D11 /transc=CT36689 /len=1197 /GB:AE003698 1,8

FB:FBgn0030968 /sym=CG7322 /name= /prod= /func=enzyme /map=17E4-17E4 /transc=CT22587 /len=729 /GB:AE003510 1,7

Cyclic-nucleotide-gated ion channel protein 1,7

Lysozyme B 1,7

FB:FBgn0036787 /sym=CG4306 /name= /prod= /func= /map=75C6-75C6 /transc=CT14001 /len=989 /GB:AE003520 1,7

FB:FBgn0034709 /sym=CG3074 /name= /prod= /func=endopeptidase /map=58C5-58C7 /transc=CT10308 /len=1982 /GB:AE003456 /note=3prime sequence from clone BDGP:GM06507.3prime-hit

1,7

FB:FBgn0022341 /sym=CG17467 /name= /prod= /func= /map=102E3-102E3 /transc=CT38621 /len=1002 /GB:AE003846 1,7

FB:FBgn0030237 /sym=CG15209 /name= /prod= /func= /map=9F4-9F4 /transc=CT35141 /len=444 /GB:AE003484 1,7

globin 1 1,7

brother of tout-velu 1,7

Myosin alkali light chain 1 1,7

FB:FBgn0032773 /sym=CG15825 /name= /prod= /func= /map=37D7-37E1 /transc=CT38769 /len=1906 /GB:AE003662 /note=3prime sequence from clone BDGP:HL01053.3prime-hit

1,7

Jonah 65Aiii 1,7

FB:FBgn0010070 /sym=Msp-300 /name=Muscle-specific protein 3 /prod= /func=actin binding /map=25C8-25C8 /transc=CT41354 /len=2016 /GB:AE003608 /note=3prime sequence from clone BDGP:SD10272.3prime-hit

1,7

Galactose-specific C-type lectin 1,7

BDGP:LD47230.3prime-hit /ESTpos=maps in FB:FBgn0034661 /sym=CG4386 /name= /prod=serine protease /func=endopeptidase /map=58A2-58A2 /transc=CT14320 /len=559

1,7

FB:FBgn0038243 /sym=CG8066 /name= /prod=cystatin-like /func=cysteine protease inhibitor /map=88C7-88C7 /transc=CT24182 /len=418 /GB:AE003705 1,7

FB:FBgn0038647 /sym=CG14302 /name= /prod= /func= /map=91C1-91C2 /transc=CT33932 /len=268 /GB:AE003723 1,7

Transferrin 1 1,7

FB:FBgn0031565 /sym=CG10030 /name= /prod= /func=DNA replication factor /map=24A5-24A5 /transc=CT28231 /len=4588 /GB:AE003579 /note=3prime sequence from clone BDGP:HL03084.3prime-hit

1,7

Myosin heavy chain 1,6

FB:FBgn0030341 /sym=CG1967 /name= /prod= /func= /map=10F1-10F2 /transc=CT6115 /len=953 /GB:AE003487 1,6

FB:FBgn0015714 /sym=Cyp6a17 /name= /prod=cytochrome P450, CYP6A17 /func=cytochrome P45 ; EC:1.14.14.1 | inferred from sequence similarity /map=51D2-51D2 /transc=CT28783 /len=1519 /GB:AE003813 /note=3prime sequence from clone BDGP:GH10635.3prime-hit

1,6

FB:FBgn0033565 /sym=CG18003 /name= /prod= /func= /map=47C6-47C6 /transc=CT40270 /len=1255 /GB:AE003828 /note=3prime sequence from clone BDGP:GH14288.3prime-hit

1,6

FB:FBgn0036234 /sym=CG17824 /name= /prod=peritrophin-like /func=cell adhesion /map=68E3-68E3 /transc=CT39558 /len=2415 /GB:AE003543 1,6

FB:FBgn0038009 /sym=CG17738 /name= /prod= /func= /map=87B8-87B8 /transc=CT34537 /len=333 /GB:AE003695 1,6

216

Page 241: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

FB:FBgn0038115 /sym=CG7966 /name= /prod=selenium-binding protein-like /func=ligand binding or carrier /map=87D11-87D11 /transc=CT23978 /len=1498 /GB:AE003698 /note=3prime sequence from clone BDGP:GH14316.3prime-hit

1,6

Tropomyosin 2 1,6

FB:FBgn0032283 /sym=CG7296 /name= /prod= /func= /map=32A1-32A1 /transc=CT22519 /len=575 /GB:AE003629 1,6

Pre-intermoult gene 1 1,6

Jonah 25Bii 1,6

FB:FBgn0031937 /sym=CG13795 /name= /prod= /func=neurotransmitter transporter /map=28C2-28C2 /transc=CT33284 /len=927 /GB:AE003618 1,6

Dromyosuppressin 1,6

pugilist 1,6

FB:FBgn0030484 /sym=CG1681 /name= /prod=glutathione transferase-like /func=enzyme /map=11F1-11F1 /transc=CT4698 /len=1098 /GB:AE003492 /note=3prime sequence from clone BDGP:GH16740.3prime-hit

1,6

Muscle protein 20 1,6

Jonah 25Bi 1,6

FB:FBgn0032897 /sym=CG9336 /name= /prod= /func= /map=38F1-38F1 /transc=CT5238 /len=660 /GB:AE003668 1,6

FB:FBgn0036525 /sym=CG13451 /name= /prod= /func= /map=71E3-71E3 /transc=CT32813 /len=195 /GB:AE003530 1,6

FB:FBgn0035917 /sym=CG6416 /name= /prod= /func=enzyme /map=66D9-66D10 /transc=CT20013 /len=1175 /GB:AE003554 /note=3prime sequence from clone BDGP:GH19182.3prime-hit

1,6

FB:FBgn0032426 /sym=CG15493 /name= /prod= /func= /map=33D4-33D4 /transc=CT35594 /len=468 /GB:AE003636 1,6

FB:FBgn0032638 /sym=CG6639 /name= /prod= /func=endopeptidase /map=36C2-36C2 /transc=CT20606 /len=1493 /GB:AE003655 1,6

FB:FBgn0036422 /sym=CG3868 /name= /prod= /func=signal transduction /map=70D7-70D7 /transc=CT12881 /len=1161 /GB:AE003534 /note=3prime sequence from clone BDGP:GH23259.3prime-hit

1,6

FB:FBgn0039644 /sym=CG11897 /name= /prod=ATP-binding cassette transporter /func=transporter /map=99A2-99A3 /transc=CT37062 /len=4458 /GB:AE003768 /note=3prime sequence from clone BDGP:LD17001.3prime-hit

1,6

Cystatin-like 1,6

Calpain-A 1,6

FB:FBgn0035112 /sym=CG13877 /name= /prod= /func= /map=61A6-61A6 /transc=CT33404 /len=324 /GB:AE003467 1,6

FB:FBgn0030679 /sym=CG8206 /name= /prod= /func= /map=13E16-13E16 /transc=CT24425 /len=997 /GB:AE003500 /note=3prime sequence from clone BDGP:GH04557.3prime-hit

1,6

FB:FBgn0036146 /sym=CG14141 /name= /prod= /func=protein kinase /map=68A8-68A8 /transc=CT33742 /len=444 /GB:AE003545 1,6

FB:FBgn0038644 /sym=CG7710 /name= /prod= /func= /map=91B8-91B8 /transc=CT23373 /len=460 /GB:AE003723 1,6

FB:FBgn0039208 /sym=CG6643 /name= /prod= /func=enzyme /map=96A7-96A7 /transc=CT20638 /len=2769 /GB:AE003748 /note=3prime sequence from clone BDGP:GH21511.3prime-hit

1,6

Sorbitol dehydrogenase 1 1,6

FB:FBgn0040741 /sym=CG14486 /name= /prod= /func= /map=54E3-54E3 /transc=CT34197 /len=171 /GB:AE003802 1,6

FB:FBgn0035742 /sym=CG8629 /name= /prod=diazepam-binding inhibitor-like protein /func=enzyme inhibitor /map=65E5-65E5 /transc=CT14596 /len=255 /GB:AE003560

1,5

FB:FBgn0039529 /sym=CG5612 /name= /prod= /func= /map=98A4-98A4 /transc=CT17752 /len=1352 /GB:AE003761 1,5

FB:FBgn0036116 /sym=CG7888 /name= /prod=amino-acid permease-like /func=transporter /map=68A2-68A2 /transc=CT36915 /len=1827 /GB:AE003546 /note=3prime sequence from clone BDGP:GH09436.3prime-hit

1,5

UDP-glycosyltransferase 35a 1,5

217

Page 242: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

Lysozyme S 1,5

FB:FBgn0034638 /sym=CG10433 /name= /prod= /func= /map=57F3-57F3 /transc=CT29298 /len=1071 /GB:AE003454 /note=3prime sequence from clone BDGP:GH10517.3prime-hit

1,5

FB:FBgn0032955 /sym=CG2201 /name= /prod=choline kinase-like /func=enzyme /map=39E3-39E3 /transc=CT7236 /len=1914 /GB:AE003781 /note=3prime sequence from clone BDGP:LD18613.3prime-hit

1,5

FB:FBgn0033205 /sym=CG2064 /name= /prod= /func=enzyme /map=43E7-43E7 /transc=CT6708 /len=996 /GB:AE003840 /note=3prime sequence from clone BDGP:SD07613.3prime-hit

1,5

FB:FBgn0033683 /sym=CG18343 /name= /prod= /func= /map=48E10-48E10 /transc=CT41671 /len=463 /GB:AE003823 1,5

FB:FBgn0036262 /sym=CG6910 /name= /prod= /func= /map=68F7-68F7 /transc=CT21406 /len=1459 /GB:AE003542 1,5

hugin 1,5

FB:FBgn0034501 /sym=CG13868 /name= /prod= /func= /map=56F17-57A1 /transc=CT33390 /len=3604 /GB:AE003792 /note=3prime sequence from clone BDGP:SD03066.3prime-hit

1,5

Trypsin 1,5

/// /// Trypsin /// Trypsin 1,5

Lobe 1,5

FB:FBgn0027544 /sym=BcDNA:GH11688 /name= /prod= /func= /map=99F6-99F6 /transc=CT6804 /len=4019 /GB:AE003773 /note=3prime sequence from clone BDGP:GH11688.3prime-hit

1,5

spatzle 1,5

Ejaculatory bulb protein III /// 1,5

ABC transporter expressed in trachea 1,5

FB:FBgn0037817 /sym=Cyp12e1 /name= /prod=cytochrome P450, CYP12E1 /func=cytochrome P45 /map=86A7-86A7 /transc=CT34465 /len=1518 /GB:AE003687

1,5

FB:FBgn0032774 /sym=CG17549 /name= /prod= /func= /map=37E1-37E1 /transc=CT38763 /len=1051 /GB:AE003662 /note=3prime sequence from clone BDGP:GH19142.3prime-hit

1,5

FB:FBgn0038774 /sym=CG5023 /name= /prod=calponin-like /func=ligand binding or carrier /map=92D2-92D2 /transc=CT16114 /len=697 /GB:AE003729 /note=3prime sequence from clone BDGP:GH21596.3prime-hit

1,5

FB:FBgn0036145 /sym=CG7607 /name= /prod= /func=cell adhesion /map=68A8-68A8 /transc=CT23219 /len=660 /GB:AE003545 1,5

FB:FBgn0038160 /sym=CG9759 /name= /prod= /func= /map=87F5-87F5 /transc=CT27579 /len=685 /GB:AE003701 1,5

lethal (2) essential for life 1,5

FB:FBgn0039754 /sym=CG9747 /name= /prod= /func=enzyme /map=99E3-99E3 /transc=CT27555 /len=2124 /GB:AE003772 /note=3prime sequence from clone BDGP:GH07782.3prime-hit

1,5

FB:FBgn0038664 /sym=CG5449 /name= /prod= /func=endopeptidase /map=91E1-91E1 /transc=CT17110 /len=2563 /GB:AE003724 /note=3prime sequence from clone BDGP:GH21941.3prime-hit

1,5

FB:FBgn0037267 /sym=CG1054 /name= /prod= /func= /map=82D5-82D5 /transc=CT1108 /len=4410 /GB:AE003605 /note=3prime sequence from clone BDGP:GH25780.3prime-hit

1,5

Annexin IX 1,5

FB:FBgn0033817 /sym=CG4688 /name= /prod= /func=enzyme /map=49F15-49F15 /transc=CT15137 /len=699 /GB:AE003819 1,5

/// Fat body protein 1 1,5

cAMP-dependent protein kinase 1,5

FB:FBgn0028526 /sym=BG:DS01759.2 /name= /prod= /func= /map=34E5-34E5 /transc=CT35241 /len=1144 /GB:AE003642 1,5

Jonah 65Aiv 1,5

218

Page 243: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

FB:FBgn0039031 /sym=CG17244 /name= /prod= /func= /map=94D8-94D9 /transc=CT33347 /len=480 /GB:AE003741 1,5

FB:FBgn0030309 /sym=CG1572 /name= /prod= /func= /map=10C4-10C4 /transc=CT4080 /len=955 /GB:AE003486 /note=3prime sequence from clone BDGP:LD04844.3prime-hit

1,5

Esterase-1 1,5

FB:FBgn0035321 /sym=CG1275 /name= /prod=cytochrome b561-like /func=electron transfer /map=62D4-62D5 /transc=CT2537 /len=1681 /GB:AE003474 /note=3prime sequence from clone BDGP:LD36721.3prime-hit

1,5

FB:FBgn0032422 /sym=CG6579 /name= /prod= /func= /map=33C3-33C4 /transc=CT20347 /len=834 /GB:AE003635 1,5

FB:FBgn0033775 /sym=Cyp9h1 /name= /prod=cytochrome P450, CYP9H1 /func=cytochrome P45 /map=49D1-49D1 /transc=CT33386 /len=1557 /GB:AE003820 1,5

FB:FBgn0034098 /sym=CG15707 /name= /prod= /func=RNA binding /map=52F11-52F11 /transc=CT35928 /len=2230 /GB:AE003807 /note=3prime sequence from clone BDGP:LD30829.3prime-hit

1,5

FB:FBgn0038353 /sym=CG5399 /name= /prod= /func= /map=89A3-89A3 /transc=CT17118 /len=762 /GB:AE003710 1,5

FB:FBgn0032413 /sym=CG16997 /name= /prod= /func=endopeptidase /map=33C1-33C1 /transc=CT34780 /len=822 /GB:AE003635 1,4

FB:FBgn0035552 /sym=CG11350 /name= /prod= /func= /map=64B12-64B12 /transc=CT31662 /len=1101 /GB:AE003481 1,4

Trehalose-6-phosphate synthase 1 1,4

Drosulfakinin 1,4

longitudinals lacking 1,4

FB:FBgn0026414 /sym=Kaz1 /name= /prod= /func=serine protease inhibitor /map=61A6-61A6 /transc=CT42525 /len=626 /GB:AE003467 1,4

FB:FBgn0033600 /sym=CG9077 /name= /prod=cuticle protein /func=structural protein /map=47E1-47E1 /transc=CT26058 /len=396 /GB:AE003826 1,4

FB:FBgn0033789 /sym=CG13324 /name= /prod= /func= /map=49F1-49F1 /transc=CT32640 /len=339 /GB:AE003820 1,4

Insulin-related peptide 1,4

FB:FBgn0035187 /sym=CG9122 /name= /prod=tryptophan 5-monooxygenase /func=enzyme /map=61F3-61F3 /transc=CT9937 /len=2160 /GB:AE003470 /note=3prime sequence from clone BDGP:GH12537.3prime-hit

1,4

FB:FBgn0038721 /sym=CG16718 /name= /prod= /func= /map=92A13-92A13 /transc=CT37197 /len=4061 /GB:AE003727 /note=3prime sequence from clone BDGP:LD10322.3prime-hit

1,4

Saccharomyces cerevisiae UAS construct a of Raisin 1,4

Imaginal disc growth factor 4 1,4

FB:FBgn0031405 /sym=CG4267 /name= /prod=lipase-like /func=enzyme /map=22D3-22D3 /transc=CT11345 /len=1301 /GB:AE003583 1,4

FB:FBgn0031936 /sym=CG13794 /name= /prod= /func= /map=28C2-28C2 /transc=CT33283 /len=213 /GB:AE003618 1,4

FB:FBgn0032284 /sym=CG7294 /name= /prod= /func= /map=32A1-32A1 /transc=CT22525 /len=527 /GB:AE003629 1,4

FB:FBgn0034885 /sym=CG4019 /name= /prod=water transporter-like /func=transporter /map=59F1-59F2 /transc=CT38997 /len=1089 /GB:AE003461 1,4

FB:FBgn0038198 /sym=CG3153 /name= /prod= /func= /map=88A10-88A10 /transc=CT10556 /len=814 /GB:AE003703 1,4

FB:FBgn0033875 /sym=CG6357 /name= /prod= /func=endopeptidase /map=50C13-50C13 /transc=CT19866 /len=1418 /GB:AE003817 /note=3prime sequence from clone BDGP:GM07827.3prime-hit

1,4

/// Trypsin /// Trypsin 1,4

phantom 1,4

FB:FBgn0033384 /sym=CG8788 /name= /prod= /func= /map=45B1-45B1 /transc=CT25336 /len=1291 /GB:AE003834 1,4

antisense 1,4

Saccharomyces cerevisiae UAS construct a of Daborn 1,4

219

Page 244: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

FB:FBgn0036710 /sym=CG6479 /name= /prod= /func= /map=74B1-74B1 /transc=CT20177 /len=1858 /GB:AE003524 /note=3prime sequence from clone BDGP:GM01209.3prime-hit

1,4

HMG Coenzyme A synthase 1,4

FB:FBgn0029766 /sym=CG15784 /name= /prod= /func= /map=4F10-4F10 /transc=CT36052 /len=1665 /GB:AE003434 1,4

FB:FBgn0037128 /sym=CG14572 /name= /prod= /func= /map=79A1-79A1 /transc=CT34303 /len=774 /GB:AE003595 1,4

FB:FBgn0040718 /sym=CG15353 /name= /prod= /func= /map=22B8-22B8 /transc=CT35362 /len=162 /GB:AE003584 1,4

FB:FBgn0040718 /sym=CG15353 /name= /prod= /func= /map=22B8-22B8 /transc=CT35362 /len=162 /GB:AE003584 1,4

FB:FBgn0038878 /sym=CG3301 /name= /prod= /func=enzyme /map=93D4-93D4 /transc=CT11093 /len=913 /GB:AE003734 /note=3prime sequence from clone BDGP:GH01837.3prime-hit

1,4

FB:FBgn0037083 /sym=CG5656 /name= /prod=alkaline phosphatase-like /func=enzyme /map=78D5-78D5 /transc=CT17844 /len=1770 /GB:AE003594 /note=3prime sequence from clone BDGP:LP05865.3prime-hit

1,4

FB:FBgn0032218 /sym=CG5381 /name= /prod= /func=transcription factor /map=31D8-31D8 /transc=CT17078 /len=1884 /GB:AE003628 1,4

FB:FBgn0036394 /sym=CG9040 /name= /prod= /func= /map=70C9-70C9 /transc=CT25958 /len=677 /GB:AE003536 1,4

FB:FBgn0037807 /sym=CG6293 /name= /prod=sodium-dependent L-ascorbic acid transporter /func=transporter /map=86A2-86A2 /transc=CT19686 /len=2088 /GB:AE003686 /note=3prime sequence from clone BDGP:LD30822.3prime-hit

1,4

FB:FBgn0039073 /sym=CG4408 /name= /prod=zinc carboxypeptidase A /func=peptidase /map=94F1-94F1 /transc=CT14368 /len=1651 /GB:AE003743 /note=3prime sequence from clone BDGP:LD41739.3prime-hit

1,4

Gelsolin 1,4

FB:FBgn0039800 /sym=CG11314 /name= /prod= /func= /map=100A5-100A5 /transc=CT31575 /len=605 /GB:AE003775 1,4

FB:FBgn0034755 /sym=CG3746 /name= /prod= /func= /map=58F2-58F2 /transc=CT10065 /len=557 /GB:AE003458 1,4

insulin-like peptide 3 1,4

FB:FBgn0037239 /sym=CG11739 /name= /prod= /func= /map=82B2-82B2 /transc=CT36785 /len=1274 /GB:AE003606 1,4

FB:FBgn0038343 /sym=CG14871 /name= /prod= /func= /map=88F6-88F6 /transc=CT34690 /len=327 /GB:AE003709 1,4

BDGP:LP01187.3prime-hit /ESTpos=maps in FB:FBgn0034226 /sym=CG4837 /name= /prod=5'-nucleotidase /func=enzyme /map=54C7-54C7 /transc=CT15539 /len=414

1,4

FB:FBgn0032899 /sym=CG9338 /name= /prod= /func= /map=38F1-38F1 /transc=CT5240 /len=660 /GB:AE003668 /note=3prime sequence from clone BDGP:GH07967.3prime-hit

1,4

FB:FBgn0039102 /sym=CG16705 /name= /prod=serine protease-like /func=endopeptidase /map=95B1-95B1 /transc=CT37173 /len=1288 /GB:AE003744 /note=3prime sequence from clone BDGP:GH28857.3prime-hit

1,4

Thor 1,4

Lamin C 1,4

Cytochrome c proximal 1,4

Glutamine synthetase 2 1,4

FB:FBgn0031563 /sym=CG10031 /name= /prod=trypsin inhibitor-like /func=enzyme inhibitor /map=24A4-24A4 /transc=CT28233 /len=333 /GB:AE003579 1,4

Imaginal disc growth factor 2 1,4

Rhythmically expressed gene 5 1,4

Paxillin 1,4

Multiple drug resistance 65 1,4

FB:FBgn0029639 /sym=CG14419 /name= /prod= /func= /map=3C3-3C3 /transc=CT34076 /len=588 /GB:AE003425 1,4

220

Page 245: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

methuselah-like 4 1,4

/// Glutathione S transferase E3 1,4

FB:FBgn0032287 /sym=CG6415 /name= /prod=aminomethyltransferase /func=enzyme /map=32A2-32A2 /transc=CT20030 /len=1241 /GB:AE003629 /note=3prime sequence from clone BDGP:GH04419.3prime-hit

1,4

FB:FBgn0039464 /sym=CG6330 /name= /prod=uridine phosphorylase /func=enzyme /map=97D10-97D11 /transc=CT19812 /len=1469 /GB:AE003758 /note=3prime sequence from clone BDGP:GH09623.3prime-hit

1,4

FB:FBgn0034648 /sym=CG15675 /name= /prod= /func= /map=57F6-57F7 /transc=CT35860 /len=1440 /GB:AE003454 /note=3prime sequence from clone BDGP:GM01014.3prime-hit

1,4

Muscle LIM protein at 84B 1,4

/// 1,4

FB:FBgn0030929 /sym=CG15043 /name= /prod= /func= /map=17B4-17B4 /transc=CT34909 /len=492 /GB:AE003509 1,4

FB:FBgn0032237 /sym=CG5362 /name= /prod= /func=enzyme /map=31E1-31E1 /transc=CT17038 /len=1183 /GB:AE003628 1,4

yellow-d 1,4

FB:FBgn0036600 /sym=CG13043 /name= /prod= /func= /map=72E3-72E3 /transc=CT32262 /len=447 /GB:AE003527 1,4

Glutathione S transferase D10 1,4

Phosphoglyceromutase 1,4

subunit of type II geranylgeranyl transferase 1,4

FB:FBgn0036298 /sym=CG10627 /name= /prod= /func=phosphoacetylglucosamine mutase /map=69C4-69C4 /transc=CT13770 /len=1866 /GB:AE003541 1,3

FB:FBgn0010578 /sym=Eb1 /name= /prod= /func= /map=42C3-42C3 /transc=CT10957 /len=2073 /GB:AE003789 1,3

FB:FBgn0030040 /sym=CG15347 /name= /prod= /func= /map=7E6-7E7 /transc=CT35350 /len=824 /GB:AE003444 1,3

FB:FBgn0032318 /sym=CG14072 /name= /prod= /func= /map=32C3-32C4 /transc=CT33642 /len=366 /GB:AE003630 1,3

FB:FBgn0034757 /sym=CG13514 /name= /prod= /func= /map=58F3-58F3 /transc=CT32885 /len=1437 /GB:AE003458 1,3

FB:FBgn0036471 /sym=CG13460 /name= /prod= /func= /map=71B1-71B1 /transc=CT32823 /len=654 /GB:AE003532 1,3

FB:FBgn0038973 /sym=CG18594 /name= /prod= /func= /map=94B4-94B4 /transc=CT42527 /len=598 /GB:AE003739 1,3

FB:FBgn0039149 /sym=CG18428 /name= /prod= /func= /map=95D10-95D10 /transc=CT29930 /len=568 /GB:AE003746 1,3

FB:FBgn0040968 /sym=CG14933 /name= /prod= /func= /map=33B2-33B2 /transc=CT34761 /len=618 /GB:AE003634 1,3

FB:FBgn0035452 /sym=CG10359 /name= /prod=fibrinogen-like /func=structural protein /map=63E3-63E4 /transc=CT29060 /len=1675 /GB:AE003478 /note=3prime sequence from clone BDGP:GM10015.3prime-hit

1,3

FB:FBgn0032603 /sym=CG17928 /name= /prod=cytochrome b5-like /func=electron transfer /map=36A9-36A9 /transc=CT39948 /len=1326 /GB:AE003652 1,3

FB:FBgn0034140 /sym=CG8317 /name= /prod= /func= /map=53C7-53C7 /transc=CT24573 /len=693 /GB:AE003806 1,3

methuselah-like 3 1,3

/// 1,3

FB:FBgn0020642 /sym=Lcp65Ac /name= /prod=larval cuticle protein 65Ac /func=structural protein of larval cuticle (Drosophila) /map=65A5-65A5 /transc=CT21559 /len=330 /GB:AE003563

1,3

FB:FBgn0037288 /sym=CG14661 /name= /prod= /func= /map=82E7-82E7 /transc=CT34439 /len=618 /GB:AE003604 1,3

FB:FBgn0037552 /sym=CG7800 /name= /prod= /func=cell adhesion /map=84F5-84F5 /transc=CT23295 /len=1745 /GB:AE003678 /note=3prime sequence from clone BDGP:GH11496.3prime-hit

1,3

Larval serum protein 1 1,3

221

Page 246: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

hikaru genki 1,3

Acetyl Coenzyme A synthase 1,3

FB:FBgn0034117 /sym=CG7997 /name= /prod= /func=enzyme /map=53B1-53B1 /transc=CT24026 /len=1725 /GB:AE003807 /note=3prime sequence from clone BDGP:LD13649.3prime-hit

1,3

spire 1,3

Cytochrome P450-18a1 1,3

Actin 57B 1,3

FB:FBgn0036449 /sym=CG5295 /name= /prod= /func= /map=70F5-70F5 /transc=CT16881 /len=2429 /GB:AE003533 /note=3prime sequence from clone BDGP:LD21785.3prime-hit

1,3

FB:FBgn0037515 /sym=CG3066 /name= /prod=monophenol monooxygenase activator /func=serine carboxypeptidase /map=84D11-84D11 /transc=CT10270 /len=1830 /GB:AE003676 /note=3prime sequence from clone BDGP:SD07170.3prime-hit

1,3

Adenylate kinase-1 1,3

FB:FBgn0030251 /sym=CG2145 /name= /prod=serine proteinase /func=endopeptidase /map=10A1-10A1 /transc=CT7008 /len=2045 /GB:AE003484 /note=3prime sequence from clone BDGP:GH10845.3prime-hit

1,3

FB:FBgn0032512 /sym=CG9305 /name= /prod= /func=transcription factor /map=34B6-34B6 /transc=CT26505 /len=2150 /GB:AE003639 -1,3

GTP-binding-protein -1,3

FB:FBgn0036848 /sym=CG10424 /name= /prod= /func= /map=75F6-75F6 /transc=CT29264 /len=1194 /GB:AE003518 -1,3

optic ganglion reduced -1,3

Chip -1,3

FB:FBgn0035872 /sym=CG7185 /name= /prod=RNA binding protein-like /func=nucleic acid binding /map=66C6-66C7 /transc=CT22187 /len=2779 /GB:AE003556 /note=3prime sequence from clone BDGP:LD25239.3prime-hit

-1,3

Kruppel homolog 1 -1,3

anterior open -1,3

hedgehog -1,3

FB:FBgn0030093 /sym=CG7055 /name= /prod= /func=DNA binding /map=8C17-8D1 /transc=CT21811 /len=2652 /GB:AE003446 /note=3prime sequence from clone BDGP:LD22190.3prime-hit

-1,3

dacapo -1,3

pannier -1,3

E(spl) region transcript m4 /// /// /// -1,3

FB:FBgn0031453 /sym=CG9894 /name= /prod= /func= /map=23B1-23B1 /transc=CT9918 /len=725 /GB:AE003582 -1,3

FB:FBgn0033089 /sym=CG17266 /name= /prod=peptidylprolyl isomerase /func=chaperone /map=42B4-42B4 /transc=CT38243 /len=552 /GB:AE003789 -1,3

FB:FBgn0038583 /sym=CG7183 /name= /prod= /func= /map=90F1-90F1 /transc=CT22177 /len=2552 /GB:AE003721 /note=3prime sequence from clone BDGP:GM01008.3prime-hit

-1,3

FB:FBgn0015805 /sym=Rpd3 /name= /prod=histone deacetylase /func=histone deacetylase /map=64B17-64B17 /transc=CT1177 /len=2141 /GB:AE003482 /note=3prime sequence from clone BDGP:GM14158.3prime-hit

-1,3

FB:FBgn0034631 /sym=CG10496 /name= /prod= /func= /map=57E6-57E8 /transc=CT29466 /len=2755 /GB:AE003454 /note=3prime sequence from clone BDGP:LD41005.3prime-hit

-1,3

mitochondrial ribosomal protein L36 -1,3

methuselah-like 8 -1,3

gliolectin -1,3

222

Page 247: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

ballchen -1,3

FB:FBgn0034801 /sym=CG11716 /name= /prod= /func= /map=59B6-59B6 /transc=CT36757 /len=614 /GB:AE003459 -1,4

Saccharomyces cerevisiae UAS construct a of O'Keefe -1,4

BDGP:LD27862.3prime-hit /ESTpos=maps in FB:FBgn0037027 /sym=CG3680 /name= /prod= /func= /map=77E8-77E8 /transc=CT12245 /len=575 -1,4

brain tumor -1,4

FB:FBgn0034354 /sym=CG5224 /name= /prod= /func=enzyme /map=55D3-55D3 /transc=CT16701 /len=734 /GB:AE003799 /note=3prime sequence from clone BDGP:LD18692.3prime-hit

-1,4

FB:FBgn0034063 /sym=CG8389 /name= /prod=monocarboxylate transporter-like /func=transporter /map=52E1-52E1 /transc=CT18579 /len=1865 /GB:AE003808 /note=3prime sequence from clone BDGP:SD04201.3prime-hit

-1,4

tailless -1,4

FB:FBgn0032705 /sym=CG10346 /name= /prod= /func=chaperone /map=37A4-37A4 /transc=CT29062 /len=1986 /GB:AE003660 -1,4

FB:FBgn0038610 /sym=CG7675 /name= /prod= /func=enzyme /map=91A2-91A2 /transc=CT30419 /len=1100 /GB:AE003721 /note=3prime sequence from clone BDGP:GH26851.3prime-hit

-1,4

Kinesin-like protein at 61F -1,4

cdc2c -1,4

FB:FBgn0035235 /sym=CG7879 /name= /prod= /func=RNA binding /map=62A7-62A7 /transc=CT23834 /len=3692 /GB:AE003472 /note=3prime sequence from clone BDGP:SD09402.3prime-hit

-1,4

twins -1,4

klumpfuss -1,4

similar -1,4

FB:FBgn0029755 /sym=CG4202 /name= /prod= /func= /map=4F2-4F2 /transc=CT13852 /len=1568 /GB:AE003433 /note=3prime sequence from clone BDGP:GH08670.3prime-hit

-1,4

FB:FBgn0027949 /sym=msb1l /name= /prod= /func= /map=37F2-37F2 /transc=CT29112 /len=1340 /GB:AE003663 /note=3prime sequence from clone BDGP:LD32040.3prime-hit

-1,4

FB:FBgn0037382 /sym=CG2031 /name= /prod= /func= /map=83C3-83C3 /transc=CT6554 /len=2288 /GB:AE003601 /note=3prime sequence from clone BDGP:LD43883.3prime-hit

-1,4

muscleblind -1,4

Microtubule-associated protein 60 -1,4

DNA replication-related element factor -1,4

FB:FBgn0038666 /sym=CG5451 /name= /prod= /func=enzyme /map=91E1-91E1 /transc=CT16994 /len=1627 /GB:AE003724 -1,4

FB:FBgn0035076 /sym=CG10142 /name= /prod=peptidyl-dipeptidase (inactive) /func=peptidase /map=60E3-60E4 /transc=CT9828 /len=2078 /GB:AE003465 /note=3prime sequence from clone BDGP:GH05754.3prime-hit

-1,4

FB:FBgn0034438 /sym=CG9416 /name= /prod= /func= /map=56D3-56D3 /transc=CT26702 /len=2758 /GB:AE003796 -1,4

FB:FBgn0034939 /sym=CG2980 /name= /prod= /func=motor /map=60A13-60A13 /transc=CT40308 /len=555 /GB:AE003462 /note=3prime sequence from clone BDGP:LD21694.3prime-hit

-1,4

FB:FBgn0039130 /sym=CG5854 /name= /prod= /func= /map=95C12-95C12 /transc=CT18371 /len=1549 /GB:AE003745 /note=3prime sequence from clone BDGP:LD23561.3prime-hit

-1,4

Epidermal growth factor receptor -1,4

FB:FBgn0033367 /sym=CG8193 /name= /prod=monophenol oxidase /func=enzyme /map=45A6-45A7 /transc=CT2817 /len=2272 /GB:AE003835 -1,4

E(spl) region transcript m7 /// /// -1,4

223

Page 248: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

FB:FBgn0038321 /sym=CG6218 /name= /prod= /func= /map=88F1-88F1 /transc=CT19478 /len=1523 /GB:AE003708 /note=3prime sequence from clone BDGP:GH07590.3prime-hit

-1,4

FB:FBgn0037670 /sym=CG8436 /name= /prod= /func= /map=85D4-85D4 /transc=CT24729 /len=940 /GB:AE003682 /note=3prime sequence from clone BDGP:LD32555.3prime-hit

-1,4

broad -1,4

deltex -1,4

yemanuclein -1,4

FB:FBgn0035030 /sym=CG3541 /name= /prod= /func= /map=60D7-60D8 /transc=CT11882 /len=2417 /GB:AE003464 /note=3prime sequence from clone BDGP:GH12163.3prime-hit

-1,4

Bearded -1,5

heat shock construct of Sigrist -1,5

FB:FBgn0033182 /sym=CG1621 /name= /prod= /func=transcription factor /map=43C7-43C7 /transc=CT4338 /len=1240 /GB:AE003841 /note=3prime sequence from clone BDGP:LD05592.3prime-hit

-1,5

FB:FBgn0038476 /sym=CG5175 /name= /prod= /func=transcription factor /map=89E13-89E13 /transc=CT16563 /len=2309 /GB:AE003716 /note=3prime sequence from clone BDGP:LD09231.3prime-hit

-1,5

FB:FBgn0029515 /sym=CG3038 /name= /prod= /func= /map=1A5-1A5 /transc=CT10170 /len=1680 /GB:AE003417 /note=3prime sequence from clone BDGP:LD16783.3prime-hit

-1,5

FB:FBgn0030710 /sym=CG8924 /name= /prod= /func=transcription factor /map=13F14-13F14 /transc=CT40370 /len=2155 /GB:AE003500 /note=3prime sequence from clone BDGP:LD19131.3prime-hit

-1,5

gluon -1,5

FB:FBgn0033998 /sym=CG8092 /name= /prod= /func=nucleic acid binding /map=51E7-51E8 /transc=CT22077 /len=4271 /GB:AE003811 /note=3prime sequence from clone BDGP:LD41072.3prime-hit

-1,5

abnormal spindle -1,5

FB:FBgn0034564 /sym=CG9344 /name= /prod=LSM6; U6 snRNP core protein /func=nucleic acid binding /map=57B14-57B14 /transc=CT26549 /len=240 /GB:AE003452

-1,5

cleavage and polyadenylation specificity factor -1,5

FB:FBgn0030762 /sym=CG4453 /name= /prod=nucleoporin /func=endopeptidase /map=14F2-14F2 /transc=CT14464 /len=7261 /GB:AE003502 /note=3prime sequence from clone BDGP:LD46585.3prime-hit

-1,5

E(spl) region transcript m /// /// -1,5

glial cells missing -1,5

FB:FBgn0027528 /sym=BcDNA:LD21405 /name= /prod= /func=endopeptidase /map=15A3-15A3 /transc=CT27236 /len=3597 /GB:AE003503 /note=3prime sequence from clone BDGP:LD21405.3prime-hit

-1,5

FB:FBgn0037377 /sym=CG1218 /name= /prod= /func= /map=83C1-83C1 /transc=CT2292 /len=1479 /GB:AE003601 /note=3prime sequence from clone BDGP:LD28626.3prime-hit

-1,5

dachs -1,5

FB:FBgn0031097 /sym=CG17052 /name= /prod=peritrophin-like /func=structural protein /map=19C1-19C1 /transc=CT37860 /len=1383 /GB:AE003572 -1,5

Serine protease inhibitor 43Aa -1,5

doublesex -1,6

FB:FBgn0032693 /sym=Cyp310a1 /name= /prod=cytochrome P450, CYP310A1 /func=cytochrome P45 /map=37A3-37A3 /transc=CT29164 /len=1628 /GB:AE003659 /note=3prime sequence from clone BDGP:LD44491.3prime-hit

-1,6

BDGP:SD10469.3prime-hit /ESTpos=maps 3prime of FB:FBgn0033913 -1,6

FB:FBgn0039717 /sym=CG7854 /name= /prod= /func= /map=99C7-99C8 /transc=CT23808 /len=1888 /GB:AE003771 /note=3prime sequence from clone -1,6

224

Page 249: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

BDGP:GH10942.3prime-hit

18 wheeler -1,6

FB:FBgn0037228 /sym=CG1092 /name= /prod= /func= /map=82A4-82A4 /transc=CT1569 /len=1388 /GB:AE003607 /note=3prime sequence from clone BDGP:HL02811.3prime-hit

-1,6

grainy head -1,6

Saccharomyces cerevisiae UAS construct a of Struhl -1,6

polo -1,6

FB:FBgn0027521 /sym=BcDNA:LD21675 /name= /prod= /func= /map=14B13-14B14 /transc=CT12345 /len=2719 /GB:AE003501 /note=3prime sequence from clone BDGP:LD21675.3prime-hit

-1,6

string -1,6

wingless -1,6

Trithorax-like -1,6

FB:FBgn0029584 /sym=CG11509 /name= /prod= /func= /map=2B6-2B6 /transc=CT36379 /len=742 /GB:AE003421 /note=3prime sequence from clone BDGP:GH13132.3prime-hit

-1,6

odd paired -1,6

FB:FBgn0039229 /sym=CG6995 /name= /prod= /func=RNA binding /map=96B1-96B2 /transc=CT21632 /len=3029 /GB:AE003749 /note=3prime sequence from clone BDGP:LP03039.3prime-hit

-1,6

engrailed -1,7

FB:FBgn0039709 /sym=CG7805 /name= /prod=cadherin /func=cell adhesion /map=99C6-99C6 /transc=CT4930 /len=5083 /GB:AE003771 /note=3prime sequence from clone BDGP:LD23052.3prime-hit

-1,7

FB:FBgn0020224 /sym=Cbl /name= /prod= /func=cell cycle regulator /map=66C12-66C13 /transc=CT21779 /len=2675 /GB:AE003555 -1,7

echinoid -1,7

Ras-related protein -1,7

E(spl) region transcript m /// /// -1,7

Hemolectin -1,7

fringe -1,8

forkhead domain 96Cb -1,8

Wrinkled -2,0

BDGP:LD29477.3prime-hit /ESTpos=maps in FB:FBgn0039021 /sym=CG17138 /name= /prod= /func= /map=94C6-94C6 /transc=CT38064 /len=443 -2,0

FB:FBgn0037305 /sym=CG12173 /name= /prod= /func=protein phosphatase /map=83A1-83A1 /transc=CT8961 /len=763 /GB:AE003603 -2,0

Enhancer of split /// -2,1

FB:FBgn0031961 /sym=CG7102 /name= /prod=Sm-E snRNP core protein /func=RNA binding /map=28D5-28D5 /transc=CT21947 /len=1180 /GB:AE003619 /note=3prime sequence from clone BDGP:LD40565.3prime-hit

-2,1

FB:FBgn0038826 /sym=CG17838 /name= /prod=RNA binding protein /func=RNA binding /map=92F10-92F10 /transc=CT39628 /len=1802 /GB:AE003732 /note=3prime sequence from clone BDGP:GH28335.3prime-hit

-2,1

FB:FBgn0039704 /sym=CG7802 /name= /prod= /func= /map=99C5-99C5 /transc=CT4915 /len=2950 /GB:AE003771 /note=3prime sequence from clone BDGP:GH22837.3prime-hit

-2,1

BDGP:LD32760.3prime-hit /ESTpos=maps 3prime of FB:FBgn0039851 -2,5

FB:FBgn0039539 /sym=CG12880 /name= /prod= /func= /map=98A8-98A8 /transc=CT32024 /len=1649 /GB:AE003762 /note=3prime sequence from clone BDGP:LD29228.3prime-hit

-2,5

225

Page 250: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX I

FB:FBgn0022342 /sym=CG4844 /name= /prod= /func=ligand binding or carrier /map=54C7-54C8 /transc=CT15561 /len=1202 /GB:AE003803 /note=3prime sequence from clone BDGP:LD10058.3prime-hit

-2,5

FB:FBgn0039484 /sym=CG6124 /name= /prod= /func=cell adhesion /map=97E6-97E6 /transc=CT19227 /len=2673 /GB:AE003759 -2,9

interference Hedgehog -3,0

FB:FBgn0031956 /sym=CG7106 /name= /prod=C-type lectin-like /func=ligand binding or carrier /map=28D3-28D3 /transc=CT21973 /len=789 /GB:AE003619 -5,4

FB:FBgn0039760 /sym=CG9682 /name= /prod= /func= /map=99E5-99E5 /transc=CT27378 /len=1449 /GB:AE003773 /note=3prime sequence from clone BDGP:LP01629.3prime-hit

-6,0

Ecdysone-inducible gene E2 -11,7

226

Page 251: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

227

Appendix II- Microarays Results for expression patterning of tshNG1 neuroblasts expression comparing to wild-type

(ordered by ontology cluster)

Table II- Results of the gene ontology clusters with a P-value <0.001 in tshNG1 homozygous neuroblats microarrays after using a

cut-off of 1.3. The positive lower bound of fold change (LBFC) represents the up regulated genes while the negative LBFC represent the

downregulated genes. Only the genes with the absolute value of LBFC upper than 1.3 are represented.

Gene

LBFC

Gene Ontology "extracellular region" genes in a list with 267 annotated genes (all: 326/8391, PValue: 0.000000) ********* FB:FBgn0028542 /sym=BG:DS00180.8 /name= /prod= /func=cell adhesion /map=34E1-34E1 /transc=CT37470 /len=3394 /GB:AE003642 2,1 FB:FBgn0031097 /sym=CG17052 /name= /prod=peritrophin-like /func=structural protein /map=19C1-19C1 /transc=CT37860 /len=1383 /GB:AE003572 -1,5 Drosulfakinin 1,4 Trypsin 1,5 wingless -1,6 Lysozyme B 1,7 Lysozyme E 1,9 Lysozyme S 1,5 hedgehog -1,3 Trypsin 2,4 /// Trypsin /// Trypsin 1,4 /// /// Trypsin /// Trypsin 1,5 Drosomycin 3,3 Drosocin 2,7 Ejaculatory bulb protein III /// 1,5 Transferrin 1 1,7 Imaginal disc growth factor 4 1,4 FB:FBgn0031558 /sym=CG16704 /name= /prod= /func= /map=24A4-24A4 /transc=CT37177 /len=318 /GB:AE003579 1,9 Saccharomyces cerevisiae UAS construct a of Struhl -1,6 Immune induced molecule 23 2,5 Immune induced molecule 1 7 Odorant-binding protein 56a 3 insulin-like peptide 3 1,4

Page 252: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX II

FB:FBgn0036234 /sym=CG17824 /name= /prod=peritrophin-like /func=cell adhesion /map=68E3-68E3 /transc=CT39558 /len=2415 /GB:AE003543 1,6 Insulin-related peptide 1,4 Imaginal disc growth factor 2 1,4 Hemolectin -1,7 Larval serum protein 1 1,3 Hikaru genki 1,3 Tissue inhibitor of metalloproteases 1,9 Ecdysone-inducible gene E2 -11,7 Attacin-A 3 spatzle 1,5 Gelsolin 1,4 Gene Ontology "imaginal disc pattern formation" genes in a list with 267 annotated genes (all: 10/8391, PValue: 0.000007) ****** twins -1,4 wingless -1,6 hedgehog -1,3 Epidermal growth factor receptor -1,4 Engrailed -1,7 Gene Ontology "cell fate specification" genes in a list with 267 annotated genes (all: 19/8391, PValue: 0.000019) ***** Bearded -1,5 E(spl) region transcript m4 /// /// /// -1,3 E(spl) region transcript m /// /// -1,7 tailless -1,4 wingless -1,6 fringe -1,8 Gene Ontology "lysozyme activity" genes in a list with 267 annotated genes (all: 12/8391, PValue: 0.000021) ***** Lysozyme B 1,7 Lysozyme A /// Lysozyme C 3,1 Lysozyme A /// Lysozyme D 1,9 Lysozyme E 1,9 Lysozyme S 1,5 Gene Ontology "defense response" genes in a list with 267 annotated genes (all: 449/8391, PValue: 0.000087) ***** FB:FBgn0033367 /sym=CG8193 /name= /prod=monophenol oxidase /func=enzyme /map=45A6-45A7 /transc=CT2817 /len=2272 /GB:AE003835 -1,4 Glutathione S transferase E6 2,3 Glutathione S transferase E7 1,8 Twins -1,4 Multiple drug resistance 65 1,4 Drosomycin 3,3

228

Page 253: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX II

Drosocin 2,7 glial cells missing -1,5 Galactose-specific C-type lectin 1,7 Transferrin 1 1,7 FB:FBgn0033089 /sym=CG17266 /name= /prod=peptidylprolyl isomerase /func=chaperone /map=42B4-42B4 /transc=CT38243 /len=552 /GB:AE003789 -1,3 FB:FBgn0033817 /sym=CG4688 /name= /prod= /func=enzyme /map=49F15-49F15 /transc=CT15137 /len=699 /GB:AE003819 1,5 Immune induced molecule 23 2,5 Immune induced molecule 1 7 Glutathione S transferase D10 1,4 lethal (2) essential for life 1,5 FB:FBgn0034638 /sym=CG10433 /name= /prod= /func= /map=57F3-57F3 /transc=CT29298 /len=1071 /GB:AE003454 /note=3prime sequence from clone BDGP:GH10517.3prime-hit

1,5

FB:FBgn0037552 /sym=CG7800 /name= /prod= /func=cell adhesion /map=84F5-84F5 /transc=CT23295 /len=1745 /GB:AE003678 /note=3prime sequence from clone BDGP:GH11496.3prime-hit

1,3

FB:FBgn0038115 /sym=CG7966 /name= /prod=selenium-binding protein-like /func=ligand binding or carrier /map=87D11-87D11 /transc=CT23978 /len=1498 /GB:AE003698 /note=3prime sequence from clone BDGP:GH14316.3prime-hit

1,6

FB:FBgn0030484 /sym=CG1681 /name= /prod=glutathione transferase-like /func=enzyme /map=11F1-11F1 /transc=CT4698 /len=1098 /GB:AE003492 /note=3prime sequence from clone BDGP:GH16740.3prime-hit

1,6

Hemolectin -1,7 18 wheeler -1,6 hikaru genki 1,3 FB:FBgn0035452 /sym=CG10359 /name= /prod=fibrinogen-like /func=structural protein /map=63E3-63E4 /transc=CT29060 /len=1675 /GB:AE003478 /note=3prime sequence from clone BDGP:GM10015.3prime-hit

1,3

Thor 1,4 FB:FBgn0039644 /sym=CG11897 /name= /prod=ATP-binding cassette transporter /func=transporter /map=99A2-99A3 /transc=CT37062 /len=4458 /GB:AE003768 /note=3prime sequence from clone BDGP:LD17001.3prime-hit

1,6

FB:FBgn0034354 /sym=CG5224 /name= /prod= /func=enzyme /map=55D3-55D3 /transc=CT16701 /len=734 /GB:AE003799 /note=3prime sequence from clone BDGP:LD18692.3prime-hit

-1,4

UDP-glycosyltransferase 35a 1,5 FB:FBgn0037515 /sym=CG3066 /name= /prod=monophenol monooxygenase activator /func=serine carboxypeptidase /map=84D11-84D11 /transc=CT10270 /len=1830 /GB:AE003676 /note=3prime sequence from clone BDGP:SD07170.3prime-hit

1,3

spatzle 1,5 Gene Ontology "Notch signaling pathway" genes in a list with 267 annotated genes (all: 38/8391, PValue: 0.000165) **** Enhancer of split /// -2,1 Bearded -1,5 deltex -1,4 E(spl) region transcript m4 /// /// /// -1,3 E(spl) region transcript m /// /// -1,7 E(spl) region transcript m /// /// -1,5 Fringe -1,8 Gene Ontology "antimicrobial humoral response (sensu Protostomia)" genes in a list with 267 annotated genes (all: 20/8391, PValue:

229

Page 254: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

APPENDIX II

0.000329) **** Lysozyme B 1,7 Lysozyme A /// Lysozyme C 3,1 Lysozyme A /// Lysozyme D 1,9 Lysozyme E 1,9 Lysozyme S 1,5 Gene Ontology "chymotrypsin activity" genes in a list with 267 annotated genes (all: 182/8391, PValue: 0.000674) **** Trypsin 1,5 Trypsin 2,4 /// Trypsin /// Trypsin 1,4 /// /// Trypsin /// Trypsin 1,5 Jonah 25Bi 1,6 Jonah 25Bii 1,6 FB:FBgn0032413 /sym=CG16997 /name= /prod= /func=endopeptidase /map=33C1-33C1 /transc=CT34780 /len=822 /GB:AE003635 1,4 FB:FBgn0032638 /sym=CG6639 /name= /prod= /func=endopeptidase /map=36C2-36C2 /transc=CT20606 /len=1493 /GB:AE003655 1,6 Jonah 65Aiv 1,5 Jonah 74E 2 FB:FBgn0038113 /sym=CG11668 /name= /prod=serine protease /func=endopeptidase /map=87D11-87D11 /transc=CT36689 /len=1197 /GB:AE003698 1,8 BDGP:LD47230.3prime-hit /ESTpos=maps in FB:FBgn0034661 /sym=CG4386 /name= /prod=serine protease /func=endopeptidase /map=58A2-58A2 /transc=CT14320 /len=559 1,7 Jonah 65Aiii 1,7 FB:FBgn0039102 /sym=CG16705 /name= /prod=serine protease-like /func=endopeptidase /map=95B1-95B1 /transc=CT37173 /len=1288 /GB:AE003744 /note=3prime sequence from clone BDGP:GH28857.3prime-hit 1,4 FB:FBgn0037515 /sym=CG3066 /name= /prod=monophenol monooxygenase activator /func=serine carboxypeptidase /map=84D11-84D11 /transc=CT10270 /len=1830 /GB:AE003676 /note=3prime sequence from clone BDGP:SD07170.3prime-hit 1,3 Gene Ontology "wing disc anterior/posterior pattern formation" genes in a list with 267 annotated genes (all: 14/8391, PValue: 0.000780) **** wingless -1,6 hedgehog -1,3 engrailed -1,7 FB:FBgn0039709 /sym=CG7805 /name= /prod=cadherin /func=cell adhesion /map=99C6-99C6 /transc=CT4930 /len=5083 /GB:AE003771 /note=3prime sequence from clone BDGP:LD23052.3prime-hit -1,7

230

Page 255: Analysis of teashirt mutants affecting cell …repositorio.ul.pt/bitstream/10451/2533/1/ulsd059715_td...Objectives, Methodological Approach and Structure of the Thesis The main goal

231