the effect of serrate transmembrane domain substitution on

38
Trinity College Trinity College Digital Repository Senior eses and Projects Student Works Spring 2015 e Effect of Serrate Transmembrane Domain Substitution on Notch Signaling James Z. Curlin Trinity College, [email protected] Follow this and additional works at: hp://digitalrepository.trincoll.edu/theses Recommended Citation Curlin, James Z., "e Effect of Serrate Transmembrane Domain Substitution on Notch Signaling". Senior eses, Trinity College, Hartford, CT 2015. Trinity College Digital Repository, hp://digitalrepository.trincoll.edu/theses/482

Upload: others

Post on 16-Mar-2022

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Effect of Serrate Transmembrane Domain Substitution on

Trinity CollegeTrinity College Digital Repository

Senior Theses and Projects Student Works

Spring 2015

The Effect of Serrate Transmembrane DomainSubstitution on Notch SignalingJames Z. CurlinTrinity College, [email protected]

Follow this and additional works at: http://digitalrepository.trincoll.edu/theses

Recommended CitationCurlin, James Z., "The Effect of Serrate Transmembrane Domain Substitution on Notch Signaling". Senior Theses, Trinity College,Hartford, CT 2015.Trinity College Digital Repository, http://digitalrepository.trincoll.edu/theses/482

Page 2: The Effect of Serrate Transmembrane Domain Substitution on

TRINITY COLLEGE

THE EFFECT OF SERRATE TRANSMEMBRANE DOMAIN SUBSTITUTION ON NOTCH SIGNALING

BY

JAMES CURLIN

A THESIS SUBMITTED TO

THE FACULTY OF THE DEPARTMENT OF BIOLOGY

IN CANDIDACY FOR THE BACCALAUREATE DEGREE

WITH HONORS IN BIOLOGY

DEPARTMENT OF BIOLOGY

HARTFORD, CONNECTICUT

1 May 2015

Page 3: The Effect of Serrate Transmembrane Domain Substitution on

ii

THE EFFECT OF SERRATE TRANSMEMBRANE DOMAIN SUBSTITUTION ON NOTCH SIGNALING

BY

JAMES CURLIN

Honors Thesis Committee

Approved:

______________________________________________

Robert J. Fleming, Advisor

______________________________________________

Kent D. Dunlap

______________________________________________

Terri A. Williams

Date: ________________________________________

Page 4: The Effect of Serrate Transmembrane Domain Substitution on

iii

Table of Contents

Section Page Number

Abstract………………………………………………………………………………………..1

Introduction…………………………………………………………………………………....2

Materials and Methods……………………………………………………………………….15

Results………………………………………………………………………………………..19

Discussion……………………………………………………………………………………25

References……………………………………………………………………………………29

Acknowledgments

I would like to sincerely thank the Trinity College Biology Department for providing me the opportunity to engage in unique research over the past several years. Dr. Terri Williams and Dr. Kent Dunlap were crucial to the completion of this thesis, by providing much needed feedback and advice.

Most importantly, I would like to thank Dr. Robert Fleming. Over the past four years he has been my research advisor, and mentor throughout my college experience. He introduced me to the world of academic research, and has helped me to further my aspirations of being a scientist.

Page 5: The Effect of Serrate Transmembrane Domain Substitution on

1

Abstract

The Notch signaling pathway is a crucial means by which organisms differentiate

cells during development. Notch is regulated primarily through the interaction of a Notch

receptor protein and a ligand protein, in two specific ways. Cis-inhibition occurs when both a

ligand and receptor are present on the same cellular membrane. This results in the cis-ligand

binding to the receptor and preventing the ligand on an adjacent cell from binding and

activating the receptor. Alternatively, trans-activation occurs when the ligand and receptor

are on adjacent cells, and results in the activation of the Notch pathway. Both the receptor

and ligand proteins are transmembrane proteins that are cleaved first extracellularly by a

metalloprotease, and then intracellularly by a γ-secretase. While the cleavages in the receptor

protein have been found to be crucial for proper Notch activation, the role of the ligand

cleavages is much less well defined. Previous studies have found that the cleavage rate of the

ligand may be inversely correlated to Notch activation. We postulated that the extracellular

cleavage of Serrate, a ligand for Notch, by a metalloprotease serves as a means of inhibiting

Notch activation, possibly by affecting the endocytosis of the ligand molecule. To test this

hypothesis, we replaced the transmembrane domain of a truncated form of Serrate with that

region from non-cleavable human tyrosine kinase receptor DDR2 (discoidin domain-receptor

2), and show that preventing this cleavage is insufficient to restore Notch activation. These

findings suggest that an additional extracellular 65 amino acid segment near the

transmembrane domain may be necessary to restore wild-type levels of Notch activation.

Page 6: The Effect of Serrate Transmembrane Domain Substitution on

2

Introduction

Intercellular communication is crucial for growth and development of specific cell

types in multicellular organisms. One primary means of cell communication is through cell

signaling pathways. Individual cells signal to surrounding cells, providing them with

instructions for differentiation, growth and even apoptosis (Artavanis-Tsakonas et al., 1999).

Notch signaling is one such communication pathway and is responsible for important

developmental changes in organisms ranging from Drosophila melanogaster to Homo

sapiens. When Notch signaling is disrupted, severe developmental defects can occur. Early

studies in Drosophila showed that various Notch mutations caused hypertrophic growth of

neural tissue, accompanied by significant hypotrophy of epidermal tissue (Jimenez and

Campos-Ortega, 1982; Xu et al., 1990). This results from the Notch pathway controlling the

differentiation of cells by lateral inhibition. Neural cells normally develop in clusters: a

central cell is determined to become a neuroblast, and signals to the surrounding cells to shut

off their proneural genes, resulting in their adopting an epidermal fate (Technau et al., 1987).

Without the Notch pathway to signal to the surrounding cells, none of the proneural genes are

shut down, producing a phenotype with no epidermis and hypertrophic neural tissue (Heitzler

and Simpson, 1991). Other studies in mammalian systems showed that Notch also controls

cell differentiation in systems outside of neurogenesis, such as controlling cell fate in the

endocrine and exocrine pancreas (Apelqvist et al., 1999). Furthermore, Notch can direct cell

fate not only through promoting differentiation, but by inhibiting cell differentiation as well,

a mechanism by which Notch controls muscle cell fate (Shawber et al., 1996).

Problems resulting from mutations arising in the Notch pathway have been implicated

in a number of human diseases. Such mutations can affect the development of the liver, heart,

Page 7: The Effect of Serrate Transmembrane Domain Substitution on

3

skeleton, and kidneys. One example is Alagille syndrome, which is caused by mutations in

either the Jagged1 ligand or the Notch2 receptor (Oda et al., 1997). Another example is

Hajdu-Cheney syndrome, which is linked to a mutation in the Notch2 receptor and is

responsible for osteoporosis, renal cysts, and problems with craniofacial bone structure in the

craniofacial region (Iwaya et al., 1979). Mutations in other Notch receptors have been

implicated in cardiac disease, and cerebral autosomal dominant arteriopathy with subcortical

infarcts and leukoencephalopathy (CADASIL; Penton et al., 2012).

The Notch pathway is extremely conserved, and therefore research on Drosophila has

clinical relevance for humans, despite minor differences between the mammalian and

Drosophila Notch systems. These differences include the mammalian receptor undergoing

additional processing prior to its presentation on the cell membrane (Kopan and Ilagan, 2009;

Westhoff et al., 2009). Many features that control the Notch pathway remain unknown.

While the receptor molecule must undergo a series of proteolytic processing steps that allows

the signal to reach the nucleus, the ligands, which interact with and regulate the receptor, are

also similarly processed for a currently unknown reason. Therefore, the purpose of my

experiments in this work is to provide insight into what role ligand processing, specifically

metalloprotease cleavage has on Notch activation. This is achieved by generating a novel

form of the ligand Serrate.

Structure of Notch Receptor

Drosophila melanogaster has long been a favored research animal by geneticists for a

variety of reasons. It has a relatively short life cycle and produces many offspring in a short

period of time. Compared to many species, it also has relatively simple genetics with its

Page 8: The Effect of Serrate Transmembrane Domain Substitution on

4

genome consisting of merely four pairs of chromosomes. The Notch pathway has been

extensively studied in Drosophila, and it is the focus of this thesis.

The Notch gene product functions as a signal receptor and belongs to a group of

single-pass transmembrane proteins (Wharton et al., 1985). This receptor interacts with the

Delta/Serrate/Lag-2 (DSL) family of ligands to constitute the major interacting elements of a

cell-signaling pathway (Fehon et al., 1990). Drosophila Notch is a single protein and was

first characterized by analyzing the mRNA sequence obtained through the overlapping of

cDNA clones (Wharton et al., 1985). After determining the DNA base pair sequence of

Notch, the correct reading frame was determined and a predicted protein, of ~300 kDa in size,

was isolated. The protein indicated the presence of an extracellular domain, a transmembrane

domain and an intracellular domain.

The Notch receptor’s extracellular domain (NECD) includes 36 epidermal growth

factor (EGF)-like repeated sequences (ELRs), which play a key role in proper Notch

signaling, as well as three lin12-Notch (LNR) repeats that are also cysteine-rich (Fig. 1).

ELRs 11 and 12 play a key role in Notch’s ability to interact with both the Delta and Serrate

ligands and are sufficient to induce cells expressing Notch or Delta to aggregate (Rebay et al.,

1991). This structure-function specificity appears highly conserved, as repeats 11 and 12

serve the same role in the Xenopus Notch homolog. However, the remaining ELRs appear to

have contrasting effects in Notch-Delta and Notch-Serrate interactions in Drosophila.

Normally, the binding of the receptor and ligand surface proteins causes cells in culture to

stick together in aggregations. Removal of extraneous ELRs causes an increase in

aggregation of Notch-Delta cells, but it produces no such increase in the aggregation of

Notch-Serrate cells (Rebay et al., 1991).

Page 9: The Effect of Serrate Transmembrane Domain Substitution on

5

The ELRs play a key role in maintaining the structural integrity of the Notch

molecule. Each ELR has six highly conserved cysteines that generate disulfide bonds and

provide a highly consistent structure, even with some variation in the composition and

number of other non-cysteine amino acids in the repeat (Fig. 2). ELRs 24 through 29 have

been implicated in ligand interactions that inhibit Notch signaling (Kelley et al., 1987).

ELRs also bind calcium ions, which

influence the receptor’s affinity for the

ligand during activation and inhibition

(Cordle et al. 2008).

The Notch intracellular domain (NICD) contains several structural motifs, including

a large segment of repeating glutamines, known as OPA, for which no purpose has been

determined, 6 cdc10/ankyrin repeats (ANK), nuclear localization sequences (NLS) that

ensure the NICD goes to the nucleus, a RBPjk association module (RAM), which has a high

A A A A

C A A A A

A A

C A A A A A

A

C A

A A

A A A C

C

A A A

A A A A A A

A

A C A A A A

Figure 2: Structure of an EGF-like repeat. It

has 6 highly conserved cysteines (red) that

maintain the structure with disulfide bonds

(Yamamoto et al., 2012).

Intracellular

Extracellular

ELRs

TMD

RAM

ANK

NLS OPA PEST

LNR

Figure 1: The Notch receptor protein structure. The

extracellular domain consists of 36 ELRs and 3 LNR repeats.

The intracellular domain consists of the RAM region, 7

cdc10/ANK repeats, Nuclear localization sequences (NLS), a

polyglutamine chain called the OPA, as well as a PEST

region that controls degradation

Page 10: The Effect of Serrate Transmembrane Domain Substitution on

6

affinity for binding to various transcription factors in the nucleus and finally a PEST domain

that controls the degradation of the NICD (Struhl and Adachi, 1998; Deregowski et al., 2006,

Kopan and Ilagan, 2009). In addition to the intracellular and extracellular domains, the Notch

receptor has a transmembrane domain (TMD), which is responsible for bridging the cellular

membrane and anchoring the protein in place. Once the receptor has bound to a ligand to

activate the signaling pathway, it undergoes several cleavages that first remove the

extracellular domain and then the intracellular domain from the molecule.

Notch Ligands

The receptor alone is not capable of activating the signaling pathway and is regulated

through its interaction with the ligands, resulting in either activation or inhibition. Several

ligands of Notch exist in mammals, Drosophila and C. elegans. The best studied ligands

belong to the DSL (Delta/Serrate/LAG-2) family, (although other ligands may exist; Kopan

and Ilagan, 2009). Each ligand in this family is identified by three structural motifs, a DSL

region found at the N-terminal end of the extracellular domain, a DOS domain (Delta and

OSM-11-like proteins), and a series of ELRs, similar those found in the Notch receptor itself

(Fig. 3). While the ligand Serrate and its mammalian homolog Jagged consist of 14 and 16

ELRs respectively, and have a cysteine-rich domain, the ligand Delta (in mammals) has only

eight or nine ELRs and lacks the cysteine-rich domain (Kopan and Ilagan 2009). ELRs 4-6 of

Serrate are known as the Notch-inhibitory region (NIR) and when any single repeat is

removed the ligand loses all inhibitory function (Fig. 3A; Fleming et al., 2013). These

ligands are also transmembrane proteins and therefore have a transmembrane domain and

intracellular domain as well.

Page 11: The Effect of Serrate Transmembrane Domain Substitution on

7

Mechanism of trans-activation/cis-inhibition

The membrane bound Notch receptor interacts with its ligands in two specific ways,

trans-activation and cis-inhibition. The first ligand-receptor interaction occurs when both

proteins are present on the same cell membrane. In this case, the ligand interacts with the

receptor in a currently unknown manner and prevents the receptors from undergoing trans-

activation with any adjacent cells (Fig. 4A). This process is known as cis-inhibition

(Jacobsen et al., 1998). When the Notch receptor and a ligand are present on separate, but

adjacent cells, the ligand binds to at least ELRs 11-12 in Notch. This creates a

conformational change, revealing an extracellular metalloprotease cleavage site, and

subsequently initiating the extracellular cleavage of the Notch protein called S2 (Fig. 4B;

Mumm et al., 2000).

B

Intracellular Extracellular

DSL

DOS

Cysteine-rich

domain

NIR

A B

Figure 3: A) Structure of the Serrate ligand, characterized by a DSL domain,

14 ELRs and a cysteine-rich domain. B) The structure of the Delta ligand

consisting of the 9 ELRs and lacking a cysteine-rich domain.

Page 12: The Effect of Serrate Transmembrane Domain Substitution on

8

Proteolytic Cleavage of the Receptor Protein

The kuzbanian (kuz) gene codes for an ADAM (a disintegrin and metalloprotease)

protein that is responsible for the extracellular cleavage in Notch receptor processing prior to

signal transduction. This protein has a conserved zinc-binding region as well as several

cysteines that allow the binding of integrin receptors. The kuz protein shares a close

homology with a mammalian homolog, BMP (bovine metalloprotease) and other ADAM

proteins cause proteolytic cleavages to Notch as well, such as TACE (TNF-alpha converting

enzyme), which uses a proteolytic cleavage to release TNF-α from its transmembrane domain

(Brou et al., 2000). In Notch, this cleavage allows for the shedding of the extracellular

Intracellular

Extracellular

Intracellular

Extracellular

S3

S2

A B

Figure 4: Regulation of Notch pathway through ligand-receptor interactions. A) Cis-inhibition: The receptor and ligand are

present on the same cell surface, so the receptor is prevented from interacting with the adjacent ligand. B) Trans-activation:

The Notch receptor binds to the ligand, which triggers a conformational shift, allowing the receptor to be cleaved and

activating the signal

Page 13: The Effect of Serrate Transmembrane Domain Substitution on

9

domain, which subsequently leads to another cleavage of Notch by a γ-secretase (Mumm et

al., 2000).

After the extracellular cleavage of Notch by ADAM, the NECD is endocytosed by the

ligand-expressing cell (Shimizu et al., 2002). This allows room for a presinilin/γ-secretase,

which may normally be blocked by the presence of the extracellular domain, to interact with

and cleave the intracellular domain of Notch at the S3 site (Fig. 4B). This initiates

transcription regulation by releasing the intracellular domain into the cell.

Following the intracellular S3 cleavage by γ-secretase, the Notch intracellular domain

(NICD) moves to the nucleus and regulates transcription through a series of nuclear

intermediates. One such nuclear factor is the Suppressor of Hairless (Su(H)), which requires

the ankyrin (ANK) repeats of the NICD to be sequestered in the cytoplasm (Fortini and

Artavanis, 1994). Once the Notch receptor is bound in trans to the ligand, Su(H) is

translocated into the nucleus. The ANK repeats then interact with the Su(H) protein to recruit

Mastermind, a coactivator necessary for recruiting a transcription activation complex (Kopan

and Ilagan 2009).

Ligand Processing

Protein cleavages are not only necessary for the receptor to function properly, but are

crucial for the ligand as well. The ligands Serrate and Delta are cleaved extracellularly by

metalloproteases produced by kuzbanian or TACE (Lavoe and Selkoe, 2003;Qi et al., 1999).

However, when secreted forms of these ligands were expressed, that are comparable to forms

that result from ADAM cleavages, they generated a phenotype similar to loss of function

Notch mutation (Sun and Artavanis-Tsakonas, 1997). It is postulated that the ligands must

Page 14: The Effect of Serrate Transmembrane Domain Substitution on

10

undergo a special endocytosis to activate Notch (Parks et al., 2000). After binding to the

receptor protein, the ligand is then tagged with ubiquitin (Deblandre et al., 2001). This allows

epsin to trigger the endocytosis of the ligand, which tugs on the NECD to reveal a

metalloprotease cleavage site (Wang and Struhl, 2004). Given the Parks model of Notch

activation (2000), cleaving the extracellular domain of the ligand might reduce its ability to

cause a conformation change in the receptor, hence antagonizing Notch activation and

emphasizes the sequence of events necessary for activation.

Effect of Altering the Ligand Protein Structure

Altering the structure of Serrate can have a significant impact on the ligand’s ability

to activate Notch. The Serrate construct Minigene is a truncated form of the ligand that only

contains the signal peptide, DSL domain, and the first six ELRs in the extracellular domain

(Fig. 5B). This construct was unable to activate Notch when ectopically expressed (Fig. 6E;

Fleming, personal communication). However, adding a 65 amino acid sequence to the

extracellular domain of Serrate restored its trans-activation capabilities (Fig. 6K).

Intracellular Extracellular

DSL

Cysteine-

rich

domain

NIR

DDR

A B C D

ELRs

Figure 5: Variations of the Serrate molecule. A) Wild-type Serrate. B) Minigene Serrate construct with a truncated

extracellular domain containing the first 6 ELRs, and a small amino acid chain directly adjacent to the

transmembrane domain. C) DDR2 Serrate construct containing the minigene construct with a DDR2

transmembrane domain substituted in. D) DDR265 Serrate construct containing the DDR2 construct with an

additional 65 amino acids added immediately adjacent to the transmembrane domain.

Page 15: The Effect of Serrate Transmembrane Domain Substitution on

11

Figure 6: Previous Serrate constructs and their ability to activate and inhibit Notch. A, D, G, and J) show ectopically

expressed ptc-Serrate using GFP in several different constructs. B, E, H, and K) Cut expression of Notch activation

in various Serrate constructs. C, F, I, L) merged images of the other images. B) Trans-activation and cis-inhibition of

Notch in ectopically expressed wild-type Serrate. The white line indicates cut expression when no ectopic Serrate is

present. E) Minigene construct, a truncated Serrate that only has the first 6 ELRs, only produces cis-inhibition, and

no trans-activation of Notch. H) Ncleave7, a construct that changed one amino acid in the metalloprotease cleavage

site, produces excess Notch activation. K) MG65, a construct that includes minigene and 65 additional amino acids,

produces wild-type trans-activation and cis-inhibition.

G H I Ncleave7

Page 16: The Effect of Serrate Transmembrane Domain Substitution on

12

Figure 7: Western Blot showing the cleavage rate of minigene (MG) and MG65. The extracellular domain of the

minigene is significantly more abundant than that of the MG65, indicating that MG is cleaved at a much higher rate

than MG65 (Fleming, personal communication). The MG65m lane shows the cleavage rate of a mutated MG65

construct.

Furthermore, the minigene construct has also been shown to cleave at a significantly

higher rate than MG65 (Fig. 7) which indicates that a correlation between the rate of

cleavage and Notch activation capacity may exist. The first attempt to test this correlation

resulted in the creation of the Ncleave7 construct, which substituted two amino acids in the

metalloprotease cleavage site, in an effort to block the cleavage from occurring. While

Ncleave7 did produce an over activation of Notch, (Fig. 6H) it was later determined that the

cleavage was not completely prevented (Fleming, personal communication).

MG MG65

Cell lysate

Medium faction

Medium fraction (concentrated)

MG65m MG MG65 MG65m MG MG65 MG65m

MGECD

anti-HA

MG65ECD

, MG65mutECD

Page 17: The Effect of Serrate Transmembrane Domain Substitution on

13

Goals of the project

As stated previously, while the cleavages of the Notch receptor serve the clear

purpose of facilitating the release of the NICD, the purpose of cleaving the ligands is still

unknown. Based on the initial evidence provided by previous constructs (Fig. 6, 7), we

theorized that the cleavage of the ligand Serrate functions as a down regulator of the Notch

signal. To study this phenomenon, I sought to interrupt the metalloprotease cleavage of

Serrate. Discoidin domain receptors (DDR) are a group of transmembrane tyrosine kinase

proteins that control the interactions between cells and collagen. While DDR1 has been

shown to be cleaved by metalloproteases such as membrane-type matrix metalloproteinase

(MT-MMP) and release an extracellular domain in a manner similar to Serrate and Notch,

the transmembrane domain of DDR2 undergoes no such cleavage by a metalloprotease (Fu et

al., 2013). The cDNA encoding the transmembrane and juxtamembrane regions of the DDR2

was used to replace the comparable region of Serrate to eliminate metalloproteinase cleavage.

The first goal of this project was the generation of a Serrate minigene molecule, with the

transmembrane domain substituted for the theoretically uncleavable DDR2 (discoidin

domain receptor) transmembrane domain from humans. Theoretically, if the cleavage of the

ligand is stopped, then the ligand will always trigger the conformational change, and Notch

will activate at a much higher rate.

In order to observe the effect of ectopically expressed DDR2 Serrate on Notch, and

consequently its effect on wing development in D. melanogaster, the construct is linked to

the patched (ptc) promoter and the Gal4/UAS expression system with green fluorescence

protein (GFP) acting as the reporter gene (Brand and Perrimon 1993). This allows the

expression of Serrate to occur ectopically in a known pattern and can be measured using the

Page 18: The Effect of Serrate Transmembrane Domain Substitution on

14

GFP. The second goal was to create a construct similar to DDR2, but with the Serrate MG65

molecule, rather than the minigene as the basis for the construct. If the new construct results

in an overexpression of Notch, then it is very clear that some portion of the 65 additional

amino acids interacts in a presently unknown manner with the Notch receptor, and would

further the hypothesis that the cleavage acts to down regulate Notch.

Page 19: The Effect of Serrate Transmembrane Domain Substitution on

15

Materials and Methods

Creation of Serrate Construct DDR2

The initial DNA used was the full-length Serrate Bsp tom DNA in pUAST attB

vector (Biscof, et al. 2007), and is called Xho+. Xho+ was initially cut with the restriction

digest enzymes XhoI and XbaI (NEB) simultaneously. The DNA from pUAST attB Xho+ #4

(Fleming, personal communication) was used as a control. For all digests, a lambda/Hind III

ladder (NEB) was used for fragment size estimations. Inoculations were made of the DDR2

construct, which was initially synthesized and cloned by InVitrogen. These inoculations were

purified according to the same procedure mentioned previously. Each inoculation was

analyzed using Nanodrop to determine the concentration of the DNA.

Using gel electrophoresis, the Xho+ digest was separated out on using the Extraction

Kit procedure (www.qiagen.com). A ligation of the two DNA fragments was performed

using Thermo Quick Ligase according to the Rapid DNA Ligation Kit procedure (Thermo

Scientific). The ligated DNA (XhoDDR2) was transformed into NEB 5-alpha competent E.

coli (NEB) according to the manufacturer’s protocol and plated in varying quantities onto

LB/AMP agar plates. PCR was used to determine if the insert was correct with the primer

Xho3 end (5’GCGTCTGGCTTATCGCTCGAGTTCGGGAATGAACTTAAC3’) and primer

5119-5080 (5’CACACTGTGTAGGATGTTCTTAGCGAAGAGAAGAGT3’), with Bsp tom used

as a control. In order to test the orientation of the insert, both samples were digested

sequentially with first XbaI and then EcoR1.

Both XhoDDR2 and an additional sample of Xho+ #4 were digested sequentially with

XbaI and EcoR1, and the lowest band from both digests were isolated and purified according

to the QIAquick Gel Extraction Kit procedure (www.qiagen.com) over a single column.

Page 20: The Effect of Serrate Transmembrane Domain Substitution on

16

These fragments were ligated with a pUAST attB vector that had been cut with EcoR1, using

the ligation procedure (Thermo Scientific). NEB 5-alpha competent E. coli (NEB) was used

for the transformation, and the cells were plated on LB/AMP agar plates in varying quantities.

Each sample was prepped for sequencing first by increasing the concentration of each

sample by using vacuum dehydration (Savant SpeedVac Plus SC110A). One microgram of

DNA was resuspended, and the primer 4336

(5’GCCGGTGGCCGCTCTTAGGAACTGGTGCTGCC3’) was added to the solution. These

samples were sequenced by Genewiz through the University of Connecticut Molecular Core

Facility. The final DNA construct was sent to Genetic Services, Inc. where it was injected

into Drosophila embryos to generate transgenic lines.

Drosophila crosses used for phenotypic observations

To determine the phenotypic effects of the constructs, several crosses involving the

injected flies were necessary. Virgin females were collected from the promoter constructs

H2S2/H2S2 or ptc;Gal4 UAS GFP lines. These females were crossed separately with males of

the DDR2/attp2 homozygous lines. The H2S2/H2S2 cross was placed in 25°C incubation,

while the ptc;Gal4 UAS GFP cross was placed in 18°C to slow development to avoid

embryonic lethality associated with high levels of expression. After 3 days, the adults were

moved to fresh tubes of food in to grow the fly lines. This process was repeated several times

until sufficient animals were obtained.

Wing Dissection

As the adult Drosophila from the H2S2/H2S2 cross emerged from their pupal cases, the

wings were removed from adult flies and stored in xylene. Approximately ten wings were

Page 21: The Effect of Serrate Transmembrane Domain Substitution on

17

placed on a microscope slide, and excess xylene was removed. Permount (Fisher Scientific)

was used to mount the wings, and a coverslip was placed on the slide. This was allowed to

dry for several hours.

Immunofluorescence Staining

Larvae from the ptc;Gal4 UAS GFP cross were dissected in phosphate buffered saline

(PBS), and the imaginal wing discs, while still attached to the larval heads, were placed in

4% paraformaldehyde in PBS for approximately 20 minutes. The heads were washed with

PBS four times. At this point, the PBS was replaced with blocking solution (1X PBS, 0.002%

Saponin and 0.003% Normal Goat Serum), and the wing discs were kept at a cool

temperature for 2 hours. The blocking solution was replaced, and 15µL of primary antibody

mouse anti-cut 2B10 (Developmental Studies Hybridoma Bank) was added, and incubated

overnight. The following day, primary antibody solution was removed and the wing discs

were washed with 4 changes of PBS, and placed in fresh blocking solution. After several

hours, the blocking solution was replaced with 1 µL of the secondary antibody Alexa Fluor

546 goat anti-mouse IgG and allowed to incubate overnight. The following day, heads were

again washed with PBS 4x, and placed in fresh blocking solution. The wing discs were

mounted on microscope slides, and were isolated from the larval heads. Excess solution was

removed, and the discs were covered in glycerol. The wing discs were coverslipped and

observed using a Nikon E600 epi- fluorescent microscope and Spot Diagnostics Software.

Page 22: The Effect of Serrate Transmembrane Domain Substitution on

18

Figure 8: A simplified diagram showing the restriction digests and ligations necessary to generate the

SerDDR2 construct. A) Xho+ Bsp tom was digested with XbaI/XhoI and the smaller fragments were

removed. B) DDR2 synthesized DNA digested with XbaI/XhoI and the smaller fragment was kept. C) The

result of ligating A and B, this was digested with EcoR1/XbaI. D) Same DNA from A, digested with

EcoR1/XbaI and the Xba fragment removed E) Bsp tom digested with EcoR1. F) The result of ligating C,

D, and E together.

Xho+ Bsp tom

in pUAST attB

EcoR1

XbaI

XbaI

XhoI EcoR1

DDR2 in

XbaI XhoI

XhoDDR2 in

pUAST

EcoR1 XbaI

XhoI

DDR2

XbaI XbaI

XhoI XbaI

Final SerDDR2

construct

EcoR1

XbaI

XhoI EcoR1

Bsp tom Xho+

5’ fragment

XhoDDR2

3’ fragment

Bsp tom in pUAST

attB

EcoR1 Xho+ Bsp tom

in pUAST attB

EcoR1

XbaI

XbaI

XbaI XbaI

A B

C

D

E

F

Page 23: The Effect of Serrate Transmembrane Domain Substitution on

19

Results

Construction of SerDDR2

The putatively uncleavable construct SerDDR2 was successfully created by replacing

the transmembrane domain of the minigene Serrate with the transmembrane domain of the

human DDR2 protein. Figure 8 shows the fragments produced by the XhoI/XbaI digest of the

DDR2 inserted piece and the Xho+ vector. The smaller of two fragments of DDR2 (Fig. 9,

lane 1) was isolated from the gel and purified, as was the larger fragment from Bsp tom Xho+

(Fig. 9, lane 4).

Figure 9: The gel electrophoresis of the XhoI/XbaI restriction digest. Lane 1. DDR2 cut with XhoI/XbaI.

The lower band was extracted and purified for further use (circled). Lane 2. Uncut DDR2 used as a

control. Lane 3. HindIII lambda ladder used for fragment size determination. Lane 4. Xho+ cut with

XhoI/XbaI. The upper band was extracted and purified for further use (circled). Lane 5. Uncut Xho+

used as a control.

3 1 2 4 5

Page 24: The Effect of Serrate Transmembrane Domain Substitution on

20

Following the successful ligation of the two fragments from the XhoI/XbaI digest (Fig.

8C), the XhoDDR2 construct was subsequently digested by EcoR1/XbaI, as was Bsp tom

Xho+. Figure 10 shows the resulting fragments produced by the digest, excluding Xho+. The

lower band (Fig. 10, Lane 1) was extracted and purified. This fragment was then ligated with

the fragment obtained from Xho+, as well as Bsp tom attB that had been digested with EcoR1

alone (Fig 8C-F).

Figure 10: An EcoR1/XbaI restriction digest used as a test for two different samples of XhoDDR2. Lane 1.

The first sample of XhoDDR2 with the desired fragment present (circled). Lane 2. The second sample of

XhoDDR2 lacking the desired fragment. Lane 3. MG65 DNA also digested with EcoR1/XbaI to serve as a

control. Lane 4. HindIII lambda ladder for fragment size comparison.

Several colonies resulted from the previously mentioned ligation, so a 50-colony PCR

was performed (Figure 11). Many samples showed the presence of the correct insertion band,

4 3 2 1

Page 25: The Effect of Serrate Transmembrane Domain Substitution on

21

and several were used for further testing (Lanes 1, 2, 3, 6, 9, 10, 14 and 15). Ultimately,

samples from lanes 3 and 6 were sent for DNA sequencing and verification, and both

appeared to have the correct sequence.

1 2 3 4 5 6 7 8 9 10 11 12 13

14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30

Figure 11: 50-colony PCR of the SerDDR2 construct following the final ligation. HA3

primer and 5119-5080 primer were used, and HAMG DNA was used as a control. Lanes

with the desired band (such as the one circled) indicate the correct ligation. Each of these

was later tested for the orientation of the insert. Lanes 3, 6, and 15 were tested and all had

the insert in the correct orientation. The samples from lanes 3 and 6 were sent for

sequencing.

30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50

Page 26: The Effect of Serrate Transmembrane Domain Substitution on

22

Figure 12: Imaginal wing discs of the H2S2/ H2S2 and ptc; Gal4 UAS GFP cross. Dorsal is up and posterior

is to the right in each panel. A) A wild-type wing disc showing ectopic Serrate expression under the

patched (ptc) promoter using green fluorescence protein (GFP) (Fleming, personal communication). B)

CUT expression of Notch activation affected by ectopic Serrate in a wild-type disc. Cut is expressed

adjacent to Serrate expression in ventral disc areas (red stripes). Notch is not activated where Serrate is

expressed at high levels, as seen by the gap in CUT expression. C) Merged image of A and B. D) ptc-GFP

expression of Serrate in the DDR2 construct. E) CUT expression of Notch activation in the DDR2

construct. Gap in CUT expression across the margin is present. F) Merged image of D and E

Upon completion of the SerDDR2 construct, genetic crosses were performed to

express the transgene in a pattern driven by the promoter of the gene patched (ptc). The

imaginal wing discs were collected. The wing discs taken from the H2S2/ H2S2 and ptc;Gal4

DDR2

Merge

E F

D

Page 27: The Effect of Serrate Transmembrane Domain Substitution on

23

UAS cross were processed for Cut expression as a marker for Notch activity and observed

(Fig. 12).

When DDR2attp2 is expressed via ptc, strong cis-inhibition is observed (Fig. 12E).

Occasionally, a very small amount of activation of Notch occurs on the posterior side of the

patched Serrate expression pattern, in some discs (Fig. 12E,F). However, this activation is

extremely limited compared to wild-type activation of Notch (Fig. 12B, C), and many discs

showed no activation at all.

The H2S2/ H2S2 and DDR/2attp2; A5/TM6 B cross produced several different

phenotypes (Fig. 13). All flies developed wings, but the vast majority of flies produced

dominant-negative wings, a scenario in which the altered copy of Serrate inhibits the wild-

type copy. These wings display severely stunted growth and heavy wing veination (Fig. 13G,

H). A smaller percentage of animals showed wings with severely nicked edges, similar to the

phenotype produced by Serrate mutants (Fig. 12E and F).

Construction of SerDDR265 is currently in progress, following the same procedures

used for the construction of SerDDR2. However, this construct uses MG65 as its framework

rather than the minigene, and therefore contains the additional 65 amino acids that were

shown to restore Notch activation (Fig. 6K). This should allow us to determine if the

cleavage of Serrate has an effect on Notch activation independent of the effect of the

additional amino acids.

Page 28: The Effect of Serrate Transmembrane Domain Substitution on

24

Figure 13: Wings collected from the H2S2/ H2S2; DDR/2attp2 cross with ptc expressed Serrate. A) Wild-

type adult wing that is phenotypically normal. B) Wild-type wing with a wild type Serrate construct

expressed by H2S2. C) Wing expressing activated Serrate construct via H2S2. D) Wing expressing a

dominant-negative Serrate construct via H2S2 E), F) Adult wings expressing DDR2/attp2 via H2S2

displaying severe wing nicking, but still somewhat developed. G), H) Adult wings expressing DDR2/attp2

via H2S2 displaying the dominant-negative phenotype with severely reduced wing size and inhibited

development. All images are at same magnification.

A

B

C

D H

E

G

F

Page 29: The Effect of Serrate Transmembrane Domain Substitution on

25

Discussion

The results produced by the SerDDR2 construct, as shown in the adult wings and

imaginal wing discs (Fig. 11,12), do not support the hypothesis that the cleavage of Serrate

functions as a down regulator of Notch activation. Previous studies have shown that forms of

Serrate such as the minigene construct, which are cleaved at a significantly higher rate than

wild-type Serrate, also lack activation capacity. However, Serrate constructs such as MG65,

which are cleaved at levels significantly lower than the minigene (Fig. 7), are capable of

activating Notch at levels equivalent to wild-type as well (Fleming, personal communication).

Therefore, it appears that the cleavage rate of Serrate may be inversely correlated to the rate

of Notch activation.

I predicted that preventing the cleavage of Serrate would increase activation of Notch

drastically. Hence a phenotype in wing discs comparable to that seen in Figure 6H was

expected of the SerDDR2 construct. On the contrary, the SerDDR2 construct resulted in

primarily Notch inhibition with perhaps an extremely small amount of Notch activation in

some discs. This could be explained by several different possibilities as outlined below.

The MG65 Serrate construct differs from the minigene construct by the presence of

an additional endogenous 65 amino acids chain located adjacent to the transmembrane

domain. This additional chain of amino acids has very little homology with the same region

in the mammalian homologous ligand Jagged (Lindsell et al., 1995), and no additional

functions have been attributed to this region so far (Fleming, personal communication).

However, the additional 65 amino acids allow MG65 to activate Notch at a significantly

higher rate than the minigene construct. This suggests that a previously undescribed

Page 30: The Effect of Serrate Transmembrane Domain Substitution on

26

component in these 65 amino acids contributes to proper activation of Notch. Part of the

reason for constructing DDR265 is to further examine whether that additional chain can

permit Notch activation, in the absence of a metalloprotease cleavage in the DDR2 construct.

It is possible that the cleavage does, in fact, down regulate Notch activation, but that

activation itself requires an additional component not present in DDR2. Without this

unknown component, Notch may not activate at significant levels regardless of ligand

cleavage. This could explain the largely dominant-negative effects observed in the adult

wings (Fig. 13G, H) when DDR2attP2 is expressed with an occasional hint of activity in

some wings (Fig. 13E, F).

Most adult wings observed from the SerDDR2 construct produced the dominant-

negative phenotype (Fig. 12G, H). This is consistent with adult wing phenotype observed

previously in the minigene construct (Fleming, personal communication), indicating that

Notch activation does not occur, or occurs in very small amounts. However, several

individual flies produced wings that were much more developed, and thus do not display as

strong of a dominant-negative phenotype, though these flies were not nearly as abundant as

the other phenotype. Wings that were not dominant-negative were characterized by severe

wing nicking, a trait often present in flies with reduced Notch activation (Fleming, personal

communication; Thomas et al., 1991). This suggests that in some SerDDR2 flies, Notch

activation was partially restored. Unfortunately, because this phenotype is not present in all

offspring, it seems unlikely that it is solely the result of the SerDDR2 construct. Ultimately,

the cause of the unexpected phenotype is still unknown.

Another factor that could be influencing the results is the substituted SerDDR2

transmembrane domain itself. While it has been shown to not be cleaved in humans, its

Page 31: The Effect of Serrate Transmembrane Domain Substitution on

27

cleavage or non-cleavage has not yet been verified in Drosophila (Fu et al., 2013). Many

components that control metalloprotease cleavage have not yet been fully described, and it is

possible that some of these could initiate a cleavage of the DDR2 transmembrane domain in

flies in a manner that has not been observed in humans. If so, the SerDDR2 construct could

still be getting cleaved, which would then down regulate Notch. It will be necessary to

measure the abundance of cleaved material using a western blot to rule out this scenario as an

explanation. Consistent with this explanation are the properties of metalloproteases

themselves.

Metalloprotease cleavage sites are shown to be more “fluid” than many types of

proteases because they are less sequence specific (Fu et al., 2013). Despite altering an amino

acid within the cleavage site of the Serrate construct Ncleave7 and having this result in

excessive Notch activation when expressed, the protein was still capable of being cleaved

(Fig. 7; Fleming, personal communication). Therefore, it is possible that the SerDDR2

construct is still cleaved, despite the complete absence of the metalloprotease cleavage site

used in humans.

Construction of SerDDR265 is still in progress due to a number of technical problems,

specifically issues relating to ligations. Once it is completed, the results should provide

insight into the role of ligand cleavage on Notch activation. If the additional 65 amino acids

present in the MG65 construct that restore its ability to activate Notch similar to wild type

Serrate are important for its activity, then I predict that the SerDDR265 construct will over-

activate Notch (Fig. 14). This is based on the idea that the cleavage functions as a down

regulator, because SerDDR265 would essentially be an uncleavable form of MG65. However,

Page 32: The Effect of Serrate Transmembrane Domain Substitution on

28

if SerDDR265 does not over activate Notch, then it is possible that the cleavage itself might

be required for Notch activation, rather than serving as a down regulator.

For further study, a control construct that contains an additional 65 amino acids that

are not native to Serrate would help confirm if some factor in that region plays a key role in

Notch activation. Otherwise, the ligand may rely on a spacing mechanism to activate the

receptor. If an unknown factor does exist, then it could revolutionize the current

understanding of the ligand’s role in Notch.

DDR

B

C

Figure 14: Variations of the Serrate molecule, cleavage rate and their effect on Notch. A) Ser minigene, a

truncated form that displayed an increased cleavage rate and no Notch activation. B) DDR2 Serrate

construct containing the minigene construct with a DDR2 transmembrane domain substituted in. It was not

cleaved and produced no Notch activation C) DDR265 Serrate construct containing the DDR2 construct with

an additional 65 amino acids added immediately adjacent to the transmembrane domain. This construct is

not cleaved and should display increased Notch activation. D) Ser MG65, the minigene construct with an

additional 65 amino acids attached. Displayed cleavage rates and Notch activation.

A

D

Cleavage: Increase Notch activation: None

Cleavage: None Notch activation: Increase?

Cleavage: Normal Notch activation: Normal

Cleavage: None Notch activation: None

Page 33: The Effect of Serrate Transmembrane Domain Substitution on

29

Literature Cited

Apelqvist, Å., Li, H., Sommer, L., Beatus, P., Anderson, D., Honjo, T., Hrab de Angelis, M.,

Lendahl, U., and Edlund, H., 1999. Notch signalling controls pancreatic cell

differentiation. Nature 400.6747: 877-881.

Artavanis-Tsakonas, S., Rand, M. D., & Lake, R. J. 1999. Notch signaling: cell fate control

and signal integration in development. Science, 284(5415), 770-776.

Bischof, J., Maeda, R., Hediger, M., Karch, F., and Basler, K.. An Optimized Transgenesis

System for Drosophila Using Germ-line-specific φC31 Integrases. 2007. Procedings

of the National Academy of Sciences of the United States of America 104.9: 3312-

317.

Blaumeuller, C. M. , Qi, H., Zagouras, P., and Artavanis-Tsakonas, S. 1997. Intracellular

cleavage of Notch leads to a heterodimeric receptor on the plasma membrane. Cell

90: 281-291.

Brand, A. H., Perrimon, N. 1993. Targeted gene expression as a means of altering cell fates

and generating dominant phenotypes. Development (Cambridge, England) 118 (2):

401–415

Brou, C., Logeat, F., Gupta, N., Bessia, C., LeBail, O., Doedens, J. R., Cumano, A., Roux, P.,

Black, R. A., and Israel, A. 2000. A novel proteolytic cleavage involved in Notch

signaling: The role of the disintegrin-metalloprotease TACE. Mol. Cell 5: 207-216.

Page 34: The Effect of Serrate Transmembrane Domain Substitution on

30

Cordle, J., Johnson, S., Tay, J.Z., Roversi, P., Wilkin, M.B., de Madrid, B.H., Shimizu, H.,

Jensen, S., Whiteman, P., Jin, B., et al. 2008. A conserved face of the Jagged/Serrate

DSL domain is involved in Notch trans-activation and cis-inhibition. Nat. Struct. Mol.

Biol. 15, 849–857.

Deblandre, G.A., Lai, E.C., and Kintner, C. 2001. Xenopus neuralized is a ubiquitin ligase

that interacts with XDelta1 and regulates Notch signaling. Dev Cell 1: 795-806

Deregowski, V., Gazzerro, E., Priest, L., Rydziel, S., and Canalis E. 2006. Role of the Ram

Domain and Ankyrin Repeats on Notch Signaling and Activity in Cells of

Osteoblastic Lineage. Journal of Bone and Mineral Research 21.8: 1317-326.

Fehon, R.G., Kooh, P.J., Rebay, I., Regan, C., Xu, T., Muskavitch, M.A.T., and Artavanis-

Tsakonas, S. 1990. Molecular Interactions between the protein products of the

neurogenic loci Notch and Delta, two EGF-homologous genes in Drosophila. Cell 61:

523-534

Fleming, R. J., Hori, K., Sen, A., Filloramo, G., Langer, J., Obar, R., Artavanis- Tsakonas,

S., and Maharaj-Best, A. 2013. A. An extracellular region of Serrate is essential for

ligand-induced cis-inhibition of Notch signaling. Development 140.9:2039-2049.

Fortini, M. E. and Artavanis-Tsakonas, S. 1994. The Suppressor of Hairless protein

participates in Notch receptor signaling. Cell 79: 273-282.

Fu Hsueh-Liang, Valiathan R., Arkwright R, Sohail A, Mihai C, Kumarasiri M, Mahasenan

K, Mobashery S, Huang P, Agarwal G,et al. 2013. Discoidin Domain Receptors:

Unique Receptor Tyrosine Kinases in Collagen-mediated Signaling. The Journal of

Biological Chemistry. 288(11):7430-7437

Page 35: The Effect of Serrate Transmembrane Domain Substitution on

31

Heitzler, P. and Simpson, P. 1991. The choice of cell fate in the epidermis of Drosophila.

Cell 64, 1083-1092

Iwaya, Tsutomu, Kazuhiko Taniguchi, Junzo Watanabe, Kazuso Iinuma, Yutaka Hamazaki,

and Seizo Yoshikawa. 1979. Hajdu-Cheney Syndrome. Archives of Othorpaedic and

Traumatic Surgery: 293-302

Jacobsen, T. L., Brennan, K., Arias, A. M., & Muskavitch, M. A. (1998). Cis-interactions

between Delta and Notch modulate neurogenic signalling in Drosophila.

Development, 125(22), 4531-4540.

Jiménez, F. and Campos-Ortega, J.A. 1982. Maternal effects of zygotic mutants affecting

early neurogenesis in Drosophila. Roux’s Arch Dev. Biol. 191: 191-201

Kelley, M. R., Kidd, S., Deutsch, W. A., and Young, M. W. 1987. Mutations altering the

structure of epidermal growth factor-like coding sequences at the Drosophila Notch

locus. Cell 51: 539-548.

Kidd, S., & Lieber, T. 2002. Furin cleavage is not a requirement for Drosophila Notch

function. Mechanisms of Development, 115(1), 41-51

Kopan, R. and Ilagen, M. X. G. 2009. The canonical Notch signaling pathway: Unfolding the

activation mechanism. Cell 137:216-233.

LaVoie, M. J. and Selkoe, D. S. 2003. The Notch ligands, Jagged and Delta, are sequentially

processed by α-secretase and Presenilin/γ-secretase and release signaling fragments.

The Journal of Biological Chemistry 278:34427-34437

Lindsell, C., Shawber, C., Boulter, J., and Weinmaster, G. 1995. Jagged: A

Mammalian Ligand That Activates Notch1. Cell 80.6: 909-17.

Page 36: The Effect of Serrate Transmembrane Domain Substitution on

32

Logeat, F., Bessia, C., Brou, C., LeBail, O., Jarriault, S., Seidah, N. G. and Israel, A. 1998.

The Notch1 receptor is cleaved constitutively by a furin-like convertase. Proc. Natl.

Acad. Sci.USA 95: 8108-8112.

Mumm, J. S., Schroeter, E. H., Saxena, M. T., Griesemer, A., Tian, X., Pan, D. J., Ray, W. J.,

and Kopan, R. 2000. A ligand-induced extracellular cleavage regulates γ-secretase-

like proteolytic activation of Notch1. Mol. Cell. 5: 197-206.

Oda, T., Elkahloun, A., Pike, B., Okajima, K., Krantz, I., Genin, A., Piccoli, D., Meltzer, P.,

Spinner, N., Collins, F., and Chandrasekharappa, S.ettara C. 1997. Mutations in the

Human Jagged1 Gene Are Responsible for Alagille Syndrome. Nature Genetics

16.3: 235-42.

Pan, D., and Rubin, G. M. 1997. Kuzbanian controls proteolytic processing of Notch and

mediates lateral inhibition during Drosophila and vertebrate neurogenesis. Cell 90:

271-280.

Parks, A.L., Kleug, K.M., Stout, J.R. and Muskavitch, M.A.T. 2000. Ligand endocytosis

drives receptor dissociation and activation in the Notch pathway. Development 127:

1373-1385

Penton A., Leonard L., and Spinner N. 2012. Notch signaling in human development and

disease. Seminars in cell & developmental biology. 23(4):450-457.

Qi, H., Rand, M.D., Wu, X., Sestan, N., Wang, W., Rakic, P., Xu, T. and Artavanis-Tsakonas,

S. 1999. Processing of the Notch ligand Delta by the metalloprotease Kuzbanian.

Science 283: 91-94.

Page 37: The Effect of Serrate Transmembrane Domain Substitution on

33

Rebay, I., Fleming, R. J., Fehon, R. G., Cherbas, L., Cherbas, P. and Artavanis-Tsakonas, S.

1991. Specific EGF repeats of Notch mediate interactions with Delta and Serrate:

Implications for Notch as a multifunctional receptor. Cell 67: 687-699.

Rooke, J., Pan, D., Xu, T., and Rubin, G.M. 1996. KUZ, a conserved metalloprotease-

disintegrin protein with two roles in Drosophila neurogenesis. Science 273: 1227 –

1231.

Sanger, F., Coulson A., Barrell B., Smith A., and Roe B. 1980. Cloning in single-stranded

bacteriophage as an aid to rapid DNA sequencing. Journal of Molecular Biology

143.2:161-178.

Shawber, C., Nofziger, D., Hsieh, J., Lindsell, C., Bogler, O., Hayward, D., and Weinmaster,

G. 1996. Notch signaling inhibits muscle cell differentiation through a CBF1-

independent pathway. Development 122.12: 3765-3773.

Shimizu, K., Chiba S., Saito T., Takahasi T., Kumano K., Hamada Y., and Hirai H.

2002. Integrity of intracellular domain of Notch ligand is indispensable for cleavage

required for release of the Notch2 intracellular domain. The EMBO Journal 21.3:

294-302.

Struhl, G. and Adachi, A. 1998. Nuclear access and action of Notch in vivo. Cell 93: 649-660.

Sun, X., and Artavanis-Tsakonas, S. 1997. Secreted forms of DELTA and SERRATE define

antagonists of Notch signaling in Drosophila. Development 124: 3439-3448.

Technau, G.M. and Campos-Ortega, J.A. 1987. Cell autonomy of expression of neurogenic

genes of Drosophila melanogaster. Proc. Natl. Acad. Sci. USA 84: 4500-4504.

Page 38: The Effect of Serrate Transmembrane Domain Substitution on

34

Thomas, U., Speicher, S. A. and Knust, E. 1991. The Drosophila gene Serrate encodes an

EGF-like transmembrane protein with a complex expression pattern in embryos and

wing discs. Development 111: 749-761.

Wang, W. and Struhl, G. 2004. Drosophila Epsin mediates a select endocytic pathway that

DSL ligands must enter to activate Notch. Development 131: 5367-5380.

Westhoff, B., Colaluca, I. N., D'Ario, G., Donzelli, M., Tosoni, D., Volorio, S., et al. 2009.

Alterations of the Notch pathway in lung cancer. Proceedings of the National

Academy of Sciences. 106(52), 22293-22298.

Wharton, K.A., Johansen,K.M., Xu, T. and Artavanis-Tsakonas, S. 1985. Nucleotide

sequence from the neurogenic locus Notch implies a gene product that shares

homology with proteins containing EGF-like repeats. Cell 43: 567-581

Xu, T., Rebay, I., Fleming, R.J., Scottgale, T.N. and Artavanis-Tsakonas, S. 1990. The Notch

locus and the genetic circuitry involved in early Drosophila neurogenesis. Genes Dev.

4: 464-475.

Yamamoto, S., Charng, W-L., Rana, N.A., Kakuda, S., Jaiswal, M., Bayat, V., Xiong, B.,

Zhang, K., Sandoval, H., David, G., Wang, H., Haltiwanger, R.S., and Bellen, H.J.

2012. A mutation in EGF repeat-8 of Notch discriminates between Serrate/Jagged and

Delta family ligands. Science 338: 1229-1232.