regulation of foxo stability and activity by mdm2 e3 ligase

188
University of South Florida Scholar Commons Graduate eses and Dissertations Graduate School 2007 Regulation of FOXO stability and activity by MDM2 E3 ligase Wei Fu University of South Florida Follow this and additional works at: hp://scholarcommons.usf.edu/etd Part of the American Studies Commons is Dissertation is brought to you for free and open access by the Graduate School at Scholar Commons. It has been accepted for inclusion in Graduate eses and Dissertations by an authorized administrator of Scholar Commons. For more information, please contact [email protected]. Scholar Commons Citation Fu, Wei, "Regulation of FOXO stability and activity by MDM2 E3 ligase" (2007). Graduate eses and Dissertations. hp://scholarcommons.usf.edu/etd/2181

Upload: others

Post on 18-Feb-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Regulation of FOXO stability and activity by MDM2 E3 ligase

University of South FloridaScholar Commons

Graduate Theses and Dissertations Graduate School

2007

Regulation of FOXO stability and activity byMDM2 E3 ligaseWei FuUniversity of South Florida

Follow this and additional works at: http://scholarcommons.usf.edu/etd

Part of the American Studies Commons

This Dissertation is brought to you for free and open access by the Graduate School at Scholar Commons. It has been accepted for inclusion inGraduate Theses and Dissertations by an authorized administrator of Scholar Commons. For more information, please [email protected].

Scholar Commons CitationFu, Wei, "Regulation of FOXO stability and activity by MDM2 E3 ligase" (2007). Graduate Theses and Dissertations.http://scholarcommons.usf.edu/etd/2181

Page 2: Regulation of FOXO stability and activity by MDM2 E3 ligase

Regulation of FOXO Stability and Activity by MDM2 E3 Ligase

By

WEI FU

A dissertation submitted in partial fulfillment of the requirements for the degree of

Doctor of Philosophy Department of Pathology and Cell Biology

College of Medicine University of South Florida

Major Professor: Wenlong Bai, Ph.D. George Blanck, Ph.D. Jiandong Chen, Ph.D. Santo V. Nicosia, M.D

John C.M. Tsibris, Ph.D.

Date of Approval: Nov., 2007

Key words: Forkhead Transcriptional Factors, MDM2, Ubiquitination, Protein Degradation, Apoptosis

©Copyright 2007, Wei Fu

Page 3: Regulation of FOXO stability and activity by MDM2 E3 ligase

Dedication

To my parents, my in-laws, my brother,

my husband, Zhigang and my little pumpkin, caroline.

Without their love, understanding and support, I could

not have done even one piece of it.

Page 4: Regulation of FOXO stability and activity by MDM2 E3 ligase

Acknowledgement

I would like to extend my sincere appreciation to Dr. Wenlong Bai for guiding me in the

development of this project and the invaluable support from him at each step throughout

my graduate studies.

My appreciation also goes to Dr. Santo V. Nicosia for being helped in every way.

I would like to thank my adviser and committee members, Drs Bai, Blanck, Chen,

Nicosia and Tsibris for their guidance, valuable suggestion and their word of

encouragement.

My appreciation also goes to Dr. Yanping Zhang for willing to be my outside chair.

Thanks to Dr. Jiandong Chen and the members in his lab, in particular to Lihong Chen,

for their generous supply of the cell lines, plasmids, antibodies and priceless technical

assistant.

Thanks to Dr. Xiaohong (Mary) Zhang for all her kindly help.

Thanks to all the colleagues in Dr. Bai’s and Dr. Zhang’s Lab: Qiuping Ma, Yonghua

Yang, Pengfei Li, Heehyoung Lee, Feng Jiang, Chunrong Li, Betty Bean, Xiaohui Zhang,

Junying Bao, Lucy Hou, Zheng Shen, Guoqing Liu, Latasha Lee, Ushiwa Jinwal, Tung

Hsieh, Yingtao Zhang, Sarah Yong, Huiqin Dong and Mu Zhang for sharing their

opinions and joy with me.

Page 5: Regulation of FOXO stability and activity by MDM2 E3 ligase

Thanks to Dr. Patricia Kruk and all the members in her lab.

Thanks to Dr. Jin Cheng and the members in his lab, especially to Mei Sun, Zengqiang

Yuan and Hua Yang for valuable suggestion.

I would like thank to all the members in the Department of Pathology and Cell Biology.

Addtionally, I would like to thank everyone in Graduate Affairs Office, especially to Dr.

Barber, Dr. Joseph Krzanowski, Ms Kathy Zahn, Ms Susan Chapman.

Thanks to Moffitt & College of Medicine core facility for some analysis.

Thanks to all my friends who guide me, encourage me and help me on the way.

This thesis is dedicated to my family. Word cannot express my grateful feeling. I want to

say thanks to my husband, Zhigang, for his loving and my daughter, Caroline, for her

shining smile that let me feel I am the happiest mother in the world. I would like to thank

my parents and my brother, for their life-long loving. I would also like to thank my in-laws

for their kindly help.

The research in this dissertation has been partially supported by Predoctoral Fellowship

from American Heart Association.

Thanks to Dr. Tsibris and Dr. Aslam for reading this thesis and their valuable

suggestions.

Page 6: Regulation of FOXO stability and activity by MDM2 E3 ligase

i

Table of Contents

List of Figures................................................................................................................... vi

List of Tables..................................................................................................................... x

Abstract ............................................................................................................................ xi

INTRODUCTION............................................................................................................... 1

1. FOXO Family...................................................................................................... 1

1.1. FOX Family................................................................................................ 1

1.2. FOXO Family ............................................................................................. 4

1.2.1. Classification of Human FOXO Factors ............................................ 5

1.2.2. Domain Structure of FOXO............................................................... 7

1.3. Function of FOXO Factors......................................................................... 9

1.3.1. FOXO and Cell Cycle Checkpoint and DNA Repair ....................... 10

1.3.1.1. FOXO and G1/S Checkpoint.................................................. 10

1.3.1.2. FOXO and G2/M Checkpoint and DNA Repair ...................... 11

1.3.2. FOXO and Apoptosis ...................................................................... 12

1.3.3. FOXO and Atrophy ......................................................................... 13

1.3.4. FOXO and ROS Detoxification in Stem Cells ................................. 14

1.3.5. FOXO and Tissue Differentiation.................................................... 15

1.3.6. FOXO and Glucose and Energy Metabolism.................................. 16

Page 7: Regulation of FOXO stability and activity by MDM2 E3 ligase

ii

1.3.7. FOXO and Longevity ...................................................................... 17

1.4. Posttranslational Modifications of FOXO Proteins................................... 18

1.4.1. Phosphorylation of FOXO............................................................... 18

1.4.1.1. PKB and FOXO...................................................................... 18

1.4.1.2. SGK and FOXO ..................................................................... 20

1.4.1.3. IκB Kinase and FOXO3A ....................................................... 21

1.4.1.4. CK1 and FOXO...................................................................... 21

1.4.1.5. CDK2 and FOXO ................................................................... 21

1.4.1.6. JNK and FOXO ...................................................................... 21

1.4.1.7. MST and FOXO3A................................................................. 22

1.4.2. Acetylation and Deacetylation of FOXO ......................................... 23

1.4.2.1. Acetylation and Deacetylation................................................ 23

1.4.2.2. Acetylation of FOXO .............................................................. 23

1.4.2.2.1. P300 and CBP .............................................................. 23

1.4.2.2.2. P300/CBP and FOXO Acetylation................................. 25

1.4.2.3. Deacetylation of FOXO by HDAC .......................................... 26

1.4.2.3.1. SIRT1 ............................................................................ 26

1.4.2.3.2. SIRT1 and FOXO Deacetylation ................................... 28

1.4.3. Ubiquitination and Deubiquitination of FOXO ................................. 30

1.4.3.1. Ubiquitination and Degradation of FOXO .............................. 30

1.4.3.2. Deubiquitination of FOXO ...................................................... 31

2. Ubiquitin, Proteasome and MDM2 as an E3 Ligase......................................... 32

2.1. Ubiquitin-proteasome System.................................................................. 32

2.1.1. Ubiquitin and Ubiqutination............................................................. 32

2.1.2. Deubiquitination .............................................................................. 33

Page 8: Regulation of FOXO stability and activity by MDM2 E3 ligase

iii

2.1.3. Ubiquitination Machinery and Proteasome Pathway ...................... 35

2.2. MDM2 as an Ubiquitin E3 Ligase ............................................................ 37

2.2.1. General Information about MDM2................................................... 37

2.2.2. Functions of MDM2......................................................................... 38

2.2.2.1. MDM2 and Cell Cycle ............................................................ 38

2.2.2.2. MDM2 and Differentiation ...................................................... 38

2.2.2.3. MDM2 and Ribosome Biogenesis.......................................... 39

2.2.2.4. MDM2 and Transcription........................................................ 40

2.2.2.5. MDM2 and Protein Ubiquitination and Degradation............... 40

2.2.3. Regulation of MDM2 E3 Activity ..................................................... 41

2.2.3.1. Sumoylation of MDM2............................................................ 41

2.2.3.1.1. SUMO and Sumoylation................................................ 41

2.2.3.1.2. Sumoylation and MDM2................................................ 43

2.2.3.2. Ubiquitination and Degradation of MDM2 .............................. 43

2.2.3.3. Phosphorylation of MDM2...................................................... 44

2.2.3.3.1. DNA-PK, ATM and MDM2 ............................................ 44

2.2.3.3.2. PKB and MDM2 ............................................................ 45

2.2.3.3.3. c-Abl and MDM2 ........................................................... 46

2.2.3.3.4. CK2 and MDM2............................................................. 46

3. Functional interaction among FOXO, p53, and MDM2..................................... 49

3.1. Interaction between MDM2 and p53........................................................ 49

3.2. Interaction between p53 and FOXO ........................................................ 50

HYPOTHESIS & OBJECTIVES ...................................................................................... 53

MATERIALS AND METHODS ........................................................................................ 54

1. Chemicals, Antibodies and Cell Lines .............................................................. 54

Page 9: Regulation of FOXO stability and activity by MDM2 E3 ligase

iv

2. Plasmids ........................................................................................................... 54

3. Transfections and Immunological Assays ........................................................ 55

4. Ni-NTA Pull-down Assay .................................................................................. 57

5. In vitro Transcription Coupled Translations and GST (Glutathione

S-transferase) Pull-down Assays .................................................................. 58

6. In Vitro Ubiquitination Assay............................................................................. 58

7. Apoptotic Analysis and Flow Cytometry ........................................................... 59

RESULTS........................................................................................................................60

1. MDM2 promotes the degradation of FOXO...................................................... 60

2. MDM2 interacts with FOXO in vivo and in vitro ................................................69

3. The fork head box of FOXO and the region of MDM2 controlling

nuclear-cytoplasmic shuttling mediate the interaction between

MDM2 and FOXO............................................................................................. 77

4. MDM2 promotes the ubiquitination of FOXO1 and FOXO3A ........................... 88

5. MDM2 suppresses the expression of FOXO target genes and

protects cells from FOXO1-induced cell death ................................................. 96

6. MDM2 transiently increases FOXO transcriptional activity............................. 103

7. p53 induces transient increase in the transcriptional activity of

FOXO factors, which is followed by FOXO degradation in an

MDM2-dependent manner.............................................................................. 108

8. Site-specific sumoylation of SIRT1 regulates FOXO1

transcriptional activity and stability ................................................................. 116

9. Genistein-induced FOXO1 expression is blocked by MDM2

expression in H1299 cells............................................................................... 121

10. ARF promotes the MDM2-induced FOXO ubiquitination................................ 123

Page 10: Regulation of FOXO stability and activity by MDM2 E3 ligase

v

DISCUSSION................................................................................................................ 125

1. MDM2 is the E3 ubiquitin ligase of FOXO ..................................................... 125

2. MDM2-promoted ubiquitination of FOXO depends on

phosphorylation on PKB sites......................................................................... 126

3. MDM2-promoted ubiquitination of FOXO happenes mainly in the

cytoplasm ....................................................................................................... 127

4. MDM2 affects both the transcriptional activity and ubiquitination of

FOXO ............................................................................................................. 128

5. Mammalian FOXO factors interact with p53 .................................................. 129

6. MDM2 regulates FOXO factors in a p53 independent way ............................ 130

7. Different domains of MDM2 play different roles in the regulation of

p53 and FOXO factors (see Table 2) ............................................................. 132

8. The sumoylation status of SIRT1 affects the stability and activity of

FOXO ............................................................................................................. 134

9. Genistein increases FOXO expression levels through down-

regulation of MDM2 ........................................................................................ 134

10. ARF differentially affects E3 function of MDM2 toward different

substrates....................................................................................................... 135

SUMMARY AND PERSPECTIVES............................................................................... 136

REFERENCES.............................................................................................................. 139

About the author.................................................................................................. End Page

Page 11: Regulation of FOXO stability and activity by MDM2 E3 ligase

vi

List of Figures

Figure 1 Structure of human FOXO............................................................................ 8

Figure 2 The regulation of FOXO ............................................................................. 29

Figure 3 Ubiquitin-proteasome degradation pathway ............................................... 36

Figure 4 Structure of MDM2 gene and protein ......................................................... 47

Figure 5 Stress-induced FOXO and p53 pathway .................................................... 52

Figure 6 Inverse correlation of MDM2 and FOXO3A protein expression in

different cancer cell lines ............................................................................ 62

Figure 7 Inverse correlation of MDM2 and FOXO protein expression in MEFs........ 63

Figure 8 Stably expressed MDM2 decreases the level of endogenous

FOXO3A expression................................................................................... 64

Figure 9 Knockdown of endogenous MDM2 by siRNA causes an increase in

endogenous FOXO3A protein .................................................................... 65

Figure 10 Knockdown of endogenous MDM2 by shRNA increases the level of

ectopic FOXO1 protein ............................................................................... 66

Figure 11 MDM2 overexpression causes a decrease in Flag-FOXO1 protein,

which is blocked by MG132........................................................................ 67

Figure 12 MDM2 overexpression results in decrease in the half-life of the

FOXO1 protein ........................................................................................... 68

Figure 13 Exogenous MDM2 and Flag-FOXO1 are colocalized in the nucleus

of MEF cells................................................................................................ 70

Page 12: Regulation of FOXO stability and activity by MDM2 E3 ligase

vii

Figure 14 Endogenous MDM2 and FOXO3A are colocalized in the nucleus of

H1299 cells................................................................................................. 71

Figure 15 Interaction of ectopic FOXO1 and MDM2 in H1299 cells ........................... 72

Figure 16 Interaction of ectopic FOXO3A and MDM2 in HEK293T cells.................... 73

Figure 17 Interaction of endogenous MDM2 and FOXO1 in H1299/V138 cells ......... 74

Figure 18 Interaction of endogenous MDM2 and FOXO3A in HEK293T cells ........... 75

Figure 19 The interaction between FOXO1 and GST-MDM2 in vitro ......................... 76

Figure 20 Diagram of different FOXO mutants used in this study .............................. 79

Figure 21 Mapping of the MDM2-interacting domain of FOXO1 by

immunoprecipitation ................................................................................... 80

Figure 22 Mapping of the MDM2-interacting domain of FOXO3A by GST pull-

down assay................................................................................................. 81

Figure 23 Alignment of FOXO members’ fork head box............................................. 82

Figure 24 Diagram of different MDM2 mutants used in this study.............................. 83

Figure 25 Mapping of the FOXO1-interacting domain of MDM2 by

coimmunoprecipitations in the absence of MG132..................................... 84

Figure 26 An N-terminal truncation mutant and an NLS-mutant of MDM2

interact with FOXO1 in the presence of MG132......................................... 85

Figure 27 Truncation of the central region of MDM2 abolishes the interaction

between MDM2 and FOXO1 ...................................................................... 86

Figure 28 Immunofluorescence images show the cellular localization of

various MDM2 mutants............................................................................... 87

Figure 29 MDM2 promotes the ubiquitination of FOXO1............................................ 90

Figure 30 MDM2 promotes the ubiquitination of FOXO3A ......................................... 91

Page 13: Regulation of FOXO stability and activity by MDM2 E3 ligase

viii

Figure 31 The MDM2 ring finger domain is critical for the ubiquitination of

FOXO1 ....................................................................................................... 92

Figure 32 Ubiquitination of FOXO1 requires MDM2 ubiquitin ligase function............. 93

Figure 33 The central region of MDM2 cannot promote the polyubiquitination

of FOXO1 ................................................................................................... 94

Figure 34 MDM2 promotes FOXO1 polyubiquitination in vitro....................................95

Figure 35 Stably expressed MDM2 in H1299 cells regulates the expression of

downstream targets of FOXO..................................................................... 97

Figure 36 MDM2 siRNA increases the expression of FOXO1 target gene

TRAIL ......................................................................................................... 98

Figure 37 MDM2 promotes the cell survival in the presence of FOXO1..................... 99

Figure 38 MDM2 protectes cells from FOXO1-induced cell death measured by

Flow Cytometry......................................................................................... 101

Figure 39 MDM2 increases the transcriptional activity of FOXO1 in a dose-

dependent manner in DU145 cells ........................................................... 104

Figure 40 MDM2 increases the transcriptional activity of FOXO1 in NIH3T3

cells .......................................................................................................... 105

Figure 41 MDM2 enhances the ability of FOXO1 to inhibit cyclin D1 ....................... 106

Figure 42 MDM2 increases the transcriptional activity of both wild type FOXO1

and FOXO1 (AAA) mutant in NIH3T3 and DU145 cells ........................... 107

Figure 43 P53 inhibits the transcriptional activity of FOXO, in a dose-

dependent manner, but not FOXO1-induced cell death ........................... 109

Figure 44 P53 inhibits the transcriptional activity of FOXO in a dose-

dependent manner in LNCaP cells........................................................... 110

Figure 45 MDM2 relieves the repression of FOXO1 activity by p53......................... 111

Page 14: Regulation of FOXO stability and activity by MDM2 E3 ligase

ix

Figure 46 MDM2 transiently increases FOXO transcriptional activity, which is

followed by FOXO degradation ................................................................ 112

Figure 47 MG132 relieves p53-induced decrease in the expression of

FOXO3A protein in H1299/V138 cells ...................................................... 113

Figure 48 p53 decreases the protein level of FOXO1 through MDM2...................... 114

Figure 49 Knockdown of MDM2 partially relieves p53-induced FOXO3A

downregulation ......................................................................................... 115

Figure 50 SIRT1 inhibits the transcriptional activity of FOXO................................... 117

Figure 51 Nicotinamide treatment increases the FOXO1 transcriptional activity...... 118

Figure 52 SIRT1 sumoylation at Lys 734 is required for FOXO1 deacetylation ....... 119

Figure 53 Mutation of Lys 734 relieves the inhibition of FOXO1 transcriptional

activity by SIRT1....................................................................................... 120

Figure 54 Genistein increases the expression of FOXO through MDM2

downregulation ......................................................................................... 122

Figure 55 ARF promotes the MDM2-induced FOXO1 ubiquitination........................ 124

Figure 56 A working model shows the functional interaction among FOXO,

p53 and MDM2 ......................................................................................... 131

Page 15: Regulation of FOXO stability and activity by MDM2 E3 ligase

x

List of Tables

Table 1 The FOXO family members in mammals ........................................................ 6

Table 2 Localization and function of different MDM2 mutants.................................. 133

Page 16: Regulation of FOXO stability and activity by MDM2 E3 ligase

xi

REGULATION OF FOXO STABILITY AND ACTIVITY BY MDM2 E3 LIGASE

WEI FU

ABSTRACT

Members of the forkhead class O (FOXO) transcription factors are tumor

suppressors and key molecules that control aging and lifespan. The stability of

mammalian FOXO proteins is controlled by proteasome-mediated degradation but

general ubiquitin E3 ligases for FOXO factors remain to be defined. The current studies

demonstrate that MDM2 bound to FOXO1 and FOXO3A and promoted their

ubiquitination and subsequent degradation, a process apparently dependent on FOXO

phopshorylation at PKB sites and on the E3 ligase activity of MDM2. The binding

occurred between endogenous proteins and was involved the forkhead box of FOXO1

and the region of MDM2 that controls its cellular localization. MDM2 promoted the

ubiquitination of FOXO1 in vitro in a cell free system. Knocking down MDM2 by siRNA

caused the accumulation of endogenous FOXO3A protein, and enhanced the

expression of FOXO target genes. In addition, MDM2 promoted the transcriptional

activity of FOXO in a transient transfection system. In cells stably expressing a

temperature sensitive mutant p53, activation of p53, by shifting to permissive

temperatures led to MDM2 induction and the degradation of endogenous FOXO3A.

These data suggested that MDM2 acts downstream of p53 as an E3 ubiquitin ligase to

promote the degradation of mammalian FOXO factors.

Page 17: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 1 -

INTRODUCTION

1. FOXO Family

1.1. FOX Family

Forkead proteins constitute a family of structurally related transcriptional factors

that have been identified in all eukaryotes ranging from yeast to human. Forkhead

transcription factors are characterized by the forkhead domain, which is a ~100 amino-

acid monomeric DNA binding domain. The three-dimensional structure of forkhead

domain consists of two W1 and W2 loops (or wings) and three α helices (Lehmann et al.,

2003). Due to its butterfly-like appearance, the domain is also known as “winged-helix”

domain.

The first member of the forkhead family was identified in 1989 as a nuclear

homeotic gene involved in the embryonic development of flies (Drosophila

melanogaster). To date, over 100 members of the forkhead family have been identified in

species ranging from Saccharomyces cerevisiae to humans (Kaestner et al., 2000). The

nomenclature of the forkhead transcription factors has recently been revised. These

genes, now termed Fox (after Forkhead box), are divided into 17 subclasses (A to Q),

according to the amino acid sequence of their conserved forkhead domains

(http://www.biology.pomona.edu/fox.html).

Comparative genome analyses have shown that the number of forkhead

transcription factors appears to have increased during evolution, with a greater number

identified in vertebrates than in invertebrates (Lehmann et al., 2003). The origin and

Page 18: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 2 -

expansion of forkhead genes is positively correlated with eukaryotic complexity because

among the organisms for which genome sequences are available, there is a correlation

between anatomical complexity and forkhead gene number: 4 FOX genes in S.

cerevisiae, 15 in Caenorhabditis elegans, 20 in Drosophila melanogaster and 43 in

humans (Mazet et al., 2003). The human FOX gene family consists of : FOXA1, FOXA2,

FOXA3, FOXB1, FOXC1, FOXC2, FOXD, FOXD2, FOXD3, FOXD4, FOXD5

(FOXD4L1), FOXD6 (FOXD4L3), FOXE1, FOXE2, FOXE3, FOXF1, FOXF2, FOXG1

(FOXG1B), FOXH1, FOXI1, FOXJ1, FOXJ2, FOXJ3, FOXK1, FOXK2, FOXL1, FOXL2,

FOXM1, FOXN1, FOXN2 (HTLF), FOXN3 (CHES1), FOXN4, FOXN5 (FOXR1), FOXN6

(FOXR2), FOXO1 (FOXO1A, FKHR), FOXO2 (FOXO6), FOXO3 (FOXO3A), FOXO4

(MLLT7), FOXP1, FOXP2, FOXP3, FOXP4, and FOXQ1. Members of FOX subfamilies

A-G, I-L and Q were grouped into class-1 FOX proteins, while members of FOX

subfamilies H and M-P were grouped into class-2 FOX protein. The presence of a C-

terminal basic region within the FOX domain is the common feature of class-1 FOX

protein (Katoh and Katoh, 2004b).

FOX transcriptional regulators play various roles during development, from

organogenesis to language acquistion. Foxb1 is reported to control development of

mammary glands and regions of the central nervous system that regulate the milk

ejection reflex in mice (Kloetzli et al., 2001). FOXC1 and FOXC2 (Kume et al., 2000;

Kume et al., 1998; Wilm et al., 2004), FOXE1 (Aza-Blanc et al., 1993; Ortiz et al., 1997;

Zannini et al., 1997), FOXF1 (Kalinichenko et al., 2001; Kalinichenko et al., 2002),

FOXJ1 (Pelletier et al., 1998) and FOXJ2 (Granadino et al., 2000) play important roles in

the development of the organs, such as thyroid organogenesis, mesonephros, gonad,

liver, gallbladder, lung and intestinal tract. FOXD1 (Hatini et al., 1996), FOXE3 (Blixt et

al., 2007; Valleix et al., 2006) and FOXL2 (De Baere et al., 2001; De Baere et al., 2002)

Page 19: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 3 -

play a critical role in the development of eyes. FOXD3 (Alkhateeb et al., 2005; Guo et

al., 2002), FOXD5 (Yu et al., 2002) and FOXJ3 (Landgren and Carlsson, 2004) are

involved in neural development. FOXD4 (Minoretti et al., 2007) and FOXP4 (Li et al.,

2004b) are related to heart development and differentiation. The FOXG1 (Pauley et al.,

2006) and FOXI1(Hulander et al., 2003) are required for the development of the

mammalian inner ear. FoxH1 (FAST) is a transcription factor that mediates signaling by

transforming growth factor-β, activin, and nodal. FoxH1 / embryos failed to orient the

anterior-posterior (A-P) axis correctly (Yamamoto et al., 2001). FOXJ1 is required for late

stages of ciliogenesis (You et al., 2004b). It is essential for nonrandom determination of

left-right asymmetry and development of ciliated cells. FOXN4 is necessary and

sufficient for commitment to the amacrine cell fate and is nonredundantly required for the

amacrine and horizontal cells (Li et al., 2004a). FOXP2 has been implicated in the

acquisition of grammatical skills (Hurst et al., 1990; Lai et al., 2001). FOXP3 is the

master regulator in the development and function of regulatory T cells and the selected

marker for them (Alvarado-Sanchez et al., 2006; Bennett et al., 2001; Brunkow et al.,

2001; Suri-Payer and Fritzsching, 2006). Human FOXN1 (Frank et al., 1999) and

FOXQ1 (Hong et al., 2001) function in hair differentiation. FOXN1 is also required for the

growth and differentiation of thymic epithelial cells (Balciunaite et al., 2002).

FOX factors are also very important for cell growth, differentiation and

tumorigenesis. FOXA1 binds to the promoters of more than 100 genes associated with

metabolic processes, the regulation of signaling and the cell cycle (Carlsson and

Mahlapuu, 2002; Kaestner, 2000; Tomaru et al., 2003). High expression of FOXA1 (Gao

et al., 2003; Lin et al., 2002b), FOXN6 (Katoh and Katoh, 2004d), FOXQ1 (Bieller et al.,

2001) and FOXM1(Nakamura et al., 2004) has been reported in various tumors,

including lung, esophageal, breast, prostate and pancreatic cancers. Several FOX

Page 20: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 4 -

factors are thought of as the candidate tumor suppressor genes. This includes FOXC1

(Zhou et al., 2002), FOXK2 (Li et al., 1992), FOXN5 (Katoh and Katoh, 2004a, c) and

FOXO family (Medema et al., 2000). These factors are reported either to be inactive in

cancer or to suppress tumor cell growth when overexpressed. FOXM1 is expressed in

proliferating cells and becomes silenced in terminally differentiated cells (Korver et al.,

1997; Yao et al., 1997; Ye et al., 1997). FOXM1 might be beneficial to patients whose

lung endothelial-cell barrier has been damaged (Zhao et al., 2006).

FOX factors also regulate chromatin remodeling and transcription. The FOXA3

acts in chromatin remodeling (Kaestner et al., 1994). FOXK1 is essential for regulating

cell cycle progression in myogenic progenitor cells (Hawke et al., 2003). FOXL1 is

capable of remodeling chromatin higher-order structure and can cause stable and site-

specific changes of chromatin by either creating or removing DNase I hypersensitive

sites, resulting in changes the proliferation, differentiation, and positioning of epithelial

cells (Kaestner et al., 1997; Yan et al., 2006). FOXN3 binds to Ski-interacting protein

(SKIP, NCoA-62), which is a transcriptional co-regulator known to associate with a

repressor complex (Scott and Plon, 2005).

It is believed that FOX factors form a network. For example, FOXG1, a class-1

FOXO member, acts as a repressor of transcriptional activity of the FOXO family (Aoki et

al., 2004). FOXA and FOXO factors cooperate in complex regulatory networks of the

pancreas and liver (Czech, 2003).

1.2. FOXO Family

FOXO factors are members of the O-subclass of the class-2 FOX family. A

unique five amino acid (GDSNS) insertion immediatedly prior to helix H3 within the

forkhead domain is characteristic of this subfamily. This insertion is absent in all other

Page 21: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 5 -

FOX transcription factors (Arden, 2006). The first member of FOXO was identified in C.

elegans (Caenorhabditis elegans) and named DAF-16 and was shown to regulate dauer

formation in C. elegans (Thomas, 1993).

1.2.1. Classification of Human FOXO Factors

In invertebrates there is only one FOXO gene, termed daf-16 in the worm and

dFOXO in the fly. In mammals there are four functional FOXO genes, FOXO1, 3A, 4,

and 6 (Carter and Brunet, 2007) (Table 1).

The FOXO1 gene is located in chromosome 13q14.1. It is the most abundant

FOXO isoform in insulin-responsive tissues such as liver, adipose tissue, pancreatic

cells, kidney, spleen and brain (Armoni et al., 2006). Embryos of FOXO1 homozygous

mice are smaller and die at embryonic day 10.5 due to several embryonic defects

(Nakae et al., 2002), such as incomplete vascular development (Hosaka et al., 2004).

Analysis of heterozygote mutant mice indicates that FOXO1 is involved in the function of

pancreatic β-cells, hepatic glucose metabolism and adipocyte differentiation (Kitamura et

al., 2002; Nakae et al., 2002; Nakae et al., 2003).

The FOXO3A gene is located in chromosome 6q21. It is highly expressed in the

heart, spleen, lung, kidney, ovary, adipose tissue and brain (Zhu et al., 2004). FOXO3A

knockout mice reveal haematological abnormalities, decreased glucose uptake in

glucose tolerance tests (Castrillon et al., 2003) and a distinct ovarian phenotype due to

premature follicular activation that leads to oocyte death and subsequent depletion of

follicles. It has been suggested that FOXO3A acts as a suppressor of follicular activation

(Castrillon et al., 2003; Hosaka et al., 2004). In mature endothelial cells, FOXO1 and

FOXO3A are the most abundant FOXO isoforms. Overexpression of constitutively active

Page 22: Regulation of FOXO stability and activity by MDM2 E3 ligase

6

Page 23: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 7 -

FOXO1 or FOXO3A significantly inhibits endothelial cell migration and tube formation in

vitro (Potente et al., 2005).

FOXO4 gene is located in chromosome Xq13.1. It is abundant in the lungs, brain,

heart and skeletal muscle and kidneys (Nakae et al., 2001). FOXO4 knockout mice show

no an obvious abnormalities (Hosaka et al., 2004).

FOXO6 gene is located in 1p34.1. Embryonic expression of FOXO6 is seen

predominantly in the developing brain where it is expressed in a specific temporal and

spatial pattern. In the adult animal, FOXO6 expression persists in the nucleus

accumbens, cingulated cortex, parts of the amygdale, and hippocampus. FOXO6 is also

expressed in kidney and thymus (Jacobs et al., 2003). The phenotype of FOXO6-

deficient animals is not reported, but its restricted expression pattern suggests that it

may play a role in embryologic development of the central nervous system (Hoekman et

al., 2006).

1.2.2. Domain Structure of FOXO

From the N-terminus to the C-terminus, the FOXO protein contains a proline-rich

domain, a forkhead domain, NLS (nuclear localization signal), NES (nuclear export

signal), a LxxLL motif and an activation domain (Figure 1). The proline rich motif binds to

the CH3 region of CBP/p300 and stabilizes the interaction between FOXO factors and

CBP/p300 (van der Heide and Smidt, 2005). The Forkhead domain is responsible for

binding to target gene promoters. The central region of FOXO includes the NLS and

NES and accounts for the cellular localization of FOXO. The LxxLL motif is reported to

bind to SIRT1 and has an important role in regulating its transcriptional activity (Nakae et

al., 2006). The C-terminus of FOXO contains the activation domain which stimulates the

promoter activity (Yang et al., 2005). In the nucleus, all FOXO proteins bind to DNA as

Page 24: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 8 -

FOX150-260

LxxLL461-466

AD596-655

NLS NESFOXO1 655aa

FOXO3 673aa

FOXO4 501aa

FOXO6 509aa

Nt Ct

FOX 148-258

FOX 88-198

FOX 78-188

368-377

303-327

300-308

PxPxP

Figure 1 Structure of human FOXO. Full-length FOXO and known motifs

are presented. PxPxP-proline rich motif; FOX-forkhead domain; LXXLL-L,

leucine; X, any amino acid; AD-activation domain; Nt-amino terminal; NLS-

nuclear localization signal; NES-nuclear export signal; Ct-carboxyl terminal.

The numbers above the drawings denote amino acid numbers.

Page 25: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 9 -

monomers through the FOX domain. The core motif of the consensus recognition site for

FOXO on DNA is GTAAA (C/T) A and it is designated as the DBE for DAF-16 family

member-binding element.

1.3. Functions of FOXO Factors

Studies in C. elegans showed that direct activation of DAF-16, or mutation of the

insulin-PI3K (phosphotidylinositol 3 kinase)-PKB (protein kinase B) pathway results in

extension of lifespan, stress resistance and arrest at the dauer diapause stage (Ogg et

al., 1997). Besides cell-autonomous inputs, DAF-16 also responds to environmental

cues such as starvation, heat and oxidative stress. All these stress signals activate DAF-

16, whereas nutrient-rich conditions deactivate it. dFOXO, the unique FOXO homologue

in Drosophila, seems to play similar roles, which are negatively controlled by the insulin-

PI3K-PKB signaling cascade and nutrients (Junger et al., 2003; Kramer et al., 2003;

Puig et al., 2003), although dFOXO-knockout flies are viable and of normal size, they are

more vulnerable to the oxidative stress. These data suggest that dFOXO offers

protection against oxidative stress.

In humans, the pivotal role of FOXO on cell fate decisions depends on the

balance between growth factor stimulation versus cellular stress and damage. Both

circumstantial and direct evidences implicate a role of FOXO factors in cancer. The first

three members of FOXO were found at chromosomal translocations in tumors. The

FOXO1 gene is fused to PAX3 or PAX7 gene in rhabdomyosarcoma (Galili et al., 1993;

Sorensen et al., 2002). The FOXO3A gene is fused to MLL gene in secondary acute

myeloblastic leukemia (Hillion et al., 1997). The FOXO4 gene is fused to MLL gene in

acute lymphocytic leukemia (Parry et al., 1994). Nuclear exclusion of FOXO3A in

primary breast tumors correlates with PI3K activation and poor survival of the patients

(Hu et al., 2004). FOXO factors reduce tumorigenicity in nude mice (Hu et al., 2004;

Page 26: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 10 -

Ramaswamy et al., 2002). Furthermore, FOXO proteins interact with many oncogenes

such as β-catenin (Essers et al., 2005) or tumor suppressors such as p53 (Brunet et al.,

2004).

The physiological importance of FOXO transcription factors is diverse as

revealed by loss and gain of function experiments in transgenic and knockout mice. All

lines of evidence point to FOXO factors as central regulators of metabolism, aging,

proliferation and differentiation (Hosaka et al., 2004). Since a wide range of human

diseases, including cancer, have striking aging-dependent onset, FOXO factors are

believed to serve as molecular links between longevity and tumor suppression (Greer

and Brunet, 2005).

1.3.1. FOXO and Cell Cycle Checkpoint and DNA Repair

Mammalian cells have evolved an intricate defense network to maintain genomic

integrity by preventing the fixation of permanent damage from endogenous and

exogenous mutagens. Cell cycle checkpoints, a major genomic surveillance mechanism

acting at the G1/S and G2/M boundaries, are regulated in response to DNA damage

(Hartwell and Weinert, 1989). Defects in these steps may result in a mutated phenotype

that is associated with tumorigenesis.

1.3.1.1. FOXO and G1/S Checkpoint

p21 is a cyclin- dependent kinase inhibitor. Loss of p21 expression has been

described as a poor prognostic factor and as an independent predictor of bladder cancer

progression in muscle invasive cancer (Stein et al., 1998). p27 is a protein that binds to

cyclins and cdk to block the entry into S phase. Loss of p27 expression has been shown

to be associated with aggressive behavior in a variety of human epithelial tumors,

including prostate (Macri and Loda, 1998) and breast (Catzavelos et al., 1997; Gillett et

Page 27: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 11 -

al., 1999; Tan et al., 1997; Wu et al., 1999) cancers. p130 belongs to the RB family. A

study in mice showed that p130 can induce cell cycle arrest (Classon et al., 2000).

Mammalian cyclins are classified into 12 different types, from cyclins A to I, based on

structural similarity, function period in the cell division cycle, and regulated protein

expression. cyclin D1 is a key regulator of the G1 phase of the cell division cycle where it

binds to and activates the cyclin-dependent kinases CDK4 and CDK6. In the cells that

have the capacity to divide, the main effect of the expression of active forms of FOXO

family members is to promote cell cycle arrest at the G1/S boundary. FOXO factors play

a major role in G1 arrest by upregulating cell cycle inhibitors such as p21 (Hauck et al.,

2007; Lawlor and Rotwein, 2000), p27 (Medema et al., 2000; Stahl et al., 2002) and

p130 (Chen et al., 2006) and by repressing cell cycles activators cyclin D1 and D2

(Schmidt et al., 2002).

1.3.1.2. FOXO and G2/M Checkpoint and DNA Repair

GADD45 is growth arrest and DNA damage-inducible protein 45. It was identified

on the basis of its rapid transcriptional induction after UV irradiation (Fornace et al.,

1989) and can induce G2/M arrest (Wang et al., 1999). Induction of GADD45 is also

observed after several types of pathological stimuli including various environmental

stresses: hypoxia, irradiation, genotoxic drug exposure and growth-factor withdrawal

(Papathanasiou et al., 1991).Three G-type cyclins have been identified: cyclins G1, G2,

and I. All three are expressed in terminally differentiated cells, act as cell cycle inhibitors

in certain cell types and may induce cell cycle arrest (Martinez-Gac et al., 2004).

Microarray analysis led to the identification of GADD45 and cyclin G2 as the

downstream target genes of FOXO3A for G2/M arrest and DNA repair (Tran et al.,

2002).

Page 28: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 12 -

In vivo experiment showed that FOXO3A and FOXO4 activate GADD45

promoter through direct interaction with the DBE. Oxidative stress activates the GADD45

promoter in a FOXO-dependent manner, resulting in increased level of GADD45 mRNA

and protein as well as G2 arrest (Furukawa-Hibi et al., 2002). In response to glucose

starvation, FOXO1 also affects GADD45 mRNA level in islets (Martinez et al., 2006).

Cyclin G2 is expressed in various amounts during the cell cycle in lymphocytes

(Horne et al., 1997; Horne et al., 1996). Ectopic expression of cyclin G2 inhibits cell

cycle progression (Bennin et al., 2002). FOXO3A interacts with cyclin G2 in mouse

fibroblasts. Active forms of FOXO3A increase cyclin G2 mRNA levels by activating the

cyclin G2 transactivation through interaction with the DBE in its promoter (Martinez-Gac

et al., 2004).

One mechanism by which cells protect themselves against stress is by repairing

the damage to their DNA and proteins that occurs upon exposure to environmental

stress. Furthermore, this capacity to repair DNA damage is closely correlated with an

increased longevity in mammals (Kirkwood and Austad, 2000). Gene array analysis

showed that FOXO3A is involved in nuclear excision DNA repair by modulating the

expression of GADD45 and DDB (damaged DNA binding) protein (Tran et al., 2002).

1.3.2. FOXO and Apoptosis

FOXO factors have been shown to mediate apoptosis by activating proapoptotic

genes in a variety of cells. FOXO promotes cell death through these downstream

targets: TRAIL (Tumor necrosis-related apoptosis-inducing ligand), FasL (Fas Ligand)

(Brunet et al., 1999), Bim (Stahl et al., 2002; Sunters et al., 2003; Urbich et al., 2005)

and PUMA (You et al., 2006a).

TRAIL is a member of the TNF family of cytokines. It induces apoptosis via death

receptors (DR4 and DR5) in a wide variety of tumor cells but not in normal cells (Suliman

Page 29: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 13 -

et al., 2001). TRAIL is a direct target of FOXO3A. FOXO3A response element is located

in TRAIL promoter spanning nucleotides -138 to -121. Decreased activity of FOXO3A

and FOXO1 in prostate cancers resulting from loss of PTEN leads to a decrease in

TRAIL expression that can contribute to increased survival of the tumor cells (Modur et

al., 2002).

FasL is a type II transmembrane protein. It functions as a homotrimer, because it

trimerizes Fas receptor, which spans the membrane of the "target" cell. This

trimerization usually leads to apoptosis.There are three putative FOXO binding elements

in the FasL promoter. In the human leukemia T cell line Jurkat, FOXO3A induces

apoptosis by activating FasL expression and the Fas-FasL apoptotic pathway

(Yamamura et al., 2006).

Bim is a BH3-only Bcl2 family member. Bim (also known as Bcl2l11) provokes

apoptosis (O'Connor et al., 1998). There are two conserved FOXO binding sites in the

Bim promoter: one at position -204 relative to the transcription start site (bim1) and one

at the boundary between exon 1 and the first intro. FOXO3A directly activates the bim

promoter via the two conserved FOXO binding sites (Gilley et al., 2003).

PUMA is an essential mediator of p53-dependent and -independent apoptosis in

vivo. Promoter analysis identifies that there is a potential consensus FOXO-responsive

element in intron 1 of PUMA which is conserved in humans and mice. FOXO3A up-

regulates PUMA transcription in response to cytokine or growth factor deprivation (You

et al., 2006a).

1.3.3. FOXO and Atrophy

Atrophy is the partial or complete decompositon of a part of the body. Causes of

atrophy include poor nourishment, poor circulation, loss of hormonal support, loss of

nerve supply to the target organ, disuse or lack of exercise or disease intrinsic to the

Page 30: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 14 -

tissue itself. Hormone and nerve inputs that maintain an organ or body part are referred

to as trophic. Atrophy is a general physiological process of reabsorption and breakdown

of tissues, involving apoptosis on a cellular level. When it occurs as a result of disease

or loss of trophic support due to other disease, it is termed pathological atrophy,

although it can be a part of normal body development and homeostasis as well.

Skeletal muscle atrophy is a debilitating response to fasting, disuse, cancer, and

other systemic diseases. FOXO3A acts on the atrogin-1 promoter to cause atrogin-1

transcription and dramatic atrophy of myotubes and muscle fibers. When FOXO

activation is blocked by a dominant-negative construct in myotubes or by RNAi in mouse

muscles in vivo, atrogin-1 induction is prevented during starvation and atrophy of

myotubes induced by glucocorticoids (Sandri et al., 2004).

In fully differentiated skeletal and cardiac muscle cells, expression of a

constitutively active form of FOXO3A causes atrophy. The atrophy is not due to

apoptosis. Instead, it is due to a decrease in cell size.

1.3.4. FOXO and ROS Detoxification in Stem Cells

FOXO proteins have been reported to allow detoxification of ROS (reactivie

oxygen species) by upregulating free radical scavenging enzymes, including MnSOD

(manganese superoxide dismutase) and catalase (Storz, 2006).

FOXO3A protects quiescent cells from oxidative stress by directly increasing

MnSOD mRNA and protein levels. This increase in protection from reactive oxygen

species antagonizes the apoptosis caused by glucose deprivation. Increased resistance

to oxidative stress is also associated with longevity. The model of Forkhead involvement

in regulating longevity stems from genetic analysis in C. elegans, but the model also

can be extendable to mammalian systems (Kops et al., 2002).

Page 31: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 15 -

Stem cells are primal cells found in all multi-cellular organisms. They have two

important characteristics that distinguish them from other types of cells. First, they are

unspecialized cells that renew themselves for long periods through cell division. The

second characteristic is that under certain physiologic or experimental conditions they

can be induced to become cells with special functions such as the beating cells of the

heart or the insulin-producing cells of the pancreas. Regulation of oxidative stress in the

HSC (hematopoietic stem cell) compartment is critical for the maintenance of HSC self-

renewal. FOXO-dependent signaling is required for the long-term regenerative potential

of the HSC compartment through regulation of HSC responses to physiologic oxidative

stress, quiescence and survival. Further analysis of ROS levels in FOXO knockout mice

showed that FOXOs affect HSC integrity by regulating ROS. Myeloid progenitor cells

isolated from FOXO1/3/4 conditional knockout animals show decreased ROS (Tothova

et al., 2007).

1.3.5. FOXO and Tissue Differentiation

Cell and tissue differentiation process systematically bases on a number of gene

expressions that commence successively along with the passage of time. In

differentiating cells, FOXO factors have been implicated in inhibiting and promoting

differentiation, depending on the cell types and different FOXO isoforms. In adipocytes,

FOXO1 is induced in the early stages of adipocyte differentiation but its activation is

delayed until the end of the clonal expansion phase. Constitutively active FOXO1

prevents the differentiation of preadipocytes, while dominant-negative FOXO1 restores

adipocyte differentiation of fibroblasts from insulin receptor-deficient mice. FOXO1

directly inhibits the differentiation through upregulation of p21 (Nakae et al., 2003).

Hematopoiesis, or blood cell formation, is the process in which a limited set of

hematopoietic stem cells is able to give rise to all types of functional blood cells via

Page 32: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 16 -

commitment to specific hematopoietic lineages. FOXO3A can promote erythroid

differentiation through BTG1 (B-cell translocation gene 1). BTG1, in turn, modulates

arginine methylation necessary for erythroid differentiation (Bakker et al., 2004).

Furthermore, recent experiments demonstrated that FOXO3A directly binds and

represses the transcription of the ID1 (inhibitor of differentiation 1), a suppressor of

erythroid differentiation, through the recruitment of an HDAC1 (histone deacetylase 1)–

mSin3a complex (Lam et al., 2006) .

1.3.6. FOXO and Glucose and Energy Metabolism

FOXO factors play an important role in upregulating genes that control glucose

metabolism. They upregulate G6Pase (glucose-6-phosphatase) which is responsible for

converting glucose-6-phosphate to glucose (Nakae et al., 2001; Onuma et al., 2006) and

PEPCK (phosphoenolpyruvate carboxykinase) which converts oxaloacetate to

phosphoenolpyruvate (Samuel et al., 2006). In the absence of insulin, FOXO1 binds to

IRE (insulin response elements) in G6Pase and PEPCK promoters and stimulates their

promoter activity. However, in the presence of insulin, FOXO1 undergoes

phosphorylation and is exported out of the nucleus, ceasing its transcriptional activity

(Kitamura et al., 2002). In liver and muscle, FOXO proteins can stimulate PDK4 (

pyruvate dehydrogenase kinase-4) expression, which limits oxidative metabolism of

glucose and conserves glucose for utilization in other tissues (Furuyama et al., 2003).

FOXOs are also involved in lipid metabolism through the regulation of AdipoR1/2

(Tsuchida et al., 2004), LPL (lipoprotein lipase) (Kamei et al., 2003), HMGCS2

(mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase) (Nadal et al., 2002) and SCP

(sterol carrier protein) (Dansen et al., 2004) gene expression. Adiponectin/Acrp30 is a

hormone secreted by adipocytes, which acts as an antidiabetic and antiatherogenic

adipokine. AdipoR1 and -R2 serve as the receptors for adiponectin and mediate

Page 33: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 17 -

increased fatty acid oxidation and glucose uptake by adiponectin. Mouse study shows

that insulin reduces the expression of AdipoR1/R2 via the PI3K/FOXO1-dependent

pathway in vitro (Tothova et al., 2007). FOXO proteins also stimulate the expression of

both PDK4 (Furuyama et al., 2003; Kwon et al., 2004), which limits the flux of pyruvate

through the Krebs cycle, and the expression of LPL (Kamei et al., 2003). HMGCS2 is a

key enzyme controling ketogenesis. Deletion analysis showed that there is a FOXO3A-

responsive sequence AAAAATA located 211 bp upstream of the HMGCS2 gene

transcription start site. FOXO3A can stimulate transcription of HMGCS2 and this

stimulation is repressed by insulin (Nadal et al., 2002). The SCP (sterol carrier protein)

gene encodes two proteins, SCPx and SCP2, which are independently regulated by

separate promoters. SCPx has been shown to be the thiolase involved in the breakdown

of branched-chain fatty acids and in the biosynthesis of bile acids. Both SCPx and SCP2

are upregulated by FOXO3A (Dansen et al., 2004).

1.3.7. FOXO and Longevity

A number of diseases, including cancer, type 2 diabetes and neurodegenerative

disorders have a striking age-dependent onset. A number of studies now support the

hypothesis that longevity could be affected by simple changes in the environment.

Organismal longevity also has a genetic component. A pivotal breakthrough in

identifying genes involved in longevity came from studies in C. elegans. Some worm

mutants can live two to three times longer than wild type worms (Kenyon et al., 1993).

These long-living worms turned out to have mutations in the insulin receptor gene

(Kimura et al., 1997). Remarkably, flies and mice that have mutations in the insulin or

the IGF-1 (insulin like growth factor-1) receptor genes are also long-lived (Bluher et al.,

2003; Holzenberger et al., 2003; Tatar et al., 2001). These results indicate that insulin

and IGF-1 regulate longevity in a conserved manner throughout species.

Page 34: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 18 -

A link between FOXO factors and aging was also initially observed in

invertebrates. In C. elegans, DAF-16 regulates lifespan (Baumeister et al., 2006). In

Drosophila melanogaster, dFOXO regulates age-linked decline of cardiac function and

longevity (Giannakou et al., 2004; Giannakou and Partridge, 2004; Hwangbo et al.,

2004; Wessells et al., 2004).

But so far, there is no direct evidence to link FOXO factors and longevity in

mammals. Since FOXO factors are involved in cellular resistance to stress and promote

ROS detoxification, DNA repair, and cell cycle arrest to allow time for the repair process,

they are projected to reduce detrimental effects of aging in mammals (Glauser and

Schlegel, 2007). Thus, a role of FOXO factors in longevity of mammals is expected.

1.4. Posttranslational Modifications of FOXO Proteins

As described above, FOXO proteins have diverse cellular functions by acting as

transcription factors. The activity of FOXO proteins can be regulated by posttranslational

modifications including phosphorylation, acetylation and ubiquitination.

1.4.1. Phosporylation of FOXO

Phosphorylation is a process wherein phosphate groups are added to proteins by

protein kinases or removed from proteins by protein phosphatases. The addition or

removal of phosphate groups dramatically alters protein function and leads to a myriad

of biological responses. Phosphorylation is shown to play a major role in the regulation

of FOXO cellular localization, transcription activity and protein stability, and is mediated

mainly by PKB.

1.4.1.1. PKB and FOXO

PKB is a serine/threonine kinase. It plays a pivotal role in cell survival and

proliferation through a number of downstream effectors. All FOXO family members,

Page 35: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 19 -

except FOXO6, contain three PKB phosphorylation sites (RXRXX(S/T)X). DAF-16, the

C. elegans FOXO, contains four putative PKB recognition motifs. Two are located at end

of the N-terminal & C-terminal of DAF-16. The other two sites are located in the forkhead

domain and overlap. Disrupting PKB-consensus phosphorylation sites in DAF-16 causes

nuclear accumulation in wild-type animals but has little effect on lifespan (Lin et al.,

2001; Van Der Heide et al., 2004).

PKB is shown to phosphorylate human FOXO family members on the three PKB

motifs: an N-terminal threonine (Thr32), a forkhead box serine (Ser253), and a more C-

terminal serine (Ser315). Thr32 is preferentially phosphorylated by PKB. This

phosphorylation is shown to inhibit p300 binding to FOXO3A, resulting in the recruitment

of RanGTPase and CRM1 protein to transport FOXO3A to cytoplasm and to bind the 14-

3-3 chaperone (Mahmud et al., 2002). Mutation of Ser253 to a neutral residue, such as

alanine (S253A), inhibits further phosphorylation of FOXO3A in other regions of the

proteins by PKB and other kinases. This suggests that this site is necessary to prime

other sites for phosphorylation. The FOXO3A S253A mutant is constitutively nuclear

under different conditions including PKB activation. Therefore, the S253 is called the

“gatekeeper site”. This site is believed to inhibit DNA binding, to change the

conformation and to inhibit nuclear re-import (Brownawell et al., 2001; Brunet et al.,

1999; Brunet et al., 2001; Woods and Rena, 2002). Phosphorylation of Ser315 is shown

to enhance nuclear export (Brunet et al., 2001; Rena et al., 2002). All FOXO proteins

have been shown to require the consensus N-terminal PKB site and the PKB site

located in the forkhead domain in order to translocate from nucleus to cytosol

(Brownawell et al., 2001).

Interestingly, the regulation of FOXO6 is different from those of FOXO1,

FOXO3A and FOXO4. FOXO6 only contains two PKB/SGK regulatory phosphorylation

Page 36: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 20 -

sites (Thr26 and Ser184 in mouse FOXO6) (Jacobs et al., 2003). Unlike the other FOXO

isoforms, FOXO6 is mostly nuclear. However, FOXO6 phosphorylation at Thr26 and

Ser184 appears to decrease its transcriptional activity (van der Heide and Smidt, 2005),

which is similar to the effect of PKB on other FOXO factors transcriptional activity.

1.4.1.2. SGK and FOXO

SGKs (serum- and glucocorticoid-induced kinases) belong to a new family of

serine/threonine kinases that are regulated at both the transcriptional and

posttranslational levels by external stimulation. SGK mediates the biological effects of

PI3K in parallel with PKB. Although SGK is closely related to PKB, SGK and PKB

display unique features. First, SGK protein expression is induced by extracellular stimuli,

but PKB might not be. Second, SGK does not have a pleckstrin homology domain and

appears not to be recruited to the plasma membrane prior to its activation. Third, the

consensus sequence that is phosphorylated by SGK is not identical to the site

phosphorylated by PKB. Fourth, SGK, in contrast to PKB, is capable of phosphorylating

peptide substrates that do not have a bulky hydrophobic amino acid immediately C-

terminal to the phosphate-acceptor site (Kobayashi and Cohen, 1999).

Activated SGK promotes cell survival in part by phosphorylating and inactivating

FOXO3A. In the presence of DNA damage, caused by UV and irradiation treatment,

SGK is induced in a p53-dependent manner, leading to the phosphorylation of FOXO3A

(You et al., 2004a). SGK, like PKB, phosphorylates FOXO3A, thereby causing FOXO3A

translocation from the nucleus to the cytoplasm and inhibition of FOXO3A-dependent

transcription. However, SGK and PKB, when expressed at physiological levels, display

differences with respect to the efficacy with which they phosphorylate the regulatory

sites on FOXO3A. Specifically, Thr32 is phosphorylated by both protein kinases, but

SGK prefers Ser315 whereas PKB favors Ser253 (Brunet et al., 2001).

Page 37: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 21 -

1.4.1.3. IκB Kinase and FOXO3A

The IKK (IκB kinase) signaling pathway is a key survival and antiapoptotic

mechanism, aberrant expression of IKK has been implicated in constitutive activation of

NF-κB in human breast cancer cell lines and primary tumors. In breast cancer, IKK

physically interacts with, phosphorylates, and inhibits FOXO3A independently of PKB.

Cytoplasmic FOXO3A correlates with expression of IKKβ or phospho-PKB in many

tumors and associates with poor survival in breast cancer (Hu et al., 2004).

1.4.1.4. CK1 and FOXO

CK1 (Casein Kinase 1) is a serine/threonine protein kinase (Hathaway and

Traugh, 1979) that phosphorylates FOXO1 at Ser322 and Ser325 following the

phosphorylation of Ser319 by PKB or SGK. Multi-site phosphorylation of the region

containing Ser319, Ser322, Ser325 and Ser329 provides a signal for the nuclear

exclusion of FOXO1 (Rena et al., 2002).

1.4.1.5. CDK2 and FOXO

CDK2 phosphorylates FOXO1 at serine-249 (Ser249) in vitro and in vivo.

Phosphorylation of FOXO1 at Ser249 results in cytoplasmic localization of FOXO1 and

the inhibition of its transcriptional activity. This phosphorylation was abrogated upon DNA

damage through the cell cycle checkpoint pathway and was dependent on the protein

kinases Chk1 and Chk2 (Huang et al., 2006). Functional interaction between CDK2 and

FOXO1 provides a mechanism that regulates apoptotic cell death after double strand

DNA breakage.

1.4.1.6. JNK and FOXO

Besides IIS (insulin/IGF signaling) pathway, lifespan can also be increased by

activating the stress-responsive JNK (Jun-N-terminal kinase) pathway. In Drosophila,

Page 38: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 22 -

JNK requires FOXO to extend lifespan. JNK antagonizes IIS, causes nuclear localization

of FOXO and induces the expression of FOXO targets including the growth control and

stress defense genes. JNK and FOXO also restrict IIS activity systemically by repressing

IIS ligand expression in neuroendocrine cells (Wang et al., 2005).

In humans, an increase in ROS levels induces activation of the small GTPase

Ral, which in turn leads to the phosphorylation and activation of the stress kinase JNK.

Active JNK induces the phosphorylation of Thr447 and Thr451 on FOXO4.

Phosphorylation of these residues is essential for FOXO4 transcriptional activity.

Consistent with this, H2O2 treatment increases FOXO transcriptional activity and

translocation of FOXO4 from the cytoplasm to the nucleus and activation of the

transcription factor. Activation of FOXO4 through Thr447/451 phosphorylation can

induce transcription of MnSOD and catalase, leading to a decrease in ROS levels. Thus,

activation of FOXO4 by oxidative stress is part of a negative feedback loop to reduce the

levels of oxidative stress in a cell and to prevent damage to DNA, lipids and proteins

(Essers et al., 2004).

1.4.1.7. MST1 and FOXO3A

MST1 is a serine/threonine protein kinase that mediates cell death induced by

oxidative stress in primary mammalian neurons through direct activation of FOXO

transcription factors. MST1 phosphorylates FOXO proteins at a conserved site within the

FOX domain (Ser207) disrupting their interaction with 14-3-3 proteins, promoting FOXO

nuclear translocation, and thereby inducing cell death in neurons under stress conditions

such as hydrogen peroxide treatment. Knockdown of the C. elegans MST1 ortholog

CST-1 shortens lifespan and accelerates tissue aging, while overexpression of cst-1

promotes lifespan and delays aging. The cst-1-induced lifespan extension requires daf-

16 (Lehtinen et al., 2006).

Page 39: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 23 -

1.4.2. Acetylation and Deacetylation of FOXO

1.4.2.1. Acetylation and Deacetylation

Protein acetylation is a post-translational modification that transfers an acetyl

group to lysine residues. The best example is histone acetylation, which “opens”

chromatin’s structure and activates transcription. Deacetylation, on the other hand,

induces closed chromatin structure and repression of gene expression (Cohen and Yao,

2004). Protein acetylation is catalyzed by a group of acetyltranferases with different

specificities and target consensus sites. CBP/p300 (calcium response element-binding

(CREB)-binding protein), PCAF (p300/CBP-associated factor), members of p160 family,

Tip60, and TAFII250 are the main acetyltransferases involved in histone regulation and

transcription factor acetylation (http://www.chromatin.us/hatdesc.html). Proteins

acetylation is reversed by HDACs (histone deacetylases). There are three classes of

HDACs. Class I includes HDAC1, 2, 3, and 8. Class II includes HDAC4, 5, 6, 7, 9 and 10

which show high homology to HAD1 yeast deacetylase. There is one HDAC (HDAC11)

that shares homology with both class I and II HDACs. Class III HDACs (SIRTs) have an

absolute requirement for NAD in vivo and in vitro (de Ruijter et al., 2003; Imai et al.,

2000).

1.4.2.2. Acetylation of FOXO

1.4.2.2.1. P300 and CBP

p300 and CBP are large nuclear proteins encoded by two distinct genes.

p300/CBP have been implicated in numerous disease processes, including several

forms of cancer, cardiac hypertrophy and Huntington’s disease (Bandyopadhyay et al.,

2002; Borrow et al., 1996; Deguchi et al., 2003; Gayther et al., 2000; Gusterson et al.,

2003; Muraoka et al., 1996; Ross et al., 2002).

Page 40: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 24 -

The relative low abundance of p300 is rate-limiting in coactivation and

corepression of many transcription factors. Thus, p300 serves to integrate diverse

signaling pathways involved in metabolism, cellular differentiation and p53-mediated

apoptosis processes (Yao et al., 1998). Orchestration of these activities by p300

involves both a scaffolding function to tether transcription factors to target promoters and

its enzymatic activity as a HAT (histone acetyl transferase) (Bannister and Kouzarides,

1996; Ogryzko et al., 1996).

In addition to histones, several other non-histone proteins including transcription

factors are also acetylated by p300 (Goodman and Smolik, 2000), including p53,

estrogen receptor (Wang et al., 2001) and MyoD (Polesskaya et al., 2000). The activity

of p300 itself is also subjected to regulation via a number of post-translational

modification including phosphorylation, methylation, sumoylation and acetylation

(Banerjee et al., 1994; Chevillard-Briet et al., 2002; Girdwood et al., 2003; Thompson et

al., 2004; Yaciuk and Moran, 1991; Yadav et al., 2003).

In terms of differentiation, p300 and CBP appear to have numerous functions.

There is embryonic lethality of mice nullizygous for p300 (with defects in neurulation and

heart development) as well as in mice double heterozygous for p300 and CBP. Animals

which are nullizygous for p300 died between days 9 and 11.5 of gestation and showed

defects in neurulation, cell proliferation and heart development. Cells derived from p300-

deficient embryos displayed specific transcriptional defects and proliferated poorly.

Surprisingly, p300 heterozygotes also manifested considerable embryonic lethality.

Moreover, double heterozygosity for p300 and CBP was invariably associated with

embryonic death. Thus, mouse development is exquisitely sensitive to the overall gene

dosage of p300 and CBP (Yao et al., 1998). Further study in the mouse oocytes within

primordial follicles finds that p300 and CBP enter into the oocyte nucleus at different

Page 41: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 25 -

stages of oocyte growth. From the two-cell stage to the blastocyst stage in the pre-

implantation mouse embryos, the localizations of p300 and CBP are different, which

provide the information that p300 and CBP have different functions in early mouse

embryogenesis (Kwok et al., 2006).

1.4.2.2.2. P300/CBP and FOXO Acetylation

Using immunoprecipitation, an interaction between FOXO1, FOXO4, and

FOXO3A and CBP as well as between FOXO3A and p300 was found in vivo and in vitro

(Brunet et al., 2004; Daitoku et al., 2004; Fukuoka et al., 2003; Motta et al., 2004; van

der Horst et al., 2004).

CBP and p300 play different roles in the regulation of FOXO1 and FOXO4.

Mutational analysis show that FOXO1 and FOXO4 interact with CBP via their C-terminal

domain, whereas the N-terminal CH1 domain of CBP is required to bind FOXO1 and

FOXO4. CBP binds and acetylates FOXO1 at the K (lysine) 242, K245, and K262

residues. Acetylation at these residues by CBP attenuates FOXO1’s ability to bind

cognate DNA sequence and promotes the PKB-dependent phosphorylation of FOXO1

(Matsuzaki et al., 2005). CBP interacts with FOXO4 and acetylates it at K186, K189, and

K408. Acetylation by CBP inhibited the transcriptional activity of FOXO4 (Daitoku et al.,

2004; Fukuoka et al., 2003; van der Horst et al., 2004). However, in transient

transfection experiments acetylation by p300 in the C-terminal region of FOXO1 showed

that it potently stimulates FOXO1-induced transcription of IGF-binding protein-1. The

intrinsic acetyltransferase activity of p300 is required for both activities (Perrot and

Rechler, 2005).

As for FOXO3A, p300 and CBP can bind and acetylate it both in vivo and in vitro,

but two different studies show some discrepancies. One study shows that FOXO3A

binds p300/CBP at the N-terminus masking the first 52 amino acids at the N-terminus

Page 42: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 26 -

under serum starvation conditions (Mahmud et al., 2002). Another study shows that

CBP/p300 binds the C-terminus and forkhead domains of FOXO3A, leading to

acetylation at lysine residues K242, K259, K271, K290, and K569 (Brunet et al., 2004;

Mahmud et al., 2002). Overall, acetylation by p300 stimulates the transcriptional activity

of FOXO3A (Motta et al., 2004).

1.4.2.3. Deacetylation of FOXO by HDAC

1.4.2.3.1. SIRT1

The SIRT1 gene is located in chromosome 10q21.3. It contains 9 exons

(NC 000010). The 4,107 bp human Sirt1 mRNA has an open reading frame of 2,244 bp

and encodes a 747 aa protein with a predictive molecular weight of 81.7 kDa (kilodalton)

(Voelter-Mahlknecht and Mahlknecht, 2006). It belongs to the family of mammalian

sirtuins. The founding member of the sirtuin protein family was the silent information

regulator 2 (Sir2) of S. cervisiae (Saccharomyces cervisiae). Sirtuin proteins are highly

conserved from yeast to humans. In yeast, worms and flies, expression of Sir2 extends

longevity (Kaeberlein et al., 1999; Rogina and Helfand, 2004; Tissenbaum and

Guarente, 2001; Wood et al., 2004). In S. cervisiae, there are five sirtuin homologs,

Hst1-4 and Sir2. In mammals, there are seven sirtuins, SIRT1-7. SIRT1 is the closest

homolog of Sir2, plays important roles in diverse cellular processes including

transcriptional silencing, rDNA recombination, glucose metabolism and energy

homeostasis, DNA repair and cell survival. Due to its dependency on NAD+, the activity

of SIRT1 is regulated by NAD+/NADH ratio and thus sensitive to the redox status and

celluar metabolism. Similar to Sir2, SIRT1 is potentially a nutrient sensor that regulates

the lifespan of mammals in response to caloric restrication or nutrient starvation.

Page 43: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 27 -

Several compounds have been shown to inhibit or activate the deacetylase

activity of the Sir2 family. One inhibitor, splitomicin, is a cell-permeable lactone derived

from β-naphthol; it inhibits the NAD+-dependent deacetylase activity of Sir2 in vitro

(Bedalov et al., 2001). Another inhibitor, nicotinamide, a product of the Sir2 deacetylation

reaction, is an inhibitor of Sir2 activity both in vivo and in vitro. Nicotinamide has been

shown to inhibit a Sir2 homolog, SIRT1, a negative p53 regulator, promoting p53-

dependent apoptosis in mammalian cells (Luo et al., 2001; Vaziri et al., 2001). The most

potent activator was resveratrol, a polyphenol found in red wine, which is implicated in a

number of health benefits. In human cells, treatment with a low concentration of

resveratrol increased cell survival following DNA damage. Moreover, low resveratrol

concentrations decreased the acetylation of p53 at lysine residue 382, a known SIRT1

substrate; however, high resveratrol concentrations caused the opposite (Howitz et al.,

2003).

By deacetylating and inactivating the substrates such as p53 (Langley et al.,

2002; Vaziri et al., 2001), NFκB (nuclear factor-κB), FOXO (Brunet et al., 2004; Daitoku

et al., 2004; Motta et al., 2004; van der Horst et al., 2004), Ku70 (Jeong et al., 2007),

PPARγ (Peroxisome proliferator-activated receptor γ) (Picard et al., 2004), PGC-1α

(Balaban et al., 2005), AceCS2 (acetyl-CoA synthetase 2), p300 (Bouras et al., 2005), α-

tubulin, and HES1 and HEY2 (basic helix–loop–helix transcriptional repressors) (Takata

and Ishikawa, 2003), SIRT1 plays important roles in regulating various cellular

processes, including stress response (Vaziri et al., 2001), embryogenesis (McBurney et

al., 2003), metabolism (Leibiger and Berggren, 2006), calorie restriction (Brunet et al.,

2004), neuronal cell survival (Anekonda and Reddy, 2006; Bordone et al., 2006), insulin

secretion (Bordone et al., 2006) and aging (Chua et al., 2005).

Page 44: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 28 -

1.4.2.3.2. SIRT1 and FOXO Deacetylation

In mammalian cells, SIRT1 interacts with all the FOXO factors. Co-

immuoprecipitation experiments showed that SIRT1 interacts with FOXO1 in vivo.

Mutation analysis showed that the LxxLL motif in FOXO1 (amino acids 459-463) is

critical for the interaction with SIRT1. Mutagenesis of the LxxLL motif eliminates FOXO1

interaction with SIRT1, sustains the acetylated state of FOXO1 and makes FOXO1

insensitive to nicotinamide and resveratrol (Nakae et al., 2006). Yeast two hybridization

experiment showed that FHL2 (Four and a half LIM 2), which interacts with the N-

terminal of FOXO1 in vivo and in vitro, can enhance the interaction of FOXO1 and

SIRT1 (Yang et al., 2005).

Mammalian SIRT1 has dual effects on FOXO3A and FOXO4. SIRT1 deacetylates

FOXO3A, attenuating FOXO-induced apoptosis but potentiates FOXO-induced cell-cycle

arrest (Motta et al., 2004). SIRT1 and FOXO3A form a complex in cells in response to

oxidative stress, and SIRT1 deacetylates FOXO3A in vitro and in intact cells. The sites of

FOXO3A that appear to be primarily deacetylated by SIRT1 are K242, K245, and K262

(Daitoku et al., 2004). The SIRT1 and FOXO3A interaction requires PKB

phosphorylation of FOXO3A because constitutive FOXO3A, in which three PKB

phosphorylation sites were mutated to alanine, failed to bind SIRT1 under any stress

condition (Brunet et al., 2004; Motta et al., 2004). SIRT1 has a dual effect on FOXO3A

function: It increases FOXO3A's ability to induce cell cycle arrest and resistance to

oxidative stress, but inhibits FOXO3A's ability to induce cell death (Brunet et al., 2004).

In the case of FOXO4, acetylation by CBP inhibits its transcriptional activity.

SIRT1- mediated deacetylation precludes FOXO4 inhibition through acetylation

andthereby prolongs FOXO4-dependent transcription of stress-regulating genes. A

FOXO4 study revealed a molecular mechanism whereby SIRT1 can promote cellular

Page 45: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 29 -

T24 S249 S256 S319 S322 S325 S329 T32 S253 S315 S318 S321 S325 S644 T28 S193 S258 S261 S264 S268 T447 T451T26 S184

FOXO1P

PKB/SGK CK1 DYRK JNK IKKβ

PP P P P P PP

FOXO1FOXO3FOXO4FOXO6

Figure 2 The regulation of FOXO. Full-length FOXO and known

phosphorylation sites and acetylation sites are presented. K-lysine; T-threonine;

S-serine; T-tyrosine; CDK2-cyclin-dependent kinase 2; PKB- protein Kinase B;

SGK- serum and glucocorticoid responsive kinase; CK1- casine kinase 1;DYRK-

dual-specificity YAK-1-related kinase; JNK- c-Jun N-terminal kinase; IKKβ- IκB

kinase β. The numbers above the drawings denote amino acid numbers. Full-

length Mdm2 and known motifs are also shown.

1 150 368 377 461 466 596 655

CDK2P

FOX

K245 K248 K259 K265 K274 K294K242 K245 K259 K262 K271 K290 K569 K182 K185 K199 K211 K233 K403 K173 K176 K190 K202 K229

FOXO1FOXO3FOXO4FOXO6

A A A A A A

P300/CBP P300/CBP1 150 208 655

SIRT1461-466

FOXG1150 260

SHP401 652

Crm1368-377

14-3-324 256

AR350 655∗ ∗

P53

SMAD3/4150 260

FHL21

Page 46: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 30 -

survival and increase lifespan (van der Horst et al., 2004), a similar Sir2 function as

observed in worms.

1.4.3. Ubiquitination and Deubiquitination of FOXO

1.4.3.1. Ubiquitination and Degradation of FOXO

The degradation of FOXO transcription factors is mediated by the ubiqutin-

proteasome pathway. In the presence of insulin and other growth factors, FOXO proteins

are relocalized from the nucleus to the cytoplasm and to be degraded via the ubiquitin–

proteasome pathway. PKB activation is necessary for insulin promoted ubiquitin-

mediated degradation of FOXO1 and FOXO3A via the proteasome pathway (Aoki et al.,

2004; Plas and Thompson, 2003).

As for insulin induced proteasomal degradation of FOXO1, insulin treatment

decreases endogenous FOXO1 proteins in HepG2 cells and this decrease is suppressed

by proteasome inhibitors. FOXO1 is ubiquitinated in vivo and in vitro; insulin enhances its

ubiquitination in cells. In addition, the insulin signal to FOXO1 degradation is mediated

by the PI3K pathway. FOXO1 mutates at the serine or threonine residues that are

phosphorylated by PKB inhibit FOXO1 ubiquitination in vivo and in vitro. These data

suggest that efficient ubiquitination of FOXO1 requires both phosphorylation and

cytoplasmic retention in the cells (Matsuzaki et al., 2003).

Skp2 is an oncogenic subunit of the Skp1/Cul1/F-box protein ubiquitin complex. It

inhibits FOXO1 during tumor suppression through ubiquitin-mediated degradation but it

only ubiquitinates and degrades FOXO1, not FOXO3A or FOXO4. The effect of Skp2 on

FOXO1 requires PKB-specific phosphorylation of FOXO1 at Ser-256. Decrease

ubiquitination is seen in the mutant of FOXO1 in which all three PKB sites are replaced

by alanine and FOXO1 is forced to remain in the cytoplasm, through a mutation in the

Page 47: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 31 -

NLS. In addition, when phosphorylated FOXO1 is confined to the nucleus by a mutation

in the NES, FOXO1 ubiquitination is decreased (Huang et al., 2005). This study also

confirmed that only cytoplasmic FOXO1 is successfully ubiquitinated by an E3 ubiquitin

ligase and subsequently degraded.

IKKβ (I kappa B kinase β) also causes the proteasome-dependent degradation of

FOXO factors. IKKβ induces the phosphorylation of FOXO3A at Ser644, in the extreme

C-terminal portion of the molecule. This phosphorylation results in the ubiquitination and

subsequent degradation of FOXO3A. Since IKKβ-induced tumorigenesis can be

suppressed by overexpression of FOXO3A (Hu et al., 2004), the regulation of FOXO

protein degradation by IKKβ may play an important role in tumorigenesis. However,

Ser644 is not conserved in other FOXO isoforms and is not present in worms and flies. It

remains to be determined whether IKKβ phosphorylates and controls the other FOXO

isoforms. It is possible that the degradation of FOXO isoforms is regulated by different

protein kinases through other independent mechanisms.

Recently, it was showed that in the presence of oxidative stress FOXO4

becomes monoubiquitinated, resulting in its re-localization to the nucleus and increased

transcriptional activity (van der Horst et al., 2006). This study demonstrated that besides

FOXO degradation, the ubiqutination of FOXO may also have other biological functions.

1.4.3.2. Deubiquitination of FOXO

USP7/HAUSP is a herpes virus-associated ubiquitin-specific protease.

Deubiquitination of FOXO4 requires the enzyme USP7/HAUSP, which interacts with and

deubiquitinates FOXO4 in response to oxidative stress. Oxidative stress-induced

ubiquitination and deubiquitination by USP7 do not influence the half-life of FOXO4

Page 48: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 32 -

protein. Moreover, USP7 does negatively affect FOXO transcriptional activity towards

endogenous promoters (van der Horst et al., 2006).

2. Ubiquitin, Proteasome and MDM2 as an E3 Ligase

Ubiquitination is a reversible post-translational modification of cellular proteins, in

which ubiquitin is attached to the target proteins. Aberrations in the ubiquitination system

are implicated in the pathogenesis of many diseases, including certain malignancies,

neurodegenerative disorders and pathologies of the inflammatory immune response.

Deubiqutination is a biological process in which one or more ubiqutin moieties are

removed from protein. Deubiquitination helps to maintain the pool of free ubiquitin.

2.1. Ubiquitin-Proteasome System

2.1.1. Ubiquitin and Ubiqutination

Ubiquitin (Ub) is a small protein composed of 76 amino acids. Ub is found only in

eukaryotic organisms and not in either eubacteria or archaebacteria. Among eukaryotes,

ubiquitin is highly conserved. Ub is a heat-stable protein folded into a compact globular

structure. Ub is found throughout the cell and can exist either in free form or as part of a

complex with other proteins. In the latter case, Ub is conjugated to proteins through a

covalent bond between glycine at the C-terminal end of Ub and the side chains of lysine

on the target proteins. A single Ub can be conjugated to the lysine of these proteins, or

more commonly, Ub-chains can be attached. Uiquitination depends on ATP.

Ub is encoded by a family of genes whose translation products are fusion

proteins. The Ub genes typically exist in two states: 1) The ubiquitin gene can be fused

to a ribosomal gene giving rise to a translation product that is an Ub-ribosomal fusion

protein; or 2) Ub genes can exist as a linear repeat, meaning that the translation product

consists of a linear chain of Ub-molecules fused together (a polyubiquitin molecule).

Page 49: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 33 -

After the fusion proteins are synthesized, another protein called Ub-C-term hydrolase

cleaves the fusion proteins at the C-terminal end of Ub. This either liberates an individual

Ub and ribosomal protein or liberates a set of Ub monomers from the polyubiquitin.

Ubiquitination is a process in which ubiquitin is conjugated to the protein

substrates. The attachment of a single ubiquitin polypeptide, monoubiquitin, to a

substrate serves as an important regulatory modification (Hicke, 2001). Monoubiquitin

acts as a sorting signal throughout the endocytic pathway and regulates diverse proteins

including histones, endocytic machinery and transcription factors. Monoubiquitination

targets proteins to the lysosome, either by directing endocytosis of cell-surface receptors

or by sorting newly synthesized hydrolases from the Golgi to their resident lysosomal

compartment (Lee and Tyers, 2001). A polyubiquitin chain is formed when ubiquitin is

attached to a lysine within ubiquitin itself and this process is repeated. Polyubiquitin

chains linked through different lysine residues are involved in distinct cellular functions.

For instance, the signal necessary for degradation of substrates by the proteasome is a

polyubiquitin chain attached through Lys-48, whereas chains linked through Lys-63 are

crucial to the role of ubiquitin in DNA damage repair (Pickart, 2001). Polyubiquitination

typically targets proteins for rapid proteasomal degradation. Polyubiquitination is also

critical in protein quality control, where it helps to eliminate improperly processed or mis-

folded proteins from the ER (endoplasmic reticulum), in a process called ER-associated

degradation (Lee and Tyers, 2001).

2.1.2. Deubiquitination

Both poly- and monoubiquitination can be reversed by DUBs (deubiquitinating

enzymes) that specifically cleave the isopeptide bond at the C terminus of ubiquitin.

DUBs also generate the pool of free ubiquitin both by liberating ubiquitin from precursor

Page 50: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 34 -

ubiquitin fusion proteins and by recycling ubiquitin from the branched polyubiquitin

chains of degraded proteins (D'Andrea and Pellman, 1998; Wilkinson, 1997).

The DUBs are comprised of two groups of enzymes, the UCHs (ubiquitin C-

terminal hydrolyases) and the USPs (ubiquitin-specific proteases; referred to as UBPs in

yeast). The UCHs are small (20-30 kDa), closely related proteases that are generally

involved in cleaving ubiquitin from small processed peptides. The USPs are more

numerous, much larger (60–300 kDa), and are thought to have specific protein targets.

USPs can be identified by conserved sequences within the active site, but sequences

outside of the catalytic domain are highly divergent, likely reflecting their role in mediating

interactions with different protein targets (Hu et al., 2002; Wilkinson, 1997; Wilkinson et

al., 1995).

There are 16 known UBPs in Saccharomyces cerevisiae, and 63 putative USP

genes in humans. Single and even multiple UBP deletions in yeast generally produce

minimal phenotypic abnormalities, suggestive of functional redundancies among the

yeast UBP family (Amerik et al., 2000). However, studies have shown that USPs can

play specific roles in various biological processes in higher eukaryotes, suggesting a

more specialized role as cellular regulators in multicellular organisms. Specific DUBs

have been shown to regulate eye development (Huang et al., 1995; Huang and Fischer-

Vize, 1996), cell growth in response to cytokines (Zhu et al., 1996), oncogenic

transformation (Gray et al., 1995; Jensen et al., 1998; Papa and Hochstrasser, 1993),

cell cycle regulation (Hu et al., 2002), chromatin structure (Robzyk et al., 2000), and

transcriptional regulation (Holowaty et al., 2003; Mimnaugh et al., 1997; Moazed and

Johnson, 1996).

Page 51: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 35 -

2.1.3. Ubiquitination Machinery and Proteasome Pathway

Ubiquitination of a protein substrate requires the concerted action of three

classes of enzymes designated E1, E2, and E3. E1 (ubiquitin activating enzyme) initially

activates ubiquitin in an ATP-dependent reaction through the formation of a thiol-ester

bond between the carboxyl terminus of ubiquitin and the thiol group of a specific cysteine

residue of E1. Ubiquitin is then transferred to a specific cysteine residue on one of

several E2 (ubiquitin-conjugating enzymes, Ubcs). E2 enzymes in turn transfer the

ubiquitin either directly to a substrate or to the final class of enzymes known as E3

(ubiquitin protein ligases). E3 enzymes catalyze the formation of an isopeptide bond

between the carboxyl terminus of ubiquitin and the amino group of lysine residues on the

target protein. A substrate may then undergo multiply ubiquitinations through the

attachment of additional ubiquitin molecules to specific lysine residues of ubiquitin itself.

In many cases the E1, E2, and E3 enzymes form large, multi-protein complexes. This

increases the efficiency of the process by allowing the rapid thiol-ester transfer of

ubiquitin molecules between proteins.

This type of protein degradation plays a role in many cellular processes, such as

cell cycle regulation, antigen presentation, and the disposal of denatured, unfolded, or

oxidized proteins. Most intracellular protein degradation is through the ubiquitin-

proteasome pathway (Ciechanover and Schwartz, 2002; Hershko and Ciechanover,

1998).

In the ubiquitin-proteasome degradation pathway, the covalent attachment of

multiple ubiquitin molecules to lysine residues of a target protein serves to signal its

recognition and rapid degradation by the 26S proteasome. The proteasome is a large,

multisubunit complex that exists in cells in two forms: a 20S and a 26S species. The

Page 52: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 36 -

Figure 3 Ubiquitin-proteasome degradation pathway. E1 (ubiquitin

activating enzyme) activates Ub in the presence of ATP. Activated Ub is then

transferred to E2 (ubiquitin conjugating enzyme). E2 in turn transfers Ub to E3

(ubiquitin protein ligase). E3 binds ubiquitin to the substrate protein.

Ubquitinated proteins are degraded by the proteasome. Ub – ubiquitin.

Page 53: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 37 -

active protease sites sequestered in its central cavity, so that only proteins entering this

chamber are degraded. The openings to this cavity permit only denatured proteins to

enter, where they are progressively cleaved to small peptides. The addition of a 19S

regulator to either or both ends of the 20S proteasome creates the 26S proteasome. The

19S regulator contains ATPases and other proteins and serves numerous functions,

including the recognition of the substrate and its translocation to the catalytic core

(Voges et al., 1999).

2.2. MDM2 as an Ubiquitin E3 Ligase

2.2.1. General Information about MDM2

MDM2 is the product of the 'murine double minute 2' gene. The MDM2 gene was

originally identified as one of three genes (mdm1, 2, 3) which were overexpressed by

more than 50-fold through amplification in a spontaneously transformed mouse BALB/c

cell line (3T3-DM). The mdm2 genes are located on small, acentromeric

extrachromosomal nuclear bodies, called double minutes, which are retained in cells

only if they provide a growth advantage. The gene product of mdm2 was later shown to

be responsible for cell transformation when MDM2 was overexpressed (Cahilly-Snyder

et al., 1987; Fakharzadeh et al., 1991). In 1992, Oliner cloned the human MDM2 gene

and mapped it to chromosome 12q13-14 (Oliner et al., 1992). Both the mdm2 gene and

its human counterpart, MDM2, consist of 12 exons that can generate many different

MDM2 proteins, as shown in Figure 4. There are two different promoters, the second of

which is responsive to p53. These promoters generate two proteins, the full-length p90

and a shorter p76 protein that initiates at an internal ATG (Olson et al., 1993; Perry et

al., 1993; Saucedo et al., 1999). p76 is missing part of the p53-binding domain and it can

act as a dominant negative inhibitor of p90 and activate p53. Alternative splicing of

Page 54: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 38 -

MDM2 to generate shorter proteins also occurs in many human and mouse tumors. More

than 40 different alternatively spliced transcript variants have been isolated from both

tumor and normal tissues. In humans, MDM2-a and MDM2-b are the major splice

variants that delete exons 4–9 and 4–11, respectively. Neither product contains the p53-

binding motif. MDM2-b, also named MDM2-ALT1, interacts with full-length MDM2 and

sequesters it in the cytoplasm (Bartel et al., 2002).

2.2.2. Functions of MDM2

2.2.2.1. MDM2 and Cell Cycle

Overexpression of MDM2 has been shown to correlate with the cyclin-dependent

kinase inhibitor p21. In breast cancer cells, overexpression of MDM2 correlates with lack

of p21 expression (Jiang et al., 1997). On the other hand, in squamous cell carcinoma,

overexpression of MDM2 is associated with high levels of p21 (Ng et al., 1999).

MDM2 reverses the growth inhibition at G1 imposed by p53 and RB (Chen et al.,

1996; Xiao et al., 1995). Overproduction of MDM2 can overcome the TGF-β-imposed

growth inhibition via the RB–E2F pathway (Sun et al., 1998).

Transgenic mice experiments showed that expression of a BLG (β-

lactoglobulin)/mdm2 transgene (BLGmdm2) in the epithelial cells of the mouse

mammary gland caused mammary epithelial cells to undergo multiple rounds of S phase

without cell division, and resulted in polyploidy and tumor formation. The effect of MDM2

on S phase is independent of the p53 status (Lundgren et al., 1997).

2.2.2.2. MDM2 and Differentiation

One phenotype of tumor cells is the lack of terminal differentiation. MDM2 plays a

very important role in epidermal differentiation (Dazard et al., 1997). MDM2

overexpression in the granular layer perturbs the differentiation program (Alkhalaf et al.,

Page 55: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 39 -

1999). In rhabdomyosarcoma, forced expression of MDM2 inhibits MyoD function and

consequently inhibits muscle differentiation (Fiddler et al., 1996). A MDM2-conditional

mice experiment showed that MDM2 plays a very important role in bone organogenesis

and homeostasis through inhibition of p53 function, which is a prerequisite for master

osteoblast transcriptional regulator Runx2 activation, osteoblast differentiation, and

proper skeletal formation (Lengner et al., 2006). NUMb is a cell fate determinant protein.

MDM2 associates with NUMb and influences cell differentiation and survival through

translocation of NUMb to nucleus and degradation of NUMb (Juven-Gershon et al.,

1998).

However, microinjection of MDM2 mRNA in two-cell stage zebrafish embryos

caused inhibition of cellular convergence during gastrulation. Clones derived from MDM2

microinjected blastomeres were significantly smaller than those derived from control

microinjections. This indicates that MDM2 expression may be important during the

differentiation of neural and muscular tissues of zebrafish (Thisse et al., 2000).

2.2.2.3. MDM2 and Ribosome Biogenesis

Ribosome biogenesis is the process of making ribosomes. It takes place both in

the cell cytoplasm and in the nucleolus of eukaryotic cells. It involves the coordinated

action of over 200 proteins in the synthesis and processing of the four rRNAs, as well as

assembly of those rRNAs with the ribosomal proteins. Proper ribosome assembly is

essential for the health of a cell.

Ribosome proteins L5, L11 and L23 exist in the same complex with MDM2 in

response to ribosome stress, such as exposure to actinomycin D. They activate p53 by

inhibiting MDM2-mediated p53 suppression (Dai et al., 2004; Lohrum et al., 2003;

Marechal et al., 1994; Zhang et al., 2003).

Page 56: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 40 -

2.2.2.4. MDM2 and Transcription

MDM2 affects the gene transcription by inhibiting p53-mediated transactivation

(Momand et al., 1992). MDM2 uses multiple mechanisms to inactivate p53 and to inhibit

its transcriptional activity. MDM2 targets p53 for ubiquitination and degradation by the

proteosome, shuttles p53 out of the nucleus, prevents its interaction with transcriptional

coactivators and recruits the known transcriptional corepressors, such as hCtBP2, to

p53.

2.2.2.5. MDM2 and Protein Ubiquitination and Degradation

E3 ubiquitin ligases are a large family of proteins engaged in the regulation of the

turnover and activity of many proteins. Together with ubiquitin-activating enzyme E1 and

ubiquitin-conjugating enzyme E2, E3 ubiquitin ligases catalyze the ubiquitination of a

variety of biologically significant protein substrates leading to their degradation through

the 26S proteasome. Because they serve as the specific substrate-recognition element

of the system, E3 ligases play an important role in the ubiquitin-mediated proteolytic

cascade. There are approximately 1000 E3 ligases in the human genome.

MDM2 possesses the activity of an E3 ubiquitin ligase. MDM2 was initially found

to promote the proteasome–dependent degradation of p53 (Haupt et al., 1997; Honda et

al., 1997; Kubbutat et al., 1997). It functions as an E3 ubiquitin ligase for p53 and for

itself through its RING finger domain at the C-terminus (Fang et al., 2000; Honda et al.,

1997; Honda and Yasuda, 2000). It is now known that MDM2 also promotes the

degradation of several other proteins in intact cells, such as: Numb (Yogosawa et al.,

2003), RB (Miwa et al., 2006; Uchida et al., 2005) and MDMX (Pan and Chen, 2003).

Page 57: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 41 -

2.2.3. Regulation of MDM2 E3 Activity

2.2.3.1. Sumoylation of MDM2

2.2.3.1.1. SUMO and Sumoylation

SUMOs (small ubiquitin-related modifiers) constitute a family of highly conserved

proteins found in all eukaryotes and are required for viability of most eukaryotic cells,

including budding yeast, nematodes, fruit flies, and vertebrate cells in culture

(Apionishev et al., 2001; Epps and Tanda, 1998; Fraser et al., 2000; Hayashi et al.,

2002; Jones et al., 2002). In multicellular organisms, SUMO conjugation takes place in

all tissues and at all developmental stages (Chen et al., 1998; Howe et al., 1998;

Joanisse et al., 1998; Kamitani et al., 1998; Kurepa et al., 2003; Lois et al., 2003;

Mannen et al., 1996; Shen et al., 1996). Since its discovery in 1996, SUMO has been

found covalently attached to more than 50 proteins, including the androgen receptor,

IκBα, c-Jun, HDACs and p53. Proteins that participate in transcription, DNA repair,

nuclear transport, signal transduction and the cell cycle have been found to be

sumoylated. In contrast to ubiquitination, however, sumoylation of a protein does not

appear to target it for rapid degradation, but rather affect the ability of the modified

protein to interact with cellular factors. Most SUMO-modified proteins that have been

characterized in mammalian systems are involved in transcription and they are often

repressed by SUMO conjugation. However, genetic studies in model organisms have

pointed to a role for SUMO in chromosome dynamics and higher order chromatin

structures, illustrating the diversity of SUMO function.

SUMO often has a positive effect on protein-protein interactions, and it promotes

assembly of several multi-protein complexes. However, the effects of SUMO on

interactions vary depending on the substrates. SUMO can also act by a completely

Page 58: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 42 -

different mechanism, including the prevention of ubiquitination of a protein by blocking

lysine residue where Ub would normally be attached (Desterro et al., 1998; Hoege et al.,

2002; Lee et al., 2003; Lin et al., 2003a).

The linkage between SUMO and its substrates is an isopeptide bond between

the C-terminal carboxyl group of SUMO and the ε-amino group of a lysine residue in the

substrate. A three-step enzyme pathway attaches SUMO to specific substrates, and

other enzymes cleave SUMO off its targets. The enzymes of the SUMO pathway,

although analogous to those of the Ub pathway, are specific for SUMO and have no role

in conjugating Ub or any of the other ubiquitin-like modifiers. The SUMO pathway begins

with a SUMO-activating enzyme (also called E1). E1 catalyses an ATP-dependent

activation of the SUMO C-terminus and then transfers activated SUMO to a SUMO-

conjugating enzyme (E2), also known as Ubc9. SUMO is then transferred from Ubc9 to

the substrate with the assistance of one of several SUMO-protein ligases (E3s). In

contrast to the ubiquitin-conjugating system, where E3 ligase is responsible for target

recognition, the recognition of SUMO targets is mediated by both E2 and E3 enzymes.

Many of the Lys residues where SUMO becomes attached are in the short consensus

sequence ψΩKXE/D, where ψ denotes a bulky aliphatic residue, Ω denotes a large

hydrophobic amino acid, generally isoleucine, leucine, or valine; K is the lysine residue

being modified; X is any residue; and E/D is a glutamic or aspartic acid. This motif is

bound directly by Ubc9. E3 ligases probably enhance specificity by interacting with other

features of the substrate. Although most known SUMO targets contain this sequence,

other conjugation sites are now beginning to be known, such as TKET in S. cerevisiae

PCNA (Hoege et al., 2002) and VKYC in Smad4 (Lee et al., 2003; Lin et al., 2003b).

Sumoylation is a reversible modification, and removal of SUMO is carried out by

enzymes that specifically cleave at the C-terminus of SUMO (Johnson, 2004). All known

Page 59: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 43 -

SUMO-cleaving enzymes belong to the family of ubiquitin like protease 1 (Ulp1) cysteine

proteases and contain a 200 amino acid C-terminal core domain (the Ulp domain). The

core domain has the SUMO cleaving activity and contains the catalytic triad Cys-His-Asn

(Mossessova and Lima, 2000). Mammals have seven members of the Ulp1 family:

SENP1-3 and SENP5-8. Only four of the SENP genes have been confirmed to encode

SUMO proteases, namely SENP1 (Bailey and O'Hare, 2002), SENP3 (SMT3IP1)

(Nishida et al., 2000), SENP6 (SUSP1) (Kim et al., 2000) and SENP2 (Axam,

SMT3IP2/Axam2, SuPr-1) (Best et al., 2002; Kadoya et al., 2002; Nishida et al., 2001).

2.2.3.1.2. Sumoylation and MDM2

SUMO-1 modification of MDM2 can differentially modulate the outcome of MDM2

E3 ligase activity in a manner that favors accmulation of p53. Upon Sumo-1 conjugation,

MDM2 is protected from self-ubiquitination and elicits greater ubiquitin-protein isopeptide

ligase (E3) activity toward p53, thereby increasing its oncogenic potential. This switch in

modification status is stress-responsive, because UV irradiation leads to a decrease in

the interaction of MDM2 with Ubc9 and a corresponding loss of MDM2 sumoylation

(Buschmann et al., 2001). Further studies showed that Ubc9 can associate with MDM2

only if amino acids 40-59 within the N-terminus of MDM2 are present. Furthermore,

addition of a peptide corresponding to amino acids 40-59 of MDM2 efficiently inhibits

MDM2 sumoylation in vitro and in vivo (Buschmann et al., 2001)

2.2.3.2. Ubiqutination and Degradation of MDM2

As mentioned earlier, MDM2 mediates autoubiquitination as well as the

ubiquitination of other substrates. The balance between auto- and substrate-

ubiquitination of MDM2 is modulated physiologically by posttranslational modifications,

including sumoylation and phosphorylation. After SUMO conjugation to MDM2, MDM2

Page 60: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 44 -

E3 ligase activity is shifted toward p53, while self-ubiquitination is minimized

(Buschmann et al., 2001).

P300 is an acetylase-possessing transcriptional co-activator that has been

shown to mediate transcription by numerous transcriptional activators. It binds to and

stabilizes MDM2. It stabilizes MDM2 by retaining it in a specific nuclear structure but

does not acetylate MDM2 in solution or in cells.

2.2.3.3. Phosphorylation of MDM2

The first demonstration of the complex nature of MDM2 phosphorylations was by

Henning who showed that the phosphorylation status of MDM2 was influenced by early

gene expression of SV40. MDM2 is stabilized in the presence of SV40. Moreover,

hyperphosphorylated MDM2 participates in a trimeric complex with p53 and T-Ag (T-

antigen, the transforming protein of SV40), which is thought to activate oncogenic

functions of MDM2 and enhance the transforming potential of T-Ag (Henning et al.,

1997).

Nearly 20% of the residues of MDM2 are either serine or threonine. MDM2

protein is phosphorylated at multiple sites in vivo. Two clusters of phosphorylation sites

are located at the N-terminal (amino acids 1–193) and central amino acids 194–293 of

murine Mdm2, respectively (Hay and Meek, 2000).

2.2.3.3.1. DNA-PK, ATM and MDM2

The PI3K family of enzymes generates lipid 'second messengers' that mediate

signal transduction. It includes four classes of proteins. Class IV of PI3K includes mTOR

(mammalian target of rapamycin), DNA-PK (DNA activated protein kinase), ATM (ataxia

telangiectasia-mutated) and ATR (ATM and Rad3-related) protein kinases. MDM2 is

phosphorylated in vitro by both DNA-PK (Mayo et al., 1997) and ATM (Maya et al., 2001)

Page 61: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 45 -

but phosphorylation by ATR has not yet been reported. Of eight potential DNA-PK

targets in MDM2, only Ser17 has been shown to be phosphorylated by this enzyme in

vitro (Mayo et al., 1997). Although physiological phosphorylation of Ser17 has been

confirmed, the phosphorylation site itself has been reported to have a significant impact

on the ability of MDM2 to regulate the response to p53. S17A mutant, mimics the

dephosphorylation form of MDM2, was significantly more effective in inhibiting p53-

dependent transactivation in cultured cells than wild-type MDM2. Nuclear magnetic

resonance studies also showed that MDM2 amino acids 16–24 can form a "flexible lid"

that folds over and stabilizes the MDM2 structure but competes only weakly with p53 for

binding to this cleft. The S17D mutant peptide which mimics the consititutive

phosphorylation form was found to have higher affinity for MDM2 than the wild-type

peptide (McCoy et al., 2003).

ATM is able to phosphorylate MDM2 at Ser395 in vitro. In response to ionizing

radiation and radiomimetic drugs, MDM2 undergoes rapid ATM-dependent

phosphorylation prior to p53 accumulation, which results in a decrease in its reactivity

with the 2A10 monoclonal antibody. MDM2 S395D is impaired in promoting p53

degradation and it is markedly less able to promote p53 cytoplasmic export (Maya et al.,

2001).

2.2.3.3.2. PKB and MDM2

Mitogen-induced activation of PKB results in phosphorylation of MDM2 on Ser166

and Ser186. Phosphorylation on these sites is necessary for translocation of MDM2 from

the cytoplasm into the nucleus (Mayo and Donner, 2001). Moreover, phosphorylation of

MDM2 not only enhances its nuclear localization but its interaction with p300, and

inhibits its interaction with p19ARF, resulting in increased p53 degradation (Zhou et al.,

2001).

Page 62: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 46 -

PKB inhibits MDM2 self-ubiquitination via phosphorylation of MDM2 on Ser166

and Ser188. Stimulation of human embryonic kidney 293 cells with IGF-1 increased

MDM2 phosphorylation on Ser166 and Ser188 in a PI3K-dependent manner. Treatment

of both human embryonic kidney 293 and COS-1 cells with PI3K inhibitor LY-294002 led

to proteasome-mediated MDM2 degradation (Feng et al., 2004).

2.2.3.3.3. c-Abl and MDM2

c-Abl is an non-receptor tyrosine kinase that can shuttle between the cytoplasmic

and nuclear compartments. In response to stress, such as DNA damage, c-Abl promotes

cell growth arrest and apoptosis. The apoptotic activity of c-Abl is mediated partly via

p73 (Agami et al., 1999; Gong et al., 1999) and to a lesser extent through a p53-

dependent pathway (Yuan et al., 1997). One mechanism for the protection of p53 by c-

Abl is that c-Abl can neutralize the ability of MDM2 to ubiquitinate p53 and degrade it

(Sionov et al., 1999). C-Abl directly interacts with MDM2 at multiple sites in the nucleus,

enhances its accumulation in a p53-independent manner. c-Abl phosphorylates MDM2

at Tyr394. Substitution of Tyr394 by Phe394 enhances the ability of MDM2 to promote

p53 degradation and to inhibit the transcriptional and apoptotic activities of p53

(Goldberg et al., 2002).

2.2.3.3.4. CK2 and MDM2

Protein kinase CK2 is a ubiquitous Ser/Thr protein kinase required for cell cycle

progression and cell viability. Serine residue at position 269 of MDM2 was established

as the most important CK2 phosphorylation site by analyses with deletion mutants of

MDM2 and a peptide library. Phosphorylation of MDM2 by CK2 is stimulated in the

presence of the C-terminal sequences of p53, but binding studies revealed that the

Page 63: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 47 -

I II III IV V VI VII VIII IX X XI XII

1 27 28 52 53 97 97 114 114 133 134 166166 220 221 272 273 298 299 491

P1 P2 ATG ATG

1 26 108 178 192 230 274 289 331 433 467 471 491

Zn-finger

NLS NES Ring-fingerAcidic domain

Zn-finger

NLS NES Ring-fingerAcidic domain

P P P PP PP NoLS

MDM2

P300

P5326 108

102 222 341 491

L5,L111 222 284 374

ARF210 224

P53 211 361

SP1,RB221 294

Numb1 134

PCAF50 384

AR

P

1 27 223 491

1 27 301 491MDM2-a

MDM2-b

MTBP 167 304

PML 50 166 202 304 340 488

PML

E2F, TFIIE

DNA polymerase ε

p731 150

1 52 223 491

1 27 389 491MDM2-c

MDM2-d1 75 483 491

MDM2-e

S17 S166 S186 S188 S269 S395DNA-PK PKB/AKT CK2 ATM

T216 Y394CyclinA/CDK2 c-Abl

Figure 4 Structure of MDM2 gene and protein

Page 64: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 48 -

Figure 4 Structure and regulation of MDM2 gene and protein. MDM2 gene

consists of 12 exons. Two promoters are shown by arrows. Full-length MDM2 p90

is translated from the first start codon ATG in exon 3 and the short form, p76, is

translated from the second ATG in exon 4. Phosphorylation sites are indicated by

the letter P within an ellipse. Their locations relative to the functional domains of

MDM2 are shown. Protein kinases, where known, are indicated in boxes with the

target residue(s) shown above. Five major alternative splice variants MDM2-a,

MDM2-b, MDM2-c, MDM2-d, and MDM2-e are shown. Full-length Mdm2 and

known motifs are also represented. NLS-nuclear localization signal; NES-nuclear

export signal; Zn-finger-zinc finger domain; NoLS-nucleolar localization signal;

RING-finger-ring finger domain. The numbers above the drawings denote amino

acid numbers and roman numerals denote the exon numbers.

Page 65: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 49 -

biological function of CK2 phosphorylation still needs to be established by further

studies because phosphorylation of MDM2 at Ser269 does not have any influence on

the binding of p53 to MDM2 (Gotz et al., 1999).

3. Functional interaction among FOXO, p53, and MDM2

3.1. Interaction between MDM2 and p53

P53 is a transcription factor that regulates the cell cycle and functions as a tumor

suppressor. p53 has been described as "the guardian of the genome" or the "master

watchman", referring to its role in conserving stability by preventing genome mutations.

In the absence of genetic damage, p53 is a very unstable protein with a half-life ranging

from 5-30 min and transcriptional activity is inert (Yuan et al., 1996). In the presence of

stress, such as DNA damage, hypoxia, telomere shortening, and oncogenic activation,

p53 becomes stable and activated by blocking its degradation (Caspari, 2000; Meek,

1994; Sakaguchi et al., 1998; Siliciano et al., 1997). p53 can kill cells via dual

transcription-dependent and -independent functions in the nucleus and mitochondria

(Mihara et al., 2003; Vousden and Lu, 2002).

The p53-MDM2 system forms a feedback loop, in which p53 upregulates MDM2

by activating MDM2 transcription (Barak et al., 1993). Experiments with knock-out mice

revealed that deletion of the mdm2 gene results in embryonic lethality, which can be

rescued by deletion of the p53 gene (Jones et al., 1995; Montes de Oca Luna et al.,

1995). Inhibition of cell growth and marked cell death is often seen in the absence of p53

regulation by MDM2, further emphasizing the importance of the p53–MDM2

autoregulatory loop in controling of cell growth and death.

MDM2 negatively regulates p53 in several ways:

Page 66: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 50 -

1) MDM2 binds to p53 and this interaction is conformation based. Site-directed

experiments have demonstrated the importance of p53 residues Leu14, Phe19, Leu22,

and Trp23 (Lin et al., 1994). A minimal MDM2-binding site on p53 residues 18-23 was

mapped with p53-derived peptides (Picksley et al., 1994). On binding to the p53

transactivation domain, MDM2 inhibits its transcriptional activity. Crystallographic data

showed that the amino terminal domain of MDM2 forms a deep hydrophobic cleft into

which the transactivation domain of p53 binds, thereby concealing itself from interacting

with the transcriptional machinery (Kussie et al., 1996).

2) MDM2 functions as an E3 ubiqutin ligase for p53 (Honda and Yasuda, 1999;

Lohrum et al., 2000; Tao and Levine, 1999; Weber et al., 1999). The level of MDM2 is

very important for the ubiquitination level of p53. In vitro studies showed that low levels

of MDM2 activity induce monoubiquitination, whereas high levels promote

polyubiquitination and nuclear degradation of p53 (Li et al., 2003a).

3) MDM2 promotes the export of p53 from the nucleus. p53 shuttles between

nucleus and cytoplasm in the cells in response to stress. MDM2 contains an NES, but

the study showed that the MDM2 RING finger domain, not the MDM2 NES, is necessary

for the efficient export of p53 to the cytoplasm (Boyd et al., 2000; Geyer et al., 2000).

Another study also showed that low MDM2 levels induce cytoplasmic translocation of

p53 (Li et al., 2003a), whereas MDM2-mediated p53 monoubiquitylation promotes its

mitochondrial translocation (Marchenko et al., 2007).

3.2. Interaction between p53 and FOXO

p53 and FOXO factors share similar characteristics. Both are involved in stress

response and can be post-translationally modified by phosphorylation and acetylation.

They also have some common downstream targets, such as: Fas ligand (Greer and

Page 67: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 51 -

Brunet, 2005), GADD45 (Greer and Brunet, 2005), PA26 (Greer and Brunet, 2005), p21

(Seoane et al., 2004) and PUMA (You et al., 2006a) (Figure 5).

Recently it was shown that p53 and FOXO3A interact with each other. In

response to oxidative stress, p53 binds to FOXO3A. In vivo, these two transcription

factors exhibit “crosstalk”. In response to DNA damage, p53 activation leads to FOXO3A

phosphorylation and subcellular localization change, which results in inhibition of

FOXO3A transcription activity. PKB is dispensable for p53-dependent suppression of

FOXO3A. By contrast, SGK1 was significantly induced in a p53-dependent manner after

DNA damage, and this induction is through extracellular signal-regulated kinase 1/2-

mediated posttranslational regulation. Furthermore, inhibition of SGK1 expression by a

small interfering RNA knockdown significantly decreased FOXO3A phosphorylation in

response to DNA damage.

Nuclear activated FOXO3A can impair p53 transcriptional activity. However,

activation of FOXO3A either by serum starvation or by expressing a constitutively active

form of FOXO3A can induce p53-dependent apoptosis, even in cells bearing a

transcriptionally inactive form of p53 (You et al., 2006b).

Page 68: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 52 -

DNA damageOncogenactivation

OxidativeStress

FOXOs

P21

P27

GADD45

Fas Ligand

Bim PUMA

TRAIL

Cell cycle Arrest Apoptosis

DNA damageOncogenactivation

Hypoxia

p53

P21

14-3-3δ

GADD45

Fas Ligand

PTEN PUMA

BAX Noxa

Cell cycle Arrest Apoptosis

Figure 5 Stress-induced FOXO and p53 pathway.

Page 69: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 53 -

HYPOTHESIS & OBJECTIVES

FOXO factors are known to be ubiquitinated, but so far no general E3 ubiquitin

ligases capable of ubiquitination of all FOXO factors have been identified. Skp2 is

reported to promote the ubiqutination and degradation of FOXO1, but this effect is

limited to FOXO1. A genetics study in C.elegans showed that skp expression is actually

needed for FOXO transcriptional activity.

Since MDM2 is known to be an E3 ubiquitin ligase for p53 and both p53 and

FOXO factors are important regulators in stress responses, aging and tumorigensis, we,

therefore, hypothesize that MDM2 interacts with and promotes the ubiquitination and

degradation of FOXO factors.

To substantiate the hypothesis, my thesis studies are to achieve the following

objectives:

1. Determine whether FOXO factors and MDM2 interact;

2. Determine whether MDM2 regulates the transcriptional activity of FOXO

factors;

3. Determine whether MDM2 decreases the stability of FOXO factors;

4. Determine whether MDM2 changes the biological function of FOXO;

5. Identify the signals that regulate effect of MDM2 on FOXO and investigate

the functional relationship among p53, FOXO and MDM2.

Page 70: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 54 -

MATERIALS AND METHODS

Chemicals, Antibodies and Cell Lines:

Cycloheximide and nicotinamide were purchased from Sigma and MG132 from

Calbiochem. Antibodies against FOXO1 (H-128, Santa Cruz Biotechnology), FOXO3A

(H-144, Santa Cruz Biotechnology), MDM2 (SMP14, Santa Cruz Biotechnology; 2A10,

Calbiochem), c-Myc (A-14, Santa Cruz Biotechnology), Flag (M2-A-2220 and F-7452,

Sigma), HA (MMS-101P and PRB-101P, Covance), MnSOD (Upstate), TRAIL (BD

Pharmingen), p27 (N-20, Santa Cruz), acetyl-K (Upstate), α-Tubulin (Sigma), and β-actin

(AC-74, Sigma) were purchased from commercial sources.

H1299/V138 cells were cultured as described (Pochampally et al., 1999). HEK

293T, NIH 3T3, DU145, JCA1, PC3, HeLa, MCF-7, Saos-2, H1299, p53 null MEFs

(mouse embryonic fibrblasts )and p53 and MDM2 double null MEFs (Huang et al., 2005;

Peng et al., 2001) were maintained in DMEM (Dulbecco's modified Eagle's medium) with

10% FBS (fetal bovine serum). LNCaP cells were cultured in RPMI 1640 with 10% FBS

at 37°C. H1299 cells stably expressing human MDM2 were generated by cotransfecting

MDM2 with pcDNA3 and selecting in the presence of 750 μg/ml G418.

Plasmids:

pcDNA3-Flag-FOXO1, pcDNA3-Flag-FOXO1 (AAA), HA-FOXO3A, HA-FOXO3A

(AAA) (Li et al., 2003b), HA-SIRT1 (Yang et al., 2005), HA-SIRT1(734R) (Yang et al.,

2007), pcMDM2 and different MDM2 mutants were described previously (Armoni et al.,

Page 71: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 55 -

2006; Chen et al., 1995; Chen et al., 1993; Freedman and Levine, 1999). To construct

pcDNA3-HA-FOXO1 (1-150), FOXO1 cDNA fragment coding the first 150 amino acids

were amplified by PCR with primers 5'-CGG GGG TCA CCG GAT CCA TGG CCG AGG

C-3' and 5' –GCG GCG GGA CGA TCT AGA CTA GCG CGG CTG C-3', which

generated BamHI and XbaI sites at 5' and 3' ends of the DNA fragment, respectively.

The amplified FOXO1 (1-150) fragment was cloned into the BamHI and XbaI sites of

pcDNA3.1 HA vector (Invitrogen). pcDNA3-HA-FOXO1 (1-270) and pcDNA3-HA-FOXO1

(256-655) were constructed similarly by generating a BamHI site at the 5' end and a XbaI

site at the 3' end of the corresponding FOXO1 cDNA fragments. The upstream primer of

FOXO1 (1-270) was same as the FOXO1 (1-150), and the downstream primer was 5'-

CTT GGC TCT AGA AGC TCG GCT TCG GCT CTT AG -3'. The upstream primer of

FOXO1 (256-655) was 5'-GGA GAA GAG CTG GAT CCA TGG ACA ACA AC-3' and the

downstream primers was 5'-CGG GCC CTC TAG ATC AGC CTG ACA CC -3'. MDM2

siRNAs were subcloned into pSilencer-Neo (Ambion). The corresponding

oligonucleotides for generating the MDM2 siRNA were 5'-GAT CCG CAG GTG TCA

CCT TGA AGG TTT CAA GAG AAC CTT CAA GGT GAC ACC TGT TTT TTG GAA A-

3'and 5'-GAT CCG TGG TTG CAT TGT CCA TGG CTT CAA GAG AGC CAT GGA CAA

TGC AAC CAT TTT TTG GAA A-3'. The oligonucleotides for GFP siRNA were from

Ambion.

Transfections and Immunological Assays:

For co-precipitation analysis, 106 cells were plated in 100 mm dishes in a

medium containing 10% fetal bovine serum. One day after plating, cells were transfected

with the indicated plasmids by Lipofectamine Plus following the protocol from Invitrogen.

Cellular extracts were prepared in a buffer containing 20 mM Tris-HCl (pH 7.5), 0.5% NP-

40, 250 mM NaCl, 3 mM EDTA, 3 mM EGTA, 10 µg/ml aprotinin, 10 µg/ml leupeptin, 10

Page 72: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 56 -

mM benzamidine and 1 mM phenylmethylsulfonyl fluoride. After pre-clearance by

incubating with protein A-agarose for 1 hour (h) followed by brief centrifugation, the

extracts were incubated sequentially with 1-3 µg antibody and protein G-agarose beads

for 4 h at 4°C. After four times washes with the lysis buffer, the immunoprecipitates were

eluted from the beads by boiling in sodium dodecyl sulfate-polyacrylamide gel

electrophoresis (SDS-PAGE) loading buffer.

For immunoblotting, cellular extracts or immunoprecipitates were separated on

SDS-polyacrylamide gels, transferred to a nitrocellulose membrane, probed with the

cognate antibody and visualized with enhanced chemiluminescence.

For immunofluorescence analyses, H1299 cells or MEFs transfected cells on

cover slips were cultured in DMEM for 16 h, washed once with PBS (phosphate buffered

saline) and fixed in 4% paraformaldehyde for 10 min at 37ºC. The cells were

permeabilized in buffer containing 1% Triton X-100 and 1% BSA at room temperature for

30 min and incubated for another 1 h in PBS containing 0.2% NP-40, 1% bovine serum

albumin and the primary antibody. After washing three times in PBS, the cells were

incubated for 45 min with goat anti-mouse IgG conjugated with Alexa Fluor 594 (red) or

fluorescein isothiocyanate (FITC; green) conjugated anti-rabbit IgG (Molecular Probes),

respectively and washed three times with PBS. The slides were dried and mounted with

Vectashield mounting medium with DAPI (4', 6'-diamidino-2-phenylindole). DAPI staining

was performed to visualize the nucleus. Regular fluorescent microscopic images were

obtained with a Nikon Diaphot microscope using a Photometrix PXL cooled CCD

camera. The microscope was equipped with the appropriate filters for three-color

imaging and a motorized stage for obtaining z-series images. Digital image files were

processed and deconvolved using the Oncor Image software (Oncor Inc.). High-

resolution images of the deconvolved and 3-D reconstructed image z-series stacks were

Page 73: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 57 -

processed for presentation with Adobe Photoshop. For confocal analysis, samples were

viewed with a Leica DMI6000 inverted microscope, TCS SP5 confocal scanner, and a

100X/1.40NA Plan Apochromat oil immersion objective (Leica Microsystems, Germany).

405 Diode and HeNe 594 laser lines were applied to excite the samples and tunable

filters were used to minimize spectral overlap between fluorochromes. DIC imaging was

performed using an argon laser line. Scale bars were created with the LAS AF software

version 1.6.0 build 1016 (Leica Microsystems, Germany).

Ni-NTA Pull-down Assay:

H1299 cells or MEFs were plated in 100 mm dishes and transfected with 4 µg

His6-ubiquitin (His-Ub) plasmid, 4 µg FOXO vectors and 4 µg MDM2 vectors using

Lipofectamine Plus; 24 h post transfection, cells were harvested and separated into two

aliquots. One aliquot (10%) was subjected to immunoblotting analysis to detect the

expression of transfected proteins. The other aliquot of cells (90%) was used to purify

the proteins of interested using Ni2+-nitrilotriacetic acid (NTA) beads. Cell pellets were

lysed in a buffer containing 0.01 M Tris-Cl (pH 8.0), 6 M guanidinium-HCl, 0.1 M sodium

phsophate, 5 mM imidazole, 10 mM β-mercaptoethanol and incubated with Ni2+-NTA

beads (Qiagen) for overnight at room temperature. The beads were washed sequentially

with the lysis buffer, a buffer containing 0.01 M Tris-Cl (pH 8.0), 8 M urea, 0.1 M sodium

phsophate, 10 mM β-mercaptoethanol, and a buffer containing 0.01 M Tris-Cl (pH 6.3), 8

M urea, 0.1 M sodium phsophate, 10 mM β-mercaptoethanol. Proteins bound to the

beads were eluted with a buffer containing 0.15 M Tris-Cl (pH 6.7), 5% SDS, 200 mM

imidazole, 30% glycerol, 0.72 M β-mercaptoethanol and were subjected to

immunoblotting analysis for the presence of Ub-conjugated FOXO proteins.

Page 74: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 58 -

In vitro Transcription Coupled Translations and GST (Glutathione S-transferase)

Pull-down assays:

FOXO1 protein was produced with pcDNA3-Flag-FOXO1 as a template using T7

polymerase-based in vitro transcription coupled translations (Promega, Madison, Wis.).

GST-MDM2 plasmids were transformed into BL21 and cultured at 37°C until the optical

density at 600 nm reached 0.6. Then, 0.2 mM of isopropylthiogalactopyranoside (IPTG)

was added and the incubation continued for another 5 h at 30°C. Bacterial cultures were

lysed by sonication in a buffer containing 50 mM Tris (pH 8.0), 10 mM NaCl, 1 mM

EDTA, 6 mM MgCl2, 1 mM dithiothreitol and 1 mM phenylmethylsulfonyl fluoride. GST

pull down analyses were performed using the MagneGST pull-down system (Promega

Madison, Wis) following the vendor’s protocols.

In Vitro Ubiquitination Assays:

Full length GST-MDM2 and GST-MDM2-NT (1-150) were expressed in E. coli

and bound to glutathione agarose beads. The substrate FOXO1 was produced by in vitro

transcription coupled translation in rabbit reticulocyte lysate using the TNT system

(Promega) in the presence of 35S- methionine. 4 µg GST-fusion proteins and 8 µl FOXO1

in vitro translation product were incubated to allow the enzyme-substrate interaction to

occur. After three times washes with PBS containing 0.2% NP-40, the bead-bound

enzyme-substrate complex was incubated at 37°C for 1 h with 250 ng GST-Ubc5Hb

(Boston Biochem), 250 ng purified rabbit E1 (Boston Biochem), 2 µg His6-Ub (Boston

Biochem) in 20 µl reaction buffer containing 50 mM Tris (pH 7.5), 2.5 mM MgCl2, 15 mM

KCl, 1 mM dithiothreitol, 0.01% Triton X-100, 1% glycerol and 8 mM ATP. The reactions

were terminated by boiling in SDS sample buffer and separated by SDS-polyacrylamide

gel electrophoresis. Gel was dried and the Ub-conjugated FOXO1 proteins were

detected by autoradiography.

Page 75: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 59 -

Apoptotic Analysis and Flow Cytometry:

The determination of the survival and apoptotic index of GFP (green fluorescent

protein)-transfected cells has been described (Li et al., 2001). In brief, transfected cells

were washed with PBS, fixed in 4% formaldehyde and stained with DAPI.

Representative micrographs were captured by a charge-coupled device camera with a

Smart Capture Program (Vysis, Downers Grove, Ill.) attached to a Leitz Orthoplan 2

fluorescence microscope. The viability of transfected cells in each well was determined

by counting the total number of green cells in each well. The apoptotic index of GFP-

positive cells was determined by scoring 300 GFP-positive cells for chromatin

condensation and apoptotic body formation.

To assay the apoptosis index induced by FOXO1, H1299 cells in 100 mm dishes

were transfected with GFP and FOXO1 with or without MDM2. Transfected cells were

collected in PBS containing 2.5 mM EDTA, washed twice with cold PBS, re-suspended

in 1× binding buffer containing 0.01 M Hepes (pH7.4), 0.14 M NaCl, 2.5 mM CaCl2 at a

concentration of 1 × 106 cells/ml and stained with Annexin-V APC and 7-AAD. Cell

sorting and flow cytometry analysis were performed on a FAC Scan (Becton Dickinson,

Mountain View, Calif.).

Page 76: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 60 -

RESULTS

FOXO proteins are ubiquitinated and their expression level is regulated by

proteasome mediated degradation (Aoki et al., 2004; Plas and Thompson, 2003), but a

general ubiquitin E3 ligase for FOXO proteins remains to be identified.

Skp2 was reported to mediate the ubiquitination and degradation of FOXO1 but

such activity appears to be restricted to FOXO1 (Huang et al., 2005). So far, no

mutational analysis and in vitro enzyme assays have been performed to show the direct

involvement of Skp2 E3 activity in FOXO ubiquitination. It remains to be determined

whether Skp2 promotes FOXO1 ubiquitination directly, through its intrinsic E3 activity, or

indirectly through another E3. The present study showed that MDM2 is the E3 ubiquitin

ligase for FOXO factors. MDM2 interacts with FOXO, promotes the degradation and

affects downstream targets and the biological function of FOXO.

1. MDM2 promotes the degradation of FOXO factors.

In an effort to investigate the interaction between p53 and FOXO signaling

pathways, an inverse correlation was noticed between the expression of FOXO3A and

the p53 target gene MDM2 using a panel of human cancer cell lines including

osteosarcoma (Saos2), prostate (LNCaP, PC3, DU145, and JCA1), breast (MCF-7),

cervical (Hela) and lung (H1299, H1299/V138) cancer cells (Figure 6). To determine

whether the inverse correlation of these two proteins among cancer cells reflects a

negative effect of MDM2 on FOXO protein, the level of FOXO3A and FOXO1 protein

expression in p53 null and p53/MDM2 double null MEFs was measured by

Page 77: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 61 -

immunoblotting. As shown in Figure 7, the knockout of MDM2 in MEFs resulted in an

increased expression of endogenous FOXO1 and FOXO3A proteins. Consistent with the

data from MEFs, the stable expression of MDM2 in H1299 cells reduced (Figure 8) and

the knockdown of endogenous MDM2 increased (Figure 9) the expression of

endogenous FOXO3A. Unlike MEFs, H1299/V138 cells express relatively low levels of

endogenous FOXO1, which is difficult to detect. Thus, we tested the effect of MDM2

silencing on the expression of ectopic FOXO1. As shown in Figure 10, the knockdown of

MDM2 by shRNA increased the level of ectopic Flag-FOXO1 expression.

Because MDM2 is an E3 ubiquitin ligase that promotes proteasome mediated

degradation of p53 (Honda et al., 1997) and other (Lin et al., 2002a) proteins, it is likely

that MDM2 decreases the expression of FOXO factors in a proteasome-dependent

manner. In transient transfection studies, co-expression of MDM2 reduced the

expression level of Flag-FOXO1 in DU145 prostate cancer cells, a process that was

prevented by treatment with two different concentrations of the proteasome inhibitor

MG132 (Figure 11).

To test whether the decreased FOXO protein expression by MDM2 is due to

protein degradation, the half-life of Flag-FOXO1 was measured in H1299 cells

cotransfected with control vector, full length MDM2 or an MDM2 (1-361) mutant which

lacks the C-terminal E3 ligase domain. As shown in Figure 12, the half life of FOXO1

was decreased by full length MDM2 from 6 h to 3 h, as compared to the control vector.

When FOXO1 was cotransfection of MDM2 (1-361), MDM2 (1-361) significantly

extended the half-life of Flag FOXO1; the exact meaning of the latter is unclear. Overall,

the data raise the possibility that MDM2 may function as an E3 ubiqitin ligase for

mammalian FOXO factors.

Page 78: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 62 -

Figure 6 Inverse correlation of MDM2 and FOXO3A protein expression

in different cancer cell lines. DU145, JCA, H1299-V138, HeLa, LNCaP,

MCF7, PC3, Saos-2, H1299 cells were plated into 100 mm dishes. MDM2

and FOXO3A proteins were detected by Western blotting with anti-MDM2

(SMP-14 ) and anti-FOXO3A (H-144) antibodies, respectively.

MDM2

DU

145

JCA1

H12

99/V

-138

Hel

a

LNC

aP

MC

F7

PC3

Saos

2

H12

99

78

105

MW(kD)

FOXO3A

Page 79: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 63 -

Figure 7 Inverse correlation of MDM2 and FOXO protein expression in MEFs.

p53-/- MEFs and p53-/-, MDM2-/- double null MEFs were plated in 100 mm dishes.

The cells were harvested the next day and cell lysates were subjected to

immunoblotting analyses with anti-MDM2 (SMP-14 ) antibody, anti-FOXO1 antibody,

and anti-FOXO3A (H-144) antibodies, respectively. The β-actin level showed the

equal loading.

P53-

/-

P53-

/-M

DM

2-/-

FOXO1

β-actin

150

100

75

50

150

100

75

50

IB:MDM2

IB:FOXO3A

MDM2

FOXO3A

IB:FOXO1

IB:β-actin

Page 80: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 64 -

Figure 8 Stably expressed MDM2 decreases the level of endogenous

FOXO3A expression. H1299 cells stably expressing MDM2 (MDM2-1, MDM2-

2) or empty vector (Control) were established and cell extracts were subjected to

Western blot analyses with the indicated antibodies.

Con

trol

MD

M2-

1

MD

M2-

2

MDM2

H1299 Stable Clones

FOXO3A

β-actin

IB:MDM2

IB:FOXO3A

IB:β-actin

Page 81: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 65 -

Figure 9 Knockdown of endogenous MDM2 by siRNA causes an

increase in endogenous FOXO3A protein. H1299 cells were transfected

with control GFP-siRNAs or siRNAs (MDM2-siRNA1, MDM2-siRNA2) for

MDM2, and cell extracts were subjected to Western blot analyses.

GFP

MD

M2-

1

MD

M2-

2

MDM2

FOXO3A

β -actin

siRNA

IB:MDM2

IB:FOXO3A

IB:β -actin

Page 82: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 66 -

Figure 10 Knockdown of endogenous MDM2 by siRNA increases

the level of ectopic FOXO1 protein. A Flag-tagged FOXO1 expression

vector was cotransfected transiently with MDM2-siRNA or control siRNA

(GFP-siRNA) into H1299-V138 cells, and whole cell extracts were

prepared and subjected to Western blot analyses.

+

-+

MDM2-shRNA-+-

Control-shRNA+-+Flag-FOXO1+--

+

-+

MDM2-shRNA-+-

Control-shRNA+-+Flag-FOXO1+--

β-actin

MDM2

Flag-FOXO1

IB:MDM2

IB:M2

IB: β-actin

Page 83: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 67 -

1010550 00MG132 (μM)+-+-+--MDM2++++++-Flag-FOXO1

1010550 00MG132 (μM)+-+-+--MDM2++++++-Flag-FOXO1

IB:M2

IB:β-Actin

IB:MDM2

FOXO1

MDM2

β-Actin

0

0.5

1

1.5

1 2 3 4 5 6 7

FOXO

1 Pr

otei

n D

ensi

ty

Figure 11 MDM2 overexpression causes a decrease in Flag-FOXO1 protein,

which is blocked by MG132. DU145 cells were transfected with Flag-FOXO1 and

pcMDM2. 24 h posttransfections, cells were incubated with or without different

concentrations of MG132 for 6 h. Cell lysates were separated in a 8% SDS-PAGE.

FOXO1 and MDM2 were detected by Western blotting with anti-Flag (M2) and anti-

MDM2 (SMP-14) antibodies, respectively. Equal amounts of protein were loaded.

Page 84: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 68 -

Flag-FOXO1

β-actin

+++++-----------MDM2(1-361)241263024126302412630-CHX treatment (h)

+++++++++++++++-Flag-FOXO1-----+++++------MDM2+++++-----------MDM2(1-361)241263024126302412630-CHX treatment (h)

+++++++++++++++-Flag-FOXO1-----+++++------MDM2

Figure 12 MDM2 Overexpression results in a decrease in half-life of the

FOXO1 protein. H1299 cells were transfected with the plasmids as indicated. 24 h

after transfection, cells were treated with CHX for different times. The cell lysates

were separated by SDS−8% PAGE. Exogenous FOXO1 was detected by Western

blotting with anti-Flag M2 antibodies. Equal amounts of protein were loaded on each

sample.

0

20

40

60

80

100

120

0 3 6 12 24

CTLMDM2MDM2(1-361)

FOXO

1/ β

–act

in(%

)

(hrs)

Page 85: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 69 -

2. MDM2 interacts with FOXO factors in vivo and in vitro.

Ubiquitin E3 ligases are known to make direct contact with their substrates.

Therefore, it was investigated whether or not MDM2 and FOXO factors form a complex

in cells. Deconvolution imaging analysis detected ectopic MDM2 and FOXO1 proteins in

both cytoplasmic and nuclear compartments of p53/MDM2 double null MEFs, but

colocalization was detected predominantly in the nucleus (Figure 13). Similarly, confocal

imaging gave the same result for endogenous MDM2 and FOXO3A in H1299 cells

(Figure 14).

To determine whether MDM2 and FOXO1 interact in mammalian cells, they were

ectopically expressed in H1299 cells and reciprocal coimmunoprecipitations performed.

MDM2 and FOXO1 were co-precipitated in cells expressing both proteins. In cells that

express either MDM2 or FOXO1, little or no coprecipitations were observed, suggesting

that co-precipitations were not due to cross reactivity of the antibodies (Figure 15).

Similar analysis showed that MDM2 and GFP-FOXO3A were specifically co-precipitated,

but not MDM2 and GFP (Figure 16).

To determine whether endogenous MDM2 and FOXO1 interacted in mammalian

cells, H1299/V138 cells expressing a temperature-sensitive p53 mutant (Pochampally et

al., 1999) were shifted to permissive temperature for 16 h to induce MDM2 expression

and cellular extracts were subjected to coimmunoprecipitation analysis. As shown in

Figure 17, FOXO1 was co-precipitated by the anti-MDM2 antibody, but not by the control

antibody, showing that the co-precipitations were not due to antibody cross reactivity.

To determine whether endogenous MDM2 and FOXO3A form a complex,

extracts of HEK293T cells were treated with DMSO or MG132 and co-

immunoprecipitations with an anti-MDM2 antibody. An anti-HA antibody was

Page 86: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 70 -

Figure 13 Exogenous MDM2 and Flag-FOXO1 are colocalized in the nucleus

of MEF cells. p53-/-, MDM2-/- MEF in a medium containing 0.5% FBS were

transfected with 1 μg pcMDM2 and 1 μg Flag-FOXO1. Cells were stained with DAPI

and incubated with anti-MDM2 (SMP14) and anti-FOXO1 (H-128) antibodies.

Immunoreactivity was detected with IgG conjugated to Alexa Fluor 594 (red for

MDM2) or FITC (green for FOXO1). Colocalization was determined by high-

resolution imaging with deconvolution microscopy.

Flag-FOXO1MDM2

Merged

20 microns

Page 87: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 71 -

Figure 14 Endogenous MDM2 and FOXO3A are colocalized in the nucleus of

H1299 cells. H1299 cells in a medium containing 1% FBS were stained with DAPI

and incubated with anti-MDM2 (SMP14) and anti-FOXO3A (H-144) antibodies.

Immunoreactivity was detected with IgG conjugated to Alexa Fluor 594 (red for

MDM2) or FITC (green for FOXO3A). Colocalization was visualized by high-

resolution imaging with confocal microscopy.

DAPI MDM2 FOXO3A

Merged

20 microns

Page 88: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 72 -

Figure 15 Interaction of ectopic FOXO1 and MDM2 in H1299 cells.

H1299 cells were cotransfected with HA-tagged FOXO1 and pcMDM2. 24 h

posttransfection, cell extracts were immunoprecipitated with either anti-HA or

anti-MDM2 antibodies followed by immunoblotting with anti-MDM2 or anti-HA

antibody.

++

MDM2+-HA-FOXO1-+

++

MDM2+-HA-FOXO1-+

60

78

109

60

78

109

60

78

47

IP: HAIB: MDM2

IP:MDM2IB:HA

IB: HA

IB: MDM2

MW(kD)

MDM2

FOXO1

MDM2

FOXO1

IB: β-actin β-actin

Page 89: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 73 -

Figure 16 Interaction of ectopic FOXO3A and MDM2 in HEK293T cells.

HEK293T cells were cotransfected with pcMDM2 and either GFP or GFP-

FOXO3A. 24 h posttransfection, cell extract were immunoprecipitated with either

an anti-MDM2 or an anti-GFP antibody followed by immunoblotting with an anti-

GFP or an anti-MDM2 antibody.

MDM2GFPGFP-FOXO3A

+-+

++

-

MDM2GFPGFP-FOXO3A

+-+

++

-

36

47

60

78

24

IP: MDM2IB: GFP

IP: GFP IB: MDM2

IB:GFP

IB:GFP

IB:MDM2

FOXO3A

MDM2

MDM2

FOXO3A

GFP

MW(kD)

Page 90: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 74 -

Figure 17 Interaction of endogenous MDM2 and FOXO1 in H1299/V138

cells. Cells were shifted from 39°C and cultured in 33°C for overnight and

were treated with or without MG132 for 6 h. Cell extracts were

immunoprecipitated with an anti-FOXO1 antibody followed by

immunoblotting with an anti-MDM2 antibody.

IPMG132

M+

M+

IPMG132

M+

M-

HAMW(kD)

IB: MDM2

IB: FOXO1

Heavy Chain50

75

50

75 FOXO1

MDM2

Page 91: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 75 -

Figure 18 Interaction of endogenous MDM2 and FOXO3A in HEK293T

cells. HEK 293T cells were treated with or without MG132 for 6 h before

harvest. Cell extracts were immunoprecipitated with either an anti-HA

antibody as control or an anti-MDM2 (SMP14) antibody followed by

immunoblotting with an anti-FOXO3A antibody.

-

HA

-

M

+

HA

+

M

MG132

IP

IB: FOXO3A

IB: MDM2

Heavy chain

78

60

60

78

109

MWkD

FOXO3A

MDM2

Page 92: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 76 -

Figure 19 The interaction between FOXO1 and GST-MDM2 in vitro.

FOXO1 synthesized by in vitro transcription-translation reactions was incubated

with GST-MDM2 fusion proteins, precipitated with glutathione beads and

detected by immunoblotting with an anti-FOXO1 antibody. The amounts of GST

proteins used in the pull-down assays were visualized by Coomassie blue

staining after separation in a SDS-PAGE gel (bottom panel).

GSTFOXO1-AAAFOXO1-WT

+-+-

--+++

+--+---+--

++-- GST-MDM2GSTFOXO1-AAAFOXO1-WT

+-+-

--+++

+--+---+--

++---

GST Pull Down

IB: FOXO1

CB: GST-MDM2

CB: GST

IB: FOXO1

Page 93: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 77 -

included as a control. As shown in Figure 18, FOXO3A proteins were co-precipitated

with MDM2 by an anti-MDM2 but not an anti-HA antibody. Treatment with MG132

increased the level of MDM2 and FOXO expression as well as the amount of proteins

co-precipitated. These studies showed that co-precipitations occur with both

endogenous and ectopically expressed proteins.

To determine whether MDM2 and FOXO1 interact in vitro, GST and GST-MDM2

fusion proteins were produced in bacteria, bound to glutathione beads and incubated

with FOXO1 proteins produced by in vitro transcription coupled translations. In these

GST pull down assays, wild type FOXO1 was precipitated with GST-MDM2, but not with

GST (Figure 19). An active form of FOXO1, FOXO1 (AAA) in which all three AKT

phosphorylation sites were mutated to alanine, was also precipitated with GST-MDM2

but not with GST (Figure 19). This analysis demonstrated that FOXO1 and MDM2 form a

complex in vitro and that complex formation occurs independently of the phosphorylation

of FOXO1 by AKT.

3. The fork head box of FOXO and the region of MDM2 controlling nuclear-

cytoplasmic shuttling mediate the interaction between MDM2 and FOXO.

To define the region in FOXO1 that mediates the interaction with MDM2, H1299

cells were transfected with full length MDM2 and FOXO1 deletion constructs fused to the

HA tag (Figure 20a). Cellular extracts were subjected to co-immunoprecipitation with an

anti-MDM2 antibody. As shown in Figure 21, the full length FOXO1 protein was co-

precipitated with MDM2. Deletion of the C-terminal region of FOXO1 did not alter the

interaction whereas further deletion into fork head box abolished it, suggesting that the

fork head box is required for the interaction.

Further define the region in FOXO3A that mediates the interaction with MDM2,

purified bacteria expressed GST or GST fused fragments of FOXO3A (peptide P1-P5)

Page 94: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 78 -

encoding five nonoverlapping FOXO3A regions (Figure 20b) were incubated with

HEK293T cell extracts overexpressed MDM2. MDM2 coprecipitated only with peptide P2

(amio acid 154-259), which contains the forkhead domain (Figure 22). These results

suggest that MDM2 specifically interacts with the forkhead domain of FOXO3A. Further

alignment the fork head domain of mouse FOXO3A, human FOXO1, FOXO3A and

FOXO4 (Figure 23), we found that the fork head domain is highly conserved in these

four kinds of FOXO. All these data indicated MDM2 has a general effect on FOXO.

To define the region of MDM2 responsible for FOXO1 binding, different MDM2

constructs (Figure 24) were transfected into H1299 cells together with Flag-FOXO1.

Cellular extracts were immunoprecipitated with MDM2 antibody and the co-precipitated

FOXO1 was detected by anti-Flag antibody. As shown in Figure 25, all MDM2 mutants,

except MDM2 (50-491) (p53 binding deficient) and MDM2 NLS-mt (182R, nuclear

localization sequence defective), interacted with FOXO1. Immunoblot analysis showed

that all MDM2 mutants were expressed at significant levels and most MDM2 proteins

were detected in multiple forms, presumably due to cleavage by proteases (Chen et al.,

1997). In the presence of proteasome inhibitor MG132, however, both MDM2 (50-491)

and MDM2 NLS-mt were co-precipitated with Flag-FOXO1 by the anti-Flag antibody

(Figure 26). The exact reason for the lack of interaction with FOXO1 in the absence of

MG132 is unclear, presumably due to the degradation of the protein complex by the

proteasome.

Further analysis with additional MDM2 mutants in the presence of MG132,

showed that the deletion of 150-230 amino acids abrogated the FOXO1 binding (Figure

27). Consistent with the fact that this region contains the nuclear localization sequence,

it was also shown (Figure 28) that this mutant is mainly localized to cytoplasm. However,

cytoplasmic localization is clearly not the reason for the lack of interaction with MDM2

Page 95: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 79 -

Figure 20 Diagram of different FOXO mutants used in this study.

a, The different mutants of human FOXO1 used in this study; b, The

different mutants of mouse FOXO3A used in this study.

FK150 2601 655

1-150

1-270

256-655

1-655

FL

FK148 2581 672

P1: 1-154

P2: 154-259

P3: 259-409

P4: 409-542

FLGST

P5: 542-672

GST

GST

GST

GST

GST

a

b

Page 96: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 80 -

Figure 21 Mapping of the MDM2-interacting domain of FOXO1 by

immunoprecipitation. H1299 cells were transfected with 2 µg MDM2 and 2 µg

of different HA-tagged FOXO1 fragments. Anti-MDM2 immunoprecipitates were

subjected to immunoblotting with anti-HA antibody and anti-MDM2 as indicated.

Immunoblotting of total cell extracts with anti-HA antibody (lower panel) showed

the expression of different FOXO1 fragments.

IP: MDM2IB: HA

IB: HA

Vect

or

1-15

0

1-27

0

256-

655

HA-FOXO1

IP: MDM2IB: MDM2

1-65

5 (F

L)

1-655

1-270

1-655

256-6551-270

1-150

MDM2

Page 97: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 81 -

++++++MDM2

P5P4P3P2P1GST

++++++MDM2

P5P4P3P2P1GST

GST pull down

IB:MDM2

MDM2

GST fusion protein

Figure 22 Mapping of the MDM2-interacting domain of FOXO3A by

GST pull-down assay. Recombinant GST or GST fused with fragments of

FOXO3A (P1-P5) were amplified by bacteria expression and purified by

GST beads. HEK 293T cells were transfected with 4 µg MDM2. Cell lysates

were subjected to GST pull down experiment. The amounts of GST proteins

used in the pull-down assays were visualized by Coomassie blue (CB)

staining after separation in a SDS-PAGE gel (bottom panel).

CB

Page 98: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 82 -

Figure 23 Alignment of FOXO member’s fork head box. Grey box

– fork head box; Green box –unique FOXO sequence.

Page 99: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 83 -

Figure 24 Diagram of different MDM2 mutants used in the study.

1-361

50-491

Δ150-230

E3-mt

NES-mt

NLS-mt

0 100 200 300 400 491

P53 binding NLS RingMDM2

Δ89-150

Δ50-89

Δ222-325

Δ222-437

361D-A

NES

Page 100: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 84 -

IP: anti-MDM2IB: anti-Flag

IB: anti-MDM21-

361

50-4

91

361D

-A

NE

S m

utan

t

182R

Δ222

-325

Δ222

-437

MDM2

++++++Flag-FOXO1 + + + +++++++Flag-FOXO1

FOXO1

MDM2 mutants

Figure 25 Mapping of the FOXO1-interacting domain in MDM2 by

coimmunoprecipitations in the absence of MG132. H1299 cells were

transfected with 2 μg Flag-FOXO1 and 2.5 µg different MDM2 fragments. Anti-

MDM2 mmunoprecipitates were probed with anti-Flag M2 antibody (upper

panel). Immunoblotting of cellular extracts with anti-MDM2 antibody (lower

panel) showed the expression of different MDM2 fragments.

457S

MD

M2

Page 101: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 85 -

Figure 26 An N-terminal truncation mutant and an NLS-mutant of MDM2

interact with FOXO1 in the presence of MG132. H1299 cells were

transfected with 2 μg Flag-FOXO1 together with 5 μg MDM2 or 5 μg mutant

MDM2. Cells were treated with MG132 for 6 h before harvest. Cells lysate were

immunoprecipitated with anti-Flag M2 antibody and detected with anti-MDM2

antibody. FL: full length MDM2, 50-491; MDM2(50-491); NLS: MDM2(NLS-mt).

FL 50-4

91

182R

75

50

IP:M2IB:MDM2

IB:MDM2

IB:M2

FL5% in

put

75

50

75

50

FOXO1

MDM2

MDM2/MDM2(182R)MDM2(50-491)

MW(kD)

Page 102: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 86 -

Figure 27 Truncation of the central region of MDM2 abolishes the

interaction between MDM2 and FOXO1. H1299 cells were transfected with 2 μg

Flag-FOXO1 and 2.5 µg of different MDM2 fragments. Cells were treated with

MG132 for 6 h before harvesting. Anti-Flag immunoprecipitates were probed with

anti-MDM2 (2A10) antibody (upper panel). Immunoblottiong of cellular extracts

with anti-MDM2 antibody (lower panel) showed the expression of different MDM2

fragments.

FLVect

or

FL Δ50-

89

Δ89-

150

Δ150

-230

50-4

91

IP: M2IB: MDM2

IB: M2

IB: MDM2

FOXO160

78

110

170

47

60

78

110

170

++

MG132++++++Flag-FOXO1++++-+

++

MG132++++++Flag-FOXO1++++-+

MW(kD)

MDM2

MDM2

60

78

110

Page 103: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 87 -

Figure 28 Immunofluorescence images show the cellular localization of

various MDM2 mutants. H1299 cells were transiently transfected with MDM2

or different MDM2 mutants. 24 h posttransfection, cells were fixed and

stained. The red signal showed the localization of MDM2 and the blue signal

showed the nucleus of the cells.

50-491

Δ222-325

Δ222-437

Δ50-89

Δ89-150

Δ150-230 E3-mt

1-361

NES-mt

NLS-mt

MDM2

MDM2 DAPI

361D-A

MDM2 DAPI

Page 104: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 88 -

because co-precipitations were done with whole cell extracts and, under the same

conditions, nuclear localization sequence mutant, the MDM2 NLS-mt, interacted with

FOXO1 (Figure 26). Besides cellular localization sequences, the 150-230 region also

contains an inhibitory domain that suppresses cell cycle progression independently of

p53. This region is also involved in interactions with several proteins, including TBP and

p300.

4. MDM2 promotes the ubiquitination of FOXO1 and FOXO3A.

To test whether MDM2 promoted the ubiqutination of FOXO factors, H1299 cells

were transfected with Flag-tagged FOXO1, wild type or an AKT phosphorylation site

FOXO1 mutant. The effect of ectopic MDM2 on FOXO1 ubiquitination by cotransfected

Myc-tagged ubiquitin was measured in the anti-Flag precipitates. In the absence of

ectopic MDM2, limited FOXO1 ubiquitination was occured. This result is consistent with

the fact that H1299 cells are p53-deficient and contain low levels of endogenous MDM2.

Cotransfection of MDM2 with increased the level of ubiquitiation of the wild type FOXO1

but not of FOXO1 (AAA). The data argue that the positive effect of MDM2 on FOXO1

ubiquitination requires phosphorylation at the AKT sites (Figure 29).

To test whether the effect of MDM2 extends to other FOXO factors, FOXO3A

and His-tagged ubiquitin were transfected into H1299 cells and the effect of MDM2 on

FOXO3A ubiquitination was measured by immunblotting with anti-HA antibody following

nickel bead pull-down under denaturing conditions. As shown in Figure 30, FOXO3A

ubiquitination was increased by MDM2 in a manner dependent on the phosphorylation

on the AKT sites. The data suggest that the stimulation of ubiquitination by MDM2 is not

restricted to FOXO1 but among the FOXO factors.

In p53 and MDM2 double-null MEFs, wild type MDM2 stimulated the

ubiquitination of FOXO1 in a dose dependent manner, whereas the MDM2 mutant

Page 105: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 89 -

lacking the C-terminal ring finger E3 region did not exert such an effect (Figure 31),

emphasizing the potential involvement of the E3 ligase activity. Nickel bead pull down

assays under denaturing conditions revealed that MDM2 containing a point mutation in

the E3 ligase domain, MDM2 E3 -mt (457S), did not stimulate FOXO1 ubiquitination

(Figure 32), confirming that FOXO1 ubiquitination by MDM2 requires its ubiquitin ligase

activity. Interestingly, on the one hand, a constitutively nuclear MDM2 in which the

nuclear export sequence was mutated, the NES-mt, did not promote FOXO1

ubiquitination (Figure 33), even though it contained an intact E3 ligase domain and

interacted with FOXO1 (Figure 25). On the other hand, two cytoplasmic MDM2 mutants,

MDM2 NLS-mt and MDM2 (∆89-150), stimulated FOXO1 ubiquitination (Figure 32 and

33), showing that the ubiquitiantion of FOXO1 by MDM2 is likely to occur in the

cytoplasm. MDM2 (∆150-230), which was located mainly in the cytoplasm (Figure 25)

and did not interact with FOXO1 (Figure 26), was unable to stimulate FOXO1

ubiquitination, suggesting that the ubiquitination requires FOXO1 interaction. Overall, the

data suggest that FOXO1 interacts with MDM2 in both the nucleus and the cytoplasm,

but the ubiquitination by MDM2 requires the interaction in the cytoplasm.

To fully establish MDM2 as an E3 ligase for FOXO1 ubiquitination, the ability of

recombinant MDM2 to catalyze the ubiquitination of FOXO1 was tested in vitro. In this

experiment, GST-MDM2 stimulated the ubiquitination of in vitro translated FOXO1 in the

presence but not in the absence of purified E1, E2 and ubiquitin (Figure 34, upper

panel). GST fused to an MDM2 N-terminal fragment had no effect on the ubiquitination of

FOXO1. In reactions performed with transcription-coupled translation product from a

control vector, the majority of the 35S-labeled ubiquitin conjugates disappeared, showing

that they were FOXO1 proteins (Figure 34, lower panel). In combination with the binding

Page 106: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 90 -

Figure 29 MDM2 promotes the ubiquitination of FOXO1. H1299 cells were

transfected with the indicated plasmids and cell extracts were either

immunoprecipitated with M2 antibody, followed by immunoblotting with anti-Myc

antibody, or directly immunoblotted with antibodies to Flag or MDM2.

++-+ MDM2-+--

Myc-Ub+++-Flag-FOXO1 (AAA)+---Flag-FOXO1wt-+++

++-+ MDM2-+--

Myc-Ub+++-Flag-FOXO1 (AAA)+---Flag-FOXO1wt-+++

IP: M2IB: myc

IB: M2

IB: MDM2

170

110

78

60

MW(kD)

Ub-FOXO1

FOXO1

MDM2

Page 107: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 91 -

Figure 30 MDM2 promotes the ubiquitination of FOXO3A. H1299 cells were

transfected with the indicated plasmids and treated with MG132 for 6 h before

harvest. Cell lysates were subjected to pull down analyses with Ni-NTA beads

followed by immunoblotting with anti-FOXO3A antibody. Cell extracts were also

subjected to direct immunoblotting by anti-MDM2 antibody.

MDM2+-+--His-Ub+++++

+-

FOXO3A (AAA)+---FOXO3Awt-++-

MDM2+-+--His-Ub+++++

+-

FOXO3A (AAA)+---FOXO3Awt-++-

MW(kD)

IB: FOXO3A

170110

7860

170

110

78

60

IB: MDM2

Ni-NTA pull down

Ub-FOXO3A

MDM2

Page 108: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 92 -

Figure 31 The MDM2 ring finger domain is critical for the ubiquination of

FOXO1. p53-/-,MDM2-/-MEF cells were transfected with the indicated plasmids

including Myc-tagged ubiquitin and cell extracts were either immunoprecipitated

with M2 antibody followed by immunoblotting with an anti-Myc antibody or

immunoblotted directly with antibodies to Flag or MDM2.

4-++

MDM2 (μg)-42--MDM2 (1-361)2----

Myc-Ub+++++Flag-FOXO1++++-

4-++

MDM2 (μg)-42--MDM2 (1-361)2----

Myc-Ub+++++Flag-FOXO1++++-

IP: M2IB: Myc

IB:MDM2

IB:M2

170

110

78

MW(kD)

Ub-FOXO1

MDM2

MDM2(1-361)

FOXO1

Page 109: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 93 -

-++++++++++FOXO1 -++++++++++FOXO1

CTL

MD

M2

MD

M2(

1-36

1)

MD

M2(

50-4

91)

MD

M2(

NES

mut

ant)

MD

M2(

NLS

mut

ant)

MD

M2(

457S

)

MD

M2(

Δ222

-325

)

MD

M2(

Δ222

-437

)

MD

M2(

361

mut

ant)

CTL

Ni-NTApull down

IB:FOXO1

IB:MDM2

IB:FOXO1

Figure 32 Ubiquitination of FOXO1 requires the MDM2 ubiquitin ligase

function. P53-/-, MDM2-/- MEFs were transfected with Flag-FOXO1, His-

Ubiquitin and MDM2 mutants. Cell extracts were subjected to pull down assays

with Ni-NTA beads followed by immunoblotting with anti-FOXO1 antibody.

170

110

78

63

47

M.W.(kD)

110

78

63

47

Ub-FOXO1

MDM2

FOXO1

Page 110: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 94 -

Figure 33 The central region of MDM2 cannot promote the

polyubiquitination of FOXO1. H1299 cells were transfected with 2 μg Flag-

FOXO1, 4 μg MDM2, and 4 μg His-ubiquitin. Cell extracts were subjected to pull

down analyses with Ni-NTA beads followed by immunoblotting with anti-FOXO1

antibody.

Vect

or

FL Δ50-

89

Δ89-

150

Δ150

-230

Ni-NTAPull Down

IB: FOXO1

IB: MDM2

IB: FOXO1

75

100

150

MW(kD)

Ub-FOXO1

MDM2

FOXO1

Page 111: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 95 -

Figure 34 MDM2 promotes FOXO1 polyubiquitination in vitro. GST-MDM2

and GST-N (containing MDM2 residues 1 to 150) were purified using glutathione

agarose beads. Loaded beads were incubated with in vitro-translated FOXO1 in

the presence or absence of E1 and E2 in an ubiquitination reaction as described

in Materials and Methods. Polyubiquitinated FOXO1 appears as a high

molecular weight smear above the unmodified FOXO1 band.

(35S labeled)

Ub-FOXO1

(35S labeled)

Ub-FOXO1

75

100

150

E1+E2+Ub+-+GST-MDM2++-GST-MDM2-NT--+FOXO1+++

E1+E2+Ub+-+GST-MDM2++-GST-MDM2-NT--+FOXO1+++

+++

E1+E2+Ub+GST-MDM2+FOXO1-

+++

E1+E2+Ub+GST-MDM2+FOXO1-

75

100

150

MW(kD)

MW(kD)

Page 112: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 96 -

data and whole-cell ubiquitination analysis with MDM2 mutants, the in vitro data

established that MDM2 functions as an ubiquitin E3 ligase for FOXO proteins.

5. MDM2 suppresses the expression of FOXO target genes and protects cells

from FOXO1-induced cell death.

FOXO target genes tumor necrosis related apoptosis inducing ligand (TRAIL),

p27 CDK inhibitor and manganese superoxide dismutase (MnSOD) mediate the effect of

FOXO proteins on cell cycle arrest, apoptosis, and detoxification of reactive oxygen

species. Their transcription products are directly regulated by FOXO factors. Consistent

with the ubiquitination and degradation of FOXO factors by MDM2, stable expression of

MDM2 in H1299 cells decreased the level of TRAIL, p27 and MnSOD expression (Figure

35) whereas the knockdown of MDM2 increased the expression of TRAIL (Figure 36).

To test whether MDM2 protects cells from FOXO induced cell death, H1299 cells

were transiently transfected with GFP and either control vector, FOXO1 or FOXO1 in

combination with MDM2. The survival of the transfected cells was analyzed. The

expression of FOXO1 decreased the number of transfected (green) cells, an effect that

is relieved by MDM2 co-expression (Figure 37a). To confirm that the change in the

viability of transfected cells is the result of cell apoptosis, transfected cells were fixed

and stained with DAPI and their nuclear morphology was examined for features of

apoptosis under a fluorescence microscope that allows the simultaneous visualization of

blue and green fluorescence. Apoptotic index, as determined by scoring apoptotic cells

in 300 green cells per sample, was 5% for controls and 25% for cells transfected with

FOXO1. Co-expression of MDM2 suppressed FOXO1-induced cell death in a dose-

dependent manner (Figure 37b). The data were reproduced by independent

Page 113: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 97 -

IB:MDM2

IB:FOXO3A

IB:MnSOD

IB:p27

IB:CyclinD

H129

9-VE

CH12

99-M

1

IB:FOXO1

Figure 35 Stable expressed MDM2 in H1299 cells regulates the

expression of downstream targets of FOXO. Extracts of control (H1299-VEC)

and MDM2 stable (H1299-M1) clones were subjected to immunoblotting with

different antibodies as indicated in the figure.

IB:β-actin

IB:TRAIL

MDM2

FOXO3A

FOXO1

MnSOD

p27

TRAIL

Cyclin D

β-actin

Page 114: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 98 -

IB:MDM2

IB:β-actin

siRNA CTL MD

M2-

1

IB:FOXO3A

IB:TRAIL

Figure 36 MDM2 siRNA increases the expression of the FOXO1 target gene

TRAIL. H1299 cells were transiently transfected with either GFP-siRNA or MDM2-

siRNA. The cells were harvested at 48 h later. Cell extracts were subjected to

immunoblotting analyses with anti-MDM2, anti-FOXO3A and anti-TRAIL

antibodies.

MDM2

FOXO3A

TRAIL

β-actin

Page 115: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 99 -

6h

FOXO1 FOXO1+MDM2

9h

20h

pcDNA3

0%

10%

20%

30%

0.40.200MDM2(ug)

+++-Flag-FOXO1

0.40.200MDM2(ug)

+++-Flag-FOXO1

Apop

tosi

s in

dex(

%)

a

b

Figure 37 MDM2 promotes the cell survival in the presence of FOXO1.

Page 116: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 100 -

Figure 37 MDM2 promotes the cell survival in the presence of FOXO1.

a, H1299 cells were transfected with pLNCE and Flag-FOXO1 in the presence

or absence of MDM2. The viability of transfected cells in each well was scored

by counting the number of green cells. Representative micrographs were

captured by the fluorescence microscope that had a charge-coupled device

camera. b, H1299 cells were transfected with the same plasmids as in a,

Apoptotic index of GFP-positive cells was determined by scoring 300 GFP-

positive cells for chromatin condensation and nuclear fragmentation. Triplicate

samples were analyzed per data point, and the graph represents three

independent experiments.

Page 117: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 101 -

0

10

20

30

40

Apop

tosi

s (%

)MDM2 (μg)

Flag-FOXO1

+

+

+

-

--

+-

MDM2 (μg)

Flag-FOXO1

+

+

+

-

--

+-

49.0%

32.4%

FOXO1

13.1%10.4%10.7%1

31.33%-16.7%2

FOXO1+MDM2MDM2ControlExp. No.

49.0%

32.4%

FOXO1

13.1%10.4%10.7%1

31.33%-16.7%2

FOXO1+MDM2MDM2ControlExp. No.

0

10

20

30

40

+

+

MDM2 (μg)--

Flag-FOXO1+-

+

+

MDM2 (μg)--

Flag-FOXO1+-

Apop

tosi

s (%

)

a

Figure 38 MDM2 protects cells from FOXO1-induced cell death measured

by Flow Cytometry.

Page 118: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 102 -

Figure 38 MDM2 protects cells from FOXO1-induced cell death measured

by Flow Cytometry. H1299 cells were transfected with GFP-spectrin and Flag-

FOXO1 in the presence or absence of MDM2. The apoptotic GFP positive cells

were detected with Annexin-V APC and 7-AAD. a, bar graphs; b,representative

flow cytometry profiles.

0.6 1.1

10.7

1.72 2.12

10.4

1.52 4.13

32.4

1.18 2.84

13.1

7AA

D

100

101

102

103

104

100

101

102

103

104

100

101

102

103

104

100

101

102

103

104

101 102 103 104 101 102 103 104

101 102 103 104 101 102 103 104

Annexin-V APC

Control MDM2

87.6 85.7

62 82.9

FOXO1 FOXO1+MDM2

b

Page 119: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 103 -

analysis of early apoptosis with the Annexin V method after FACS-based sorting. As

shown in Figure 36, two independent analyses of cells cotransfected with GFP-spectrin

showed that FOXO1 expression increased apoptosis in transfected cells by about 3-fold,

which is partially suppressed by the co-tranefection of MDM2. The degree of induction

by FOXO1 and the suppression by MDM2 varied between two experiments because the

basal line of the FACS machine varied from time to time.

6. MDM2 transiently increased FOXO transcriptional activity.

Since FOXO factors are transcriptional factors, an important question which must

be addressed is whether or not MDM2 affects the FOXO transcriptional activity. In order

to answer this question, the following reporter genes were used in our experiment.

Synthetic reporter 3 × IRSLuc which contained three conserved insulin response

sequence (IRS). FOXO1, like insulin, promotes the promoter activity through an IRS.

Cyclin D is the downstream target of FOXO factors in the cell cycle check point. FOXO

factors expression results in reduced levels of cyclin D protein expression. Cyclin D-Luc

is the reporter gene which can be used to specifically measure cyclin D activity in cells.

In order to measure the activity of the reporter gene, a transient transfection was

performed by transfecting the reporter gene, CMV-gal as an internal control, pcDNA3

control or Flag-FOXO1, and different doses of MDM2 into the DU145 cells and NIH3T3

cells. The data showed that MDM2 increased the 3 × IRSLuc activity, and this increase

occurred in a dose dependent manner (Figure 39 & Figure 40). FOXO1 decreased the

cyclin D-Luc activity and MDM2 decreased it further in a dose dependent manner

(Figure 41).

Our earlier data revealed that MDM2 interacted with both wild-type FOXO1 and

FOXO1 (AAA), however it only promoted the polyubiquitination of wild-type FOXO1.

Page 120: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 104 -

05

1015202530354045

MDM2-0ugMDM2-0.1ugMDM2-0.3ugMDM2-0.5ug

3×IR

SLuc

act

ivity

(R

LU, ×

105 )

Endo-FOXO Flag-FOXO1wt

IB:M2

IB:MDM2

Figure 39 MDM2 increases the transcriptional activity of FOXO1 in a

dose-dependent manner in DU145 cells. DU145 cells were transfected with

0.5 μg 3×IRSLuc, 0.1 μg CMV-Gal, 0.1 μg control vector or Flag-FOXO1, and

different amounts of MDM2. FOXO activity was measured by the Promega

luciferase activity kit.

Page 121: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 105 -

02468

10121416

3×IR

SLuc

act

ivity

(R

LU, ×

105 )

MDM2

NIH3T3

Figure 40 MDM2 increases the transcriptional activity of FOXO1 in

NIH3T3 cells. NIH3T3 cells were transfected with 0.5 μg 3×IRSLuc, 0.1 μg

CMV-Gal, 0.1 μg Flag-FOXO1, and different amounts of MDM2. FOXO

activity was measured by the Promega luciferase activity kit.

Page 122: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 106 -

Cyc

linD

Luc

activ

ity (R

LU, ×

105 )

-MDM2

++++-FOXO1

-MDM2

++++-FOXO1

0

2

4

6

8

10H1299 cells

Figure 41 MDM2 enhances the ability of FOXO1 to inhibit cyclin D1.

H1299 cells were transfected with 0.5 μg cyclin D-Luc, 0.1 μg CMV-Gal, 0.1

μg Flag-FOXO1 and different amounts of MDM2. FOXO activity was

measured by the Promega luciferase activity kit.

Page 123: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 107 -

Figure 42 MDM2 increases the transcriptional activity of both wild type

FOXO1 and the FOXO1(AAA) mutant in NIH3T3 and DU145 cells. Cells were

transfected with 0.5 μg 3×IRSLuc, 0.1 μg CMV-gal, 0.1 μg Flag-FOXO1 or

FOXO1 (AAA) and different amounts of MDM2. FOXO activity was measured by

the Promega luciferase activity kit. a, DU145; b, NIH3T3.

3×IR

SLuc

act

ivity

(R

LU, ×

105 )

3×IR

SLuc

act

ivity

(R

LU, ×

106 )

a

b

0

5

10

15

20

25

30

FOXO1wt FOXO1(AAA)

MDM2-0ugMDM2-0.3ugMDM2-0.5ug

0

5

10

15

20

25

30

endo-FOXO1 FOXO1wt FOXO1 (AAA)

MDM2-0ugMDM2-0.3ugMDM2-0.5ug

Page 124: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 108 -

MDM2 also increased the transcriptional activity of FOXO1 (AAA) (Figure 42). The data

further indicated that MDM2 promotes FOXO transcriptional activity in an AKT-

independent fashion.

7. p53 induces transient increase in the transcriptional activity of FOXO factors,

which is followed by FOXO degradation in an MDM2-dependent manner.

To test whether p53 affects the transcriptional activity of FOXO1, reporter gene 3

× IRSLuc, CMV-gal, pcDNA3 control or Flag-FOXO1 or Flag-FOXO1 (AAA), and

different doses of p53 were transfected into DU145 cells (contain endogenous mutated

p53) and LNCaP cells (contain endogenous wild type p53) and the transcriptional activity

of FOXO1 was detemined. p53 decreased FOXO1 transcriptional activity in both cell

lines (Figure 43a & Figure 44), but did not inhibit FOXO1-induced decrease in cell

viability (Figure 43b). Introduction of MDM2 into the cells relieved the inhibition of FOXO

activity by p53 (Figure 45).

In order to better understand the interaction among FOXO factors, p53 and

MDM2, H1299/V138 stable cell line that expresses a temperature-sensitive p53 was

used. Shifting to a permissive temperature allows the activation of p53. At different time

points after p53 activation, the transcriptional activity of endogenous FOXO factors and

the level of FOXO3A and MDM2 protein expression were determined. Reporter analyses

showed that FOXO activity was transiently induced between 5 and 24 hours after p53

activation and subsequently decreased (Figure 46a). Immunoblotting analysis showed

that the MDM2 protein was induced to the highest level 5 hours after p53 activation

follwed by gradually decrease of FOXO3A and MDM2 level (Figure 46b). MG132

treatment prevented the time-dependent decrease in FOXO3A levels (Figure 47),

suggesting that the decrease is due to proteasome-mediated degradation. In order to

Page 125: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 109 -

Figure 43 P53 inhibits the transcriptional activity of FOXO in a dose-

dependent manner in DU145 cells, but not FOXO1-induced cell death. a, DU145

cells were transfected with 0.5 μg 3×IRSLuc, 0.1 μg CMV-Gal, 0.1 μg Flag-FOXO1

and different amounts of p53. FOXO activity was measured by the Promega

luciferase activity kit. b, Cells were transfected with pLNCE and plasmids as

described in panel a. Representative micrographs were captured by the

fluorescence microscope that had a charge-coupled device camera.

0

20

40

60

80

100

120

pcDNA3 FOXO1wt FOXO1(AAA)

p53-0ugp53-0.02ugp53-0.1ugp53-0.3ug

3XIR

SLuc

act

ivity

(R

LU, ×

104 )

CTL FOXO1

FOXO1+p53 FOXO1+p53+MDM2

b

a

Page 126: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 110 -

Figure 44 P53 inhibits the transcriptional activity of FOXO in a dose-

dependent manner in LNCaP cells. DU145 cells were transfected with 0.5

μg 3×IRSLuc, 0.1 μg CMV-Gal, 0.1 μg Flag-FOXO1 and different amounts

of p53. FOXO activity was measured by the Promega luciferase activity kit.

0

2

4

6

8

10

pcDNA3 FOXO1wt FOXO1(AAA)

p53-0ugp53-0.02ugp53-0.1ugp53-0.3ug

3XIR

SLuc

act

ivity

(R

LU, ×

104 )

Page 127: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 111 -

3×IR

SLuc

act

ivity

(R

LU ×

106 )

Figure 45 MDM2 relieves the repression of FOXO1 activity by p53.

DU145 cells were transfected with 0.5 μg 3×IRSLuc, 0.1 μg CMV-gal, 0.1 μg

Flag-FOXO1, 0.1 μg HA-p53 and 0.5 μg of either control vector or MDM2.

FOXO activity was measured by luciferase activity kit (Promega).

0

1

2

3

4

5

6

FOXO1wt FOXO1(AAA)

mtp53wtp53wtp53+MDM2

Page 128: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 112 -

FOXO3A

MDM2

p53

0 1 3 5 7 12 18 32 ºC (h)

Figure 46 MDM2 transiently increases FOXO transcriptional activity,

which is followed by FOXO degradation . H1299/V138 cells cultured at 39oC

were shifted to 32ºC for the indicated length of time. Cell extracts were

prepared and assayed by luciferase assay (panel a) or by immunoblotting

analyses (panel b).

0

0.5

11.5

2

2.5

3

0h 1h 3h 5h 9h 24h 48h 72h

3×IR

SLuc

act

ivity

(R

LU, ×

105 )

a

b

IB:FOXO3A

IB:MDM2

IB:p53

Page 129: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 113 -

18950950

++++----

18950950

++++----

FOXO3A

β-actin

18 32ºC (h)

MG132

32ºC (h)

MG132

75

50

100

150

Figure 47 MG132 relieves p53-induced decrease in the expression of

FOXO3A protein in H1299/V138 cells. Cells cultured at 39°C were shifted to

32ºC for indicated length of time. Cells were treated with MG132 for 6 h before

cell extracts were prepared and subjected to immunoblot analysis.

IB: FOXO3A

IB: β-actin

Page 130: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 114 -

HA-p53MDM2Flag-FOXO1

-++-+-++---+++++--

HA-p53MDM2Flag-FOXO1

-++-+-++---+++++--

IB:HA

IB:MDM2

IB:M2

IB: Tubulin

p53

MDM2

FOXO1

Tubulin

Figure 48 p53 decreases the protein level of FOXO1 through MDM2.

H1299 cells were transiently transfected as noted on the Figure. 24h after

transfection, cell lysates were subjected to immunoblotting with M2 antibody

(to detect FOXO1 expression), anti-HA antibody (to detect p53 expression),

and anti-MDM2 (SMP14). Tubulin blots was included to show even loading in

each sample.

Page 131: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 115 -

FOXO3A

32-+

MDM2-siRNA-++Temperature (ºC)373237

GFP-siRNA+--

32-+

MDM2-siRNA-++Temperature (ºC)373237

GFP-siRNA+--

β-actin

MDM2

Figure 49 Knockdown of MDM2 partially relieves p53-induced FOXO3A

downregulation. H1299/V138 cells were transfected with scrambled or MDM2

siRNA. 24 h posttransfection, cells were shifted to 32ºC and cultured for

another 18 h. Then immunoblotting was performed with the indicated

antibodies.

IB: FOXO3A

IB:MDM2

IB:β-actin

Page 132: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 116 -

determine which protein led to the degradation of FOXO factors, a transient transfection

experiment was performed in H1299 cells. Immunoblotting analyses showed that p53 did

not decrease the level of FOXO1 protein expression. MDM2, either alone or in

combination with p53, decreased the level of FOXO1 protein expression (Figure 48).

Knockdown of MDM2 by siRNA partially relieved FOXO3A down-regulation by active

p53, supporting the conclusion that the p53-induced decrease is MDM2-dependent

(Figure 49).

8. Site-specific sumoylation of SIRT1 regulates FOXO1 transcriptional activity

and stability.

Acetylation can affect protein degradation via ubiquitination (Ito et al., 2002;

Jeong et al., 2002; Li et al., 2002). The study in pancreatic β cells showed that FOXO1

acetylation inhibited the degradation of FOXO1 via the ubiquitin pathway (Kitamura et

al., 2005).

SIRT1 is a mammalian HDAC. As a potential nutrient sensor, it regulates the

lifespan of mammals in response to caloric restriction or nutrient starvation, and protects

cells from apoptosis induced by DNA damage.

Previously (Brunet et al., 2004) and our studies (Figure 50) showed that SIRT1

inhibited FOXO1 transcriptional activity. After treating the H1299/V138 cells with SIRT1

inhibitor nicotinamide, we found that nicotinamide caused further increase of FOXO1

activity from 24 hours to 48 hours post p53 activiation (Figure 51). Therefore SIRT1 has

a role in the action of p53-MDM2 on the activity and degradation of FOXO1.

Since SIRT1 affects the FOXO1 transcriptional activity and plays a potential role

in FOXO1 stability, two very interesting questions arise: How does SIRT1 affect the

transcriptional activity of FOXO1 and how is SIRT1 activity regulated. Our previous study

Page 133: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 117 -

Figure 50 SIRT1 inhibits the transcriptional activity of FOXO. H1299 cells

were transfected with 0.5 μg 3×IRS Luc, 0.1 μg CMV-Gal, 0.1 μg Flag-FOXO1 and

0.5 μg HA-SIRT1. FOXO activity was measured by the Promega luciferase activity

kit.

3×IR

SLuc

act

ivity

(R

LU ×

105 )

0

5

10

15

20

25

1 2 3

+--SIRT1

++-FOXO1

+--SIRT1

++-FOXO1

Page 134: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 118 -

3×IR

SLuc

act

ivity

(R

LU, ×

105 )

Figure 51 Nicotinamide treatment increases the FOXO transcriptional

activity. H1299 and H1299/V138 cells were transfected with 0.5 μg 3×IRS

Luc and 0.1 μg CMV-Gal. 16 h later, the cells were treated with nicotinamide

and transferred from 37°C to 32°C at indicated time points. FOXO activity

was measured by the Promega luciferase activity kit.

0

2

4

6

8

10

12

14

16

18

0h 1h 3h 5h 8h 24h 48h

H1299-V

H1299-V-NIC

H1299

H1299-NIC

Page 135: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 119 -

Figure 52 SIRT1 sumoylation at Lys 734 is required for FOXO1

deacetylation. H1299 cells were co-transfected with Flag-FOXO1, HA-p300

and either wild-type HA-SIRT1 (WT) or HA-SIRT1 mutated at Lys 734

(734R). Anti-Flag M2 immunoprecipitates were immunoblotted with antibody

to acetylated FOXO1 or with an antibody to FOXO1. Cellular extracts were

also immunoblotted with antibody to HA.

IP:M2

IB:Acetyl -K

IB:M2

IB:HA

IB:HA

+++-HA-p300

734RWT--HA-SIRT1

++++Flag-FOXO1

+++-HA-p300

WT--HA-SIRT1

++++Flag-FOXO1

p300

SIRT1

Page 136: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 120 -

CTL p300

Figure 53 Mutation of Lys 734 relieves the inhibition of FOXO1

transcriptional activity by SIRT1. The activity of transfected FOXO1 was

measured using an IGFBP1 promoter-based reporter in H1299 cells expressing

wild type SIRT1 or SIRT1 (734R) in the absence or presence of p300. CTL-

control.

0

5

10

15

20

25

30

35

1 2

pcDNA3FOXO1FOXO1+SIRT1FOXO1+SIRT1(734R)

3×IR

SLuc

act

ivity

(RLU

, ×10

5 )

Page 137: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 121 -

showed that the sumoylation status of SIRT1 affects its deacetylase activity since a

sumoylation site specific mutation abolished the deacetylase activity of SIRT1.

FOXO1 is a known substrate of SIRT1. In order to determine whether this

sumoylation site affects the effect of SIRT1 on FOXO, Flag-tagged FOXO1 was

cotransfected with p300, wild-type SIRT1 or SIRT1 (734R) in which the sumoylation site

of SIRT1 was mutated, acetylated FOXO1 was detected by Western blotting of M2

immunoprecipitates with an antibody that recognizes acetylated FOXO1. As shown in

Figure 52, expression of wild type SIRT1 reduced amount of acetylated Flag-FOXO1

induced by p300, whereas expression of SIRT1 mutated at Lys 734 did not. When the

cells transfected either FOXO1 with SIRT1, SIRT1 decreased FOXO1 transcriptional

activity which is consistent with the previous study, whereas expression of SIRT1

mutated at Lys734 could not decrease the transcriptional activity of these two proteins

(Figure 53). These data showed that the sumoylation status of SIRT1 is critcal for its

effect on FOXO1.

9. Genistein-induced FOXO1 expression is blocked by MDM2 expression in

H1299 cells.

Genistein is considered the primary anticancer component of soybeans. Its in

vitro and/or in vivo activities include the antagonism of estrogen, inhibition of protein

tyrosine phosphorylation, suppression of angiogenesis, inhibition of hydrogen peroxide

formation induced by tumor promoters, inhibition of topoisomerases, induction of

apoptosis and cell differentiation, scavenging of free radicals, and inhibition of

carcinogenesis and tumor promotion. A previous paper showed that genistein down-

regulated the MDM2 oncogene, induced apoptosis and inhibited proliferation in a variety

of human cancer cell lines, regardless of p53 status(Li et al., 2005). This raised an

Page 138: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 122 -

FOXO1

MDM2

Genistein (uM) 0 5 15 50 0 5 15 50Genistein (uM) 0 5 15 50 0 5 15 50

H1299-VEC H1299-M1

Figure 54 Genistein increases the expression of FOXO through MDM2

downregulation. H1299-VEC and H1299-M1 cells were plated in 100 mm

dishes and treated with indicated doses of genistein for 24 h. Cell lysates were

subjected to immunoblotting with anti-FOXO1 and anti-MDM2 antibodies.

IB:FOXO1

IB:MDM2

IB:β-actin β-actin

Page 139: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 123 -

interesting question whether genistein’s anti-tumor activity is mediated through FOXO

factors.

To address this question, parental H1299 cells and H1299 cells that stably

express MDM2 (H1299-M) were plated in 100 mm dishes and treated with genistein at

different doses. The expression of MDM2 and FOXO1 was determined by Western blot

ananlysis. The analyses revealed that genistein decreased MDM2 and increased

FOXO1 level in a dose-dependent manner in H1299 parental cells. In H1299-M cells,

MDM2 expression was detected at a very high level. Genistein treatment had

little effect on the expression of MDM2 or FOXO1 proteins (Figure 54). The data

indicated that genistein increases FOXO1 level through MDM2 down regulation. It also

indicated that FOXO factors may be one of the nuclear targets of genistein in

suppressing tumorigenesis.

10. ARF promotes the MDM2-induced FOXO ubiquitination.

ARF is encoded by the INK4a-ARF locus and it is a known inhibitor of the MDM2

E3 ligase function on p53. To test whether ARF affects MDM2 E3 ligase function of

FOXO factors, H1299 cells were transfected with Flag-FOXO1, MDM2, myc-ARF and

His-ubiquitin as indicated in the figure 53. The cell lysates were purified by Ni-NTA

beads. The ubiquitinated-FOXO1 was detected by M2 anti-Flag antibody. As shown in

Figure 63, ARF increased FOXO1 ubiquitination level to a degree similar to that induced

by MDM2 (Figure 55). The combination of ARF and MDM2 increased the level of

FOXO1 ubiquitination to a degree comparable with that of MDM2 alone, suggesting that

AR may act through MDM2 to regulate FOXO ubiqutination (Figure 55). These data are

quite opposite to p53 ubiquitnation induced by MDM2 but are consistant with our

conclusion that FOXO ubiquitination occurs in the cytoplasm. It is known that ARF

inhibits the target of MDM2 to nucleoli to supprees the ubiquitination of p53.

Page 140: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 124 -

+++++His-Ubiquitin+-+--Myc-ARF++---MDM2++++-FOXO1

+++++His-Ubiquitin+-+--Myc-ARF++---MDM2++++-FOXO1

Ni-NTA pull down

IB:FOXO1

IB:myc

IB:M2

13

18

1107860

47

170

60

78

110

170

IB:MDM2

Ub-FOXO1

ARF

FOXO1

MDM2

Figure 55 ARF promotes the MDM2-induced FOXO1 ubiquitination. H1299

cells were transfected with 2μg Flag-FOXO1, 4 μg MDM2, 4 μg myc-ARF, 4 μg

His-ubiquitin as described on the Figure. Cell extracts were pull-down by Ni-NTA

beads and the expression levels were detected by an anti-FOXO1 antibody.

Page 141: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 125 -

DISCUSSION

Mammlian FOXO factors play very important roles in development, glucose

metabolism, cancer, aging, and energy homeostatis. Several signaling pathways

regulate their activity, such as the insulin pathway, glucose, IGF1 and oxidative stress. In

this study, we found that MDM2 is the E3 ubiquitin ligase for FOXO factors. MDM2

interacts with FOXO factors and promotes their ubiquitination and degradation. It also

affects their biological function and downstream targets expression of FOXO.

1. MDM2 is the E3 ubiquitin ligase of FOXO.

FOXO proteins are known to be ubiquitinated and their level of expression is

regulated by proteasome mediated degradation (Aoki et al., 2004; Plas and Thompson,

2003), but a general ubiquitin E3 ligase for FOXO proteins remains to be identified.

Skp2 was reported to inhibit FOXO1 in tumor suppression through ubiquitin-

mediated degradation but such activity appears to be restricted to FOXO1 (Huang et al.,

2005).

The present study identified MDM2 as a general ubiquitin E3 ligase for

mammalian FOXO factors. We present multiple lines of evidences to support this

conclusion. First, the manipulation of MDM2 expression by genetic deletion, knock down

MDM2 with siRNA or overexpression of MDM2 using transient or stable transfection

leads to change of the level of FOXO1 and FOXO3A proteins in opposite directions.

Second, the down regulation of FOXO1 protein by MDM2 is relieved by treatment with a

proteasome inhibitor, MG132, and the half-life of FOXO1 protein is decreased by ectopic

Page 142: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 126 -

MDM2. Third, MDM2 binds to FOXO1 and FOXO3A and promotes their ubiquitination.

The ubiquitination depends on both the ability of MDM2 to bind FOXO proteins as well

as its E3 ligase activity. This is supported by the evidence that mutant MDM2 (∆150-230)

that is missing in the FOXO binding region did not stimulate FOXO1 ubiquitination

although it contains an intact E3 ring finger domain. Moreover, MDM2 mutants with

either deletion MDM2 (1-361) or mutation MDM2 E3-mt (457S) at the C-terminus ring

finger region lost their ability to stimulate FOXO ubquitination even though they are still

able to interact with FOXO1. Finally, in test tube reaction, recombinant MDM2 catalyzed

the ubiquitination of FOXO1, supporting a direct substrate-enzyme relationship.

2. MDM2-promoted ubiquitnation of FOXO depends on phosphorylation on PKB

sites.

In response to stimulation by IL-3, insulin or PDGF (platelet derived growth

factor), the activated PKB oncogene triggered proteasome-dependent degradation of its

substrates including FOXO1 and FOXO3A (Aoki et al., 2004; Matsuzaki et al., 2003;

Plas and Thompson, 2003). In our study, ubiquitination was found to depend on

phosphorylation at sites that mediate the cytoplasmic localization of FOXO factors by

PKB. FOXO1 (AAA), in which all three PKB sites are mutated to non-phosphorylatable

alanine, did bind to MDM2 (Figure 19) but its level of ubiquitination was not affected by

MDM2 (Figure 29). Similarly, ubiquitination of FOXO3A (AAA) was unaffected by MDM2

(Figure 30). These data suggest that MDM2 acts as a conditional E3 ligase for FOXO

proteins, which is only functional after FOXO phosphorylation at AKT sites. Previous

studies (Feng et al., 2004; Zhou et al., 2001) showed that phosphorylation of MDM2 by

PKB inhibits its interaction with ARF, thereby increasing its stability and promoting its

ability to bind and degrade p53. It remains to be determined whether the MDM2

phosphorylation active PKB regulates its ability to stimulate FOXO ubiquitination.

Page 143: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 127 -

3. MDM2-promoted ubiqutination of FOXO happens mainly in the cytoplasm.

Protein ubiquitination and degradation occur in both the nucleus and cytoplasm

(Yu et al., 2000). In our analysis, the nuclear MDM2 in which the nuclear export

sequence is mutated contains an intact E3 region and is able to bind FOXO1 but unable

to increase FOXO1 ubiquitination, on the other hand, the cytoplasmic MDM2, both the

MDM2 NLS-mt and MDM2 (∆89-150), stimulated ubiquitination, suggesting that it is the

cytoplasmic MDM2 that functions as an E3 ligase for FOXO factors. In combination with

the data that showed that nuclear FOXO1 (AAA) is not ubiquitinated by MDM2, it is

reasonable to assume that FOXO ubiquitination occurs in the cytoplasm. However, such

a conclusion might seem incompatible with the immunofluorescence staining data

showing that the colocalization of MDM2 and FOXO occured mainly in the nucleus. It

should be pointed out that the colcoalization studies were performed in cells grown in a

medium containing low levels of serum, which minimizes PKB activity and inhibits FOXO

cytoplasmic localization and degradation. Consistent with the degradation in the

cytoplasm, the interaction between cytoplasmic MDM2 and FOXO1 was difficult to

detect unless proteasome activity was inhibited by MG132. The fact that MDM2 NLS-mt

that cannot enter the nucleus interacting with FOXO1 and stimulating its ubiquitination

suggests that a nuclear interaction between MDM2 and FOXO proteins is not required

for FOXO ubiquitination. Presently, the details of the nuclear interaction between MDM2

and FOXO proteins are unclear. It is possible that nuclear interaction between FOXO

factors and MDM2 may exert an effect on FOXO factors that is distinct from degradation.

In the case of p53 ubiquitination, it has been shown that monoubiquitination by MDM2

promotes the cytoplasmic localization of p53 whereas the polyubiquintination stimulates

the degradation of p53 in nucleus (Li et al., 2003a).

Page 144: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 128 -

4. MDM2 affects both the transcriptional activity and ubiquitination of FOXO.

FOXO factors are transcription factors. On one hand, FOXO1, FOXO3A and

FOXO4 directly bind to the promoters of TRAIL, Bim, p27, MnSOD, GADD45, G6Pase,

PEPCK, TAT and IGFBP-1 and of others. On the other hand, FOXO can also indirectly

regulate other genes, such as cyclin D1. In our studies, a positive effect of MDM2 on the

transcriptional activity of FOXO1 was detected in transient transfection reporter assays.

It also promoted the ubiquitination and degradation of FOXO factors, leading to FOXO

protein level decreases. It is not fully understood as to how MDM2 decreases FOXO

proteins expression level. There are three possible mechanisms that might be

responsible for MDM2’s positive effect on transcriptional activity of FOXO: 1) MDM2

promotes the transcriptional activity first, followed by its role in FOXO degradation

because transcriptional activity data in H1299/V138 cells showed that FOXO activity was

transiently induced between 5h and 48 h post p53 activation and subsequently FOXO

activity decreased (Figure 47a); 2) FOXO4 was recently shown to be monoubiquitinated,

which increases its transcriptional activity (van der Horst et al., 2006) and this raises the

second possibility. MDM2 plays two roles in the regulation of FOXO factor. It may

promote FOXO transcriptional activity through monoubiquitnation but degrade FOXO

protein through polyubiquitnation; 3) About 20 proteins are reported to interact with

MDM2, and p300 is reported to play a critical role in the MDM2-directed turnover of p53

(Grossman et al., 1998) and this raises the third possibility. Different cofactors are

involved in the regulation of FOXO factors by MDM2 leading to different outcomes.

In our p53 study, Mdm2-mediated monoubiquitylation of p53 greatly promoted its

mitochondrial translocation and thus its apoptosis in the mitochondria. Upon entrance in

the mitochondria, p53 undergoes rapid deubiquitylation by mitochondrial HAUSP via a

stress-induced mitochondrial p53-HAUSP complex (Marchenko et al., 2007). FOXO4

Page 145: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 129 -

was also reported to become monoubiquitinated in response to increased cellular

oxidative stress, resulting in its re-localization to the nucleus and an increase in its

transcriptional activity. Deubiquitination of FOXO is catalyzed by the deubiquitinating

enzyme HAUSP (herpesvirus-associated ubiquitin-specific protease), which interacts

with and deubiquitinates FOXO in response to oxidative stress. We found that MDM2

suppresses the expression of FOXO1 target genes and protects cells from FOXO1-

induced apoptosis. These observations are consistent with the effect of MDM2 on the

translocation change and degradation of FOXO facotors. Further study is required to

understand the biological effect of MDM2 promoted transcriptional activity of FOXO

factors and to estabolish whether FOXO factors have any mitochondrial function.

5. Mammalian FOXO factors interact with p53.

Mammalian FOXO factors and p53 are tumor suppressors and the regulators of

aging that act similarly in many ways. They induce apoptosis (Modur et al., 2002) and

cell cycle arrest (Medema et al., 2000; Nakamura et al., 2000; Schmidt et al., 2002) and

regulate cellular responses to DNA damages and stress (Essers et al., 2004; Kajihara et

al., 2006) through transcriptional induction of a similar set of target genes such as the

p21 CDK inhibitor, Fas ligand and GADD45. In our study, we found that ectopically

expressed p53 inhibited FOXO transcriptional activity in either DU145 cells (contain

inactive p53) or LNCaP (contain wild type p53) but did not decrease FOXO1 induced cell

death (Figure 44b). Two subsequent papers showed that p53 interacted with FOXO3A in

the presence of stress and active FOXO3A could induce p53-dependent apoptosis and

promote p53 cytoplasmic accumulation by increasing its association with the nuclear

exporting machinery. All these evidence indicate that the regulation of transcriptional

activity of p53 and FOXO factor is independent of the regulation of the proapoptotic

activity of p53 and of FOXO factors.

Page 146: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 130 -

p53 and MDM2 form a feedback loop, in which p53 induces MDM2 by activating

MDM2 transcription, and MDM2 in turn negatively regulates p53 by binding and

promoting p53 ubiquitination and degradation (Jin and Levine, 2001). This feedback loop

keeps the p53 activity in check under normal conditions. Upon DNA damages, the

interaction between MDM2 and p53 is suppressed, resulting in an increased p53 activity

that triggers apoptosis. Our data suggest that MDM2 might act as a general coordinator

to turn off multiple negative growth regulators when p53 senses that the DNA damage

has been repaired (Figure 56). During our investigations, several studies documented a

functional interaction between p53 and FOXO3A. In the presence of hydrogen peroxide,

p53 and FOXO3A form a complex (Brunet et al., 2004). DNA damage promotes

FOXO3A nuclear export through p53-dependent activation of serum and glucocorticoid

activated kinases (You et al., 2004a). In endothelial cells (Miyauchi et al., 2004) and

dermal fibroblasts (Kyoung Kim et al., 2005), inhibition of FOXO3A by PKB or siRNA

promoted senescence-like growth arrest in a p53- and p21-dependent manner. Overall,

p53 and FOXO factors appear to have a complex relationship, the outcome of which is

likely to depend on cellular status and environment.

6. MDM2 regulates FOXO factors in a p53 independent way.

In addition to the negative regulation of p53 activity, MDM2 is overexpressed in

tumor cells and functions as an oncogene to promote cancer cell growth independent of

p53 (Daujat et al., 2001; Ganguli and Wasylyk, 2003). Activated FOXO3A was also

reported to impair the transcriptional activity of p53, but enhanced its pro-apoptotic

function in mitochondria (You et al., 2006b). The present studies suggest that

suppression of FOXO factors may be one of the p53-independent mechanisms by which

MDM2 promotes cancer cell survival and cell growth. Decreased FOXO3A and FOXO1

Page 147: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 131 -

MDM2E3 Ligase

p53

FOXO

Proteasome

StimuliOxidative stress,DNA damages,& therapeutic treatments

Ub

UbUb

Ub

UbUb

Degradation

Tumor SuppressionApoptosis,

Senescence,cell cycle arrests,

& DNA repair

MDM2 Overexpressionin Tumor Cells

FOXO

p53

Figure 56 A working model shows the functional interaction among FOXO,

p53 and MDM2 .

Page 148: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 132 -

activity from loss of PTEN led to a decrease in TRAIL expression and increased survival

of prostatic tumor cells (Modur et al., 2002). Loss of p27 expression is associated with

aggressive behavior in a variety of human epithelial tumors (Catzavelos et al., 1997;

Macri and Loda, 1998). Stable expression of MDM2 in H1299 cells decreased the

expression of FOXO3A together with TRAIL, p27 and MnSOD (Figure 35). Knockdown

of MDM2 by siRNA in H1299 cells increased the level of FOXO3A together with TRAIL

(Figure 36). These results suggest that MDM2 promotes tumorigenesis through down-

regulation of FOXO target genes and that the targeted interruption of FOXO

ubiquitination and degradation by MDM2 may represent an effective strategy for cancer

prevention and therapy.

7. Different domains of MDM2 play different roles in the regulation of p53 and

FOXO factors (see Table 2).

FOXO factor and p53 are the central regulators of cellular reponse to stress,

genotoxic insult and DNA damage. MDM2 interacts with and promotes the degradation

of p53. Our study shows that MDM2 also interacts with and promotes the ubiquitination

and degradation of FOXO. But it became quite clear that different domains of MDM2

play different roles in the regulation of these two proteins. MDM2 (50-491), MDM2 (Δ50-

89) and MDM2 (Δ 90-150) cannot bind with p53 and promotes its ubiquitination, but they

do bind with FOXO and can promote the ubquitination of FOXO factors. MDM2 (Δ 150-

230) binds with p53 and promotes the ubiquitinaion of p53 but it does not interact with

FOXO and promotes the ubiquitnation of FOXO. The nucleo-cytoplasmic shuttling is

critical for MDM2-promoted ubiquitination of p53 because even MDM2 (NES mutant)

and MDM2 (NLS mutant) are capable of binding to p53, but cannot promote its

Page 149: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 133 -

N+(Pochampally,1999)

+-+MDM2(361D-A)

N-(Pan,2003)

+-+MDM2(457S)

N-(Kubbutat,1998)

+++MDM2(Δ 222-437)

NND+-+MDM2(Δ 222-325)

C-(Tao, 1999)

+++MDM2(NLS mutant)

N-(Roth,1998)

+-+MDM2(NES mutant)

C+(Dai, 2004)

+--MDM2(Δ 150-230)

C/NND-+±MDM2(Δ 90-150)

NND-++MDM2(Δ50-89)

NND-++MDM2(50-491)

N-(Pochampally,1999)

+-+MDM2(1-361)

N+(Haupt,1997)

+++MDM2

Localization

Promotes the ubquitination of

p53

Binds to

p53

Increases ubiquitination of FOXO1

Binds to

FOXO1

N+(Pochampally,1999)

+-+MDM2(361D-A)

N-(Pan,2003)

+-+MDM2(457S)

N-(Kubbutat,1998)

+++MDM2(Δ 222-437)

NND+-+MDM2(Δ 222-325)

C-(Tao, 1999)

+++MDM2(NLS mutant)

N-(Roth,1998)

+-+MDM2(NES mutant)

C+(Dai, 2004)

+--MDM2(Δ 150-230)

C/NND-+±MDM2(Δ 90-150)

NND-++MDM2(Δ50-89)

NND-++MDM2(50-491)

N-(Pochampally,1999)

+-+MDM2(1-361)

N+(Haupt,1997)

+++MDM2

Localization

Promotes the ubquitination of

p53

Binds to

p53

Increases ubiquitination of FOXO1

Binds to

FOXO1

Table 2 Localization and function of different MDM2 mutants. ND- Not

done yet ; N-nucleus; C-cytoplasm.

Page 150: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 134 -

ubiquination. MDM2 (Δ 222-437) is capable of binding to p53 and FOXO; as for

ubiquitnaiton, this mutant can only promote the ubiquitnation of FOXO but not p53. One

possible reason for this effect is the nucleolar localization of MDM2 (Δ 222-437). MDM2

(1-361) is a mutant which is deleted of the two highly conserved RING finger motifs in

the C-terminus. MDM2 (457S) carries a mutation critical for E3 function of MDM. It

prevents the polyubiquitination of p53, significantly lowers the efficiency of MDM2

interaction with MDMX and promotes MDMX polyubiquitination. Both of these mutants

can bind to p53 and FOXO factors but cannot promote their ubiquitination. This indicates

the essential role of the E3 region of MDM2 in the ubiquitination of both p53 and FOXO.

8. The sumoylation status of SIRT1 affects the stability and activity of FOXO.

Acetylation affects protein degradation via ubiquitination pathway (Ito et al., 2002;

Jeong et al., 2007; Li et al., 2002). In β-cells, acetylation of FOXO1 decreased its

degradation via the ubiquitin pathway (Kitamura et al., 2005). The current study showed

that sumoylation of wild type SIRT1 increased its deacetylase activity,wild type SIRT1,

but not SIRT1 (734R), decreased the acetylation level and transcriptional activity of

FOXO1. This study suggests that sumoylation of SIRT1 affects its ability to regulate the

transcriptional activity of FOXO1. The study also raised the possibility that SIRT1

sumoylation may regulate the stability of FOXO1.

9. Genistein increases FOXO expression level through down-regulation of MDM2.

Genistein (5,7,4'-trihydroxyisoflavone), a kind of isoflavone, is now considered to

be the primary anticancer component of soybeans. Experimental data showed that in

H1299 lung cancer cells, genistein increases the FOXO1 protein expression level in a

dose dependent manner, overexpression of MDM2 abolishes the effect of genestein on

FOXO1 (Figure 54). The data indicates that genstein increases FOXO1 protein

Page 151: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 135 -

expression level and this effect is MDM2-dependent. Previous data showed that

genistein down-regulates MDM2 expression at both the transcriptional and

posttranslational levels, independently of p53, in both human cancer cell lines and

primary cells. Up-regulateion of the tumor suppressor p21Waf1/CIP1 by genistein can be

regulated by both p53 and FOXO factors. Thus, the inhibition of MDM2 expression by

genistein may be essential for its antitumor activities.

10. ARF differentially affects E3 function of MDM2 toward different substrates.

p19ARF is the product of an alternative open reading frame of the mouse INK4a-

ARF locus. It is a known fact that ARF is an inhibitor of the MDM2 E3 ligase function on

p53 (Zhang and Xiong, 2001). In our study, also it was found that ARF stimulated

FOXO1 ubiqutination as well (Figure 55). The data indicate that ARF interaction with

MDM2 differentially affects its E3 function toward different substrates rather than

inactivating its E3 function in general. This effect is plausible because ARF does not

directly interact with the RING domain of MDM2, which may be involved in recruiting E2.

Binding of ARF to the acidic domain of MDM2 may simply alter its ability to properly

orient E2 for the transfer of ubiquitin to certain substrates. In the case of p53, ubiquitin

conjugation is blocked. In the case of FOXO1, ubiquitination is stimulated. There are two

possible explanations for this. The first one is that ARF may stablize MDM2 interaction

with FOXO, which may account for increased FOXO1 ubiqutination. The second one is

that ARF may also qualitatively stimulate the E3 activity of MDM2 toward FOXO. Further

experiments using the in vitro ubiquitination system will be required to address this

matter.

Page 152: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 136 -

SUMMARY AND PERSPECTIVES

Cancer is a major health problem in the USA and worldwide. One third of the

people suffer from some form of cancer and 20% of all deaths are cancer related. In

developed countries, cancer care represents about 10% of total health costs. Since the

incidence of cancer increases with age and people live longer, some important questions

need to be addressed: Why is cancer more prominent in older people and should cancer

treatment be different for the various age groups. Our studies focus on the regulation of

FOXO factors which are key elements of tumor initiation and of the mechanisms that

regulate an organism’s lifespan. They are potential candidates to serve as molecular

linker between longevity and cancer. FOXO factors also regulate numerous cellular

processes, such as stress resistance, the cell cycle, apoptosis, DNA repair/metabolism

and tumorigenesis

Ubiqitination controls the stability/degradation of FOXO factors, although the

mechanism is not clearly defined. Our data allow us to draw the following conclusions:

(1) MDM2 binds directly to FOXO1. Specifically, the binding between MDM2 and FOXO1

is through the central region of MDM2 and the N-terminal region of FOXO1.

Interestingly, FOXO protein phosphorylated by AKT is not required for this binding; (2)

MDM2 binding promotes the ubiquitin-dependent degradation of FOXO1 and FOXO3A.

Knockdown of MDM2 by siRNA caused accumulation of both FOXO1 and FOXO3A.

MDM2 mediated polyubiquitination of FOXO appears to occur in the cytoplasm and is an

AKT phosphorylation dependent; (3) Recombinant MDM2 catalyzes the ubiquitination of

Page 153: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 137 -

FOXO1 in test tube reactions, demonstrating a direct substrate-enzyme relationship; (4)

MDM2 affects the expression of downstream FOXO targets and protects cells from

FOXO1 induced cell death; (5) p53 transiently induced the transcription activity of

FOXO3A, followed by degradation of FOXO3A protein through MDM2.

In addition to the above conclusions, we also made several preliminary but

potentially important observations that need to be investigated in further studies.

1) In the transient transfection experiments, MDM2 increased the transcriptional

activity of both wild type FOXO factors and FOXO (AAA), which is no longer

phosphorylated by AKT anymore. The molecular mechanism underlying this effect

needs further clarification and monoubiquitination could play a decisive role. Other

important questions to be addressed are: whether monoubiquiyination of FOXO factors

is induced by MDM2 and is yes, whether monoubiquitination and polyubiquitination play

different roles in FOXO function; whether monoubiquitination increase the transcriptional

activity of FOXO without an effect on protein stability; whether MDM2 levels has a

differential effect on the ubiqutination status of FOXO; and whether cofactors, such as

p300 and SIRT1, have any effect on the ubiqutination status of FOXO1.

2) Genistein increases the expression of FOXO1 protein in a dose dependent

manner in H1299 lung cancer cells. This effect is abolished in the H1299 cells that stably

express MDM2. Our data suggested that the anti-tumor effect of genistein may be

mediated, at least in part, through the down-regulation of MDM2, causing the up-

regulation of FOXO factors (our present data) and p53 (earlier data of others). It remains

to be seen whether this effect of genistein on FOXO1 can be extended to other FOXO

factors and other cells lines and if so, whether it only affects FOXO protein stability or

through other mechanisms. It is also important to determine how genistein may affect

downstream targets of FOXO factors.

Page 154: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 138 -

3) It is an estabolished fact that PKB and insulin promote FOXO ubiquitination

and degradation. It remains to be elucidated whether MDM2 is required for the PKB and

insulin promoted degradation of FOXO factors and whether the growth factors control

the levels of mono- vs. poly-ubiquitination.

4) SIRT1 acts as a “double-edged sword” promoting survival of aging cells, but

also increasing the cancer risk. Several studies had shown that acetylation inhibits

FOXO protein degradation. One key question is whether SIRT1 affects MDM2 promoted

ubiquitination and degradation, and if so, whether sumoylation mutant of SIRT1 will

affect stability of FOXO since the mutant is associated with a decrease in its deacetylase

activity and with reduced ability to inhibit the transcriptional activity of FOXO1.

5) ARF is known to inhibit MDM2-induced p53 ubiquitination, but enhance

MDM2-induced MDMX ubiquitination. In our study, ARF promoted FOXO ubiquitination.

It is very important to define the molecular mechanism behind the differential effect of

ARF on the MDM2-induced ubiquitination of p53 and FOXO factors.

In summary, my thesis work is the first to identify MDM2 as an E3 ubiquitin ligase

for FOXO factors and suggests that the targeted inhibition of MDM2 E3 activity may be a

more effective strategy for tumor supprerssion than the current strategy that targets

solely the interaction with p53.

Page 155: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 139 -

REFERENCES

Agami, R., Blandino, G., Oren, M., and Shaul, Y. (1999). Interaction of c-Abl and p73alpha and their collaboration to induce apoptosis. Nature 399, 809-813.

Alkhalaf, M., Ganguli, G., Messaddeq, N., Le Meur, M., and Wasylyk, B. (1999). MDM2 overexpression generates a skin phenotype in both wild type and p53 null mice. Oncogene 18, 1419-1434.

Alkhateeb, A., Fain, P.R., and Spritz, R.A. (2005). Candidate functional promoter variant in the FOXD3 melanoblast developmental regulator gene in autosomal dominant vitiligo. The Journal of investigative dermatology 125, 388-391.

Alvarado-Sanchez, B., Hernandez-Castro, B., Portales-Perez, D., Baranda, L., Layseca-Espinosa, E., Abud-Mendoza, C., Cubillas-Tejeda, A.C., and Gonzalez-Amaro, R. (2006). Regulatory T cells in patients with systemic lupus erythematosus. Journal of autoimmunity 27, 110-118.

Amerik, A.Y., Li, S.J., and Hochstrasser, M. (2000). Analysis of the deubiquitinating enzymes of the yeast Saccharomyces cerevisiae. Biological chemistry 381, 981-992.

Anekonda, T.S., and Reddy, P.H. (2006). Neuronal protection by sirtuins in Alzheimer's disease. Journal of neurochemistry 96, 305-313.

Aoki, M., Jiang, H., and Vogt, P.K. (2004). Proteasomal degradation of the FoxO1 transcriptional regulator in cells transformed by the P3k and Akt oncoproteins. Proceedings of the National Academy of Sciences of the United States of America 101, 13613-13617.

Apionishev, S., Malhotra, D., Raghavachari, S., Tanda, S., and Rasooly, R.S. (2001). The Drosophila UBC9 homologue lesswright mediates the disjunction of homologues in meiosis I. Genes Cells 6, 215-224.

Arden, K.C. (2006). Multiple roles of FOXO transcription factors in mammalian cells point to multiple roles in cancer. Experimental gerontology 41, 709-717.

Armoni, M., Harel, C., Karni, S., Chen, H., Bar-Yoseph, F., Ver, M.R., Quon, M.J., and Karnieli, E. (2006). FOXO1 represses peroxisome proliferator-activated receptor-

Page 156: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 140 -

gamma1 and -gamma2 gene promoters in primary adipocytes. A novel paradigm to increase insulin sensitivity. The Journal of biological chemistry 281, 19881-19891.

Aza-Blanc, P., Di Lauro, R., and Santisteban, P. (1993). Identification of a cis-regulatory element and a thyroid-specific nuclear factor mediating the hormonal regulation of rat thyroid peroxidase promoter activity. Molecular endocrinology (Baltimore, Md 7, 1297-1306.

Bailey, D., and O'Hare, P. (2002). Herpes simplex virus 1 ICP0 co-localizes with a SUMO-specific protease. The Journal of general virology 83, 2951-2964.

Bakker, W.J., Blazquez-Domingo, M., Kolbus, A., Besooyen, J., Steinlein, P., Beug, H., Coffer, P.J., Lowenberg, B., von Lindern, M., and van Dijk, T.B. (2004). FoxO3a regulates erythroid differentiation and induces BTG1, an activator of protein arginine methyl transferase 1. The Journal of cell biology 164, 175-184.

Balaban, R.S., Nemoto, S., and Finkel, T. (2005). Mitochondria, oxidants, and aging. Cell 120, 483-495.

Balciunaite, G., Keller, M.P., Balciunaite, E., Piali, L., Zuklys, S., Mathieu, Y.D., Gill, J., Boyd, R., Sussman, D.J., and Hollander, G.A. (2002). Wnt glycoproteins regulate the expression of FoxN1, the gene defective in nude mice. Nature immunology 3, 1102-1108.

Bandyopadhyay, D., Okan, N.A., Bales, E., Nascimento, L., Cole, P.A., and Medrano, E.E. (2002). Down-regulation of p300/CBP histone acetyltransferase activates a senescence checkpoint in human melanocytes. Cancer research 62, 6231-6239.

Banerjee, A.C., Recupero, A.J., Mal, A., Piotrkowski, A.M., Wang, D.M., and Harter, M.L. (1994). The adenovirus E1A 289R and 243R proteins inhibit the phosphorylation of p300. Oncogene 9, 1733-1737.

Bannister, A.J., and Kouzarides, T. (1996). The CBP co-activator is a histone acetyltransferase. Nature 384, 641-643.

Barak, Y., Juven, T., Haffner, R., and Oren, M. (1993). mdm2 expression is induced by wild type p53 activity. The EMBO journal 12, 461-468.

Bartel, F., Taubert, H., and Harris, L.C. (2002). Alternative and aberrant splicing of MDM2 mRNA in human cancer. Cancer cell 2, 9-15.

Baumeister, R., Schaffitzel, E., and Hertweck, M. (2006). Endocrine signaling in Caenorhabditis elegans controls stress response and longevity. The Journal of endocrinology 190, 191-202.

Page 157: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 141 -

Bedalov, A., Gatbonton, T., Irvine, W.P., Gottschling, D.E., and Simon, J.A. (2001). Identification of a small molecule inhibitor of Sir2p. Proceedings of the National Academy of Sciences of the United States of America 98, 15113-15118.

Bennett, C.L., Christie, J., Ramsdell, F., Brunkow, M.E., Ferguson, P.J., Whitesell, L., Kelly, T.E., Saulsbury, F.T., Chance, P.F., and Ochs, H.D. (2001). The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nature genetics 27, 20-21.

Bennin, D.A., Don, A.S., Brake, T., McKenzie, J.L., Rosenbaum, H., Ortiz, L., DePaoli-Roach, A.A., and Horne, M.C. (2002). Cyclin G2 associates with protein phosphatase 2A catalytic and regulatory B' subunits in active complexes and induces nuclear aberrations and a G1/S phase cell cycle arrest. The Journal of biological chemistry 277, 27449-27467.

Best, J.L., Ganiatsas, S., Agarwal, S., Changou, A., Salomoni, P., Shirihai, O., Meluh, P.B., Pandolfi, P.P., and Zon, L.I. (2002). SUMO-1 protease-1 regulates gene transcription through PML. Molecular cell 10, 843-855.

Bieller, A., Pasche, B., Frank, S., Glaser, B., Kunz, J., Witt, K., and Zoll, B. (2001). Isolation and characterization of the human forkhead gene FOXQ1. DNA and cell biology 20, 555-561.

Blixt, A., Landgren, H., Johansson, B.R., and Carlsson, P. (2007). Foxe3 is required for morphogenesis and differentiation of the anterior segment of the eye and is sensitive to Pax6 gene dosage. Developmental biology 302, 218-229.

Bluher, M., Kahn, B.B., and Kahn, C.R. (2003). Extended longevity in mice lacking the insulin receptor in adipose tissue. Science 299, 572-574.

Bordone, L., Motta, M.C., Picard, F., Robinson, A., Jhala, U.S., Apfeld, J., McDonagh, T., Lemieux, M., McBurney, M., Szilvasi, A., Easlon, E.J., Lin, S.J., and Guarente, L. (2006). Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic beta cells. PLoS biology 4, e31.

Borrow, J., Stanton, V.P., Jr., Andresen, J.M., Becher, R., Behm, F.G., Chaganti, R.S., Civin, C.I., Disteche, C., Dube, I., Frischauf, A.M., Horsman, D., Mitelman, F., Volinia, S., Watmore, A.E., and Housman, D.E. (1996). The translocation t(8;16)(p11;p13) of acute myeloid leukaemia fuses a putative acetyltransferase to the CREB-binding protein. Nature genetics 14, 33-41.

Bouras, T., Fu, M., Sauve, A.A., Wang, F., Quong, A.A., Perkins, N.D., Hay, R.T., Gu, W., and Pestell, R.G. (2005). SIRT1 deacetylation and repression of p300 involves lysine residues 1020/1024 within the cell cycle regulatory domain 1. The Journal of biological chemistry 280, 10264-10276.

Page 158: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 142 -

Boyd, S.D., Tsai, K.Y., and Jacks, T. (2000). An intact HDM2 RING-finger domain is required for nuclear exclusion of p53. Nature cell biology 2, 563-568.

Brownawell, A.M., Kops, G.J., Macara, I.G., and Burgering, B.M. (2001). Inhibition of nuclear import by protein kinase B (Akt) regulates the subcellular distribution and activity of the forkhead transcription factor AFX. Molecular and cellular biology 21, 3534-3546.

Brunet, A., Bonni, A., Zigmond, M.J., Lin, M.Z., Juo, P., Hu, L.S., Anderson, M.J., Arden, K.C., Blenis, J., and Greenberg, M.E. (1999). Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96, 857-868.

Brunet, A., Park, J., Tran, H., Hu, L.S., Hemmings, B.A., and Greenberg, M.E. (2001). Protein kinase SGK mediates survival signals by phosphorylating the forkhead transcription factor FKHRL1 (FOXO3a). Molecular and cellular biology 21, 952-965.

Brunet, A., Sweeney, L.B., Sturgill, J.F., Chua, K.F., Greer, P.L., Lin, Y., Tran, H., Ross, S.E., Mostoslavsky, R., Cohen, H.Y., Hu, L.S., Cheng, H.L., Jedrychowski, M.P., Gygi, S.P., Sinclair, D.A., Alt, F.W., and Greenberg, M.E. (2004). Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 303, 2011-2015.

Brunkow, M.E., Jeffery, E.W., Hjerrild, K.A., Paeper, B., Clark, L.B., Yasayko, S.A., Wilkinson, J.E., Galas, D., Ziegler, S.F., and Ramsdell, F. (2001). Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nature genetics 27, 68-73.

Buschmann, T., Lerner, D., Lee, C.G., and Ronai, Z. (2001). The Mdm-2 amino terminus is required for Mdm2 binding and SUMO-1 conjugation by the E2 SUMO-1 conjugating enzyme Ubc9. The Journal of biological chemistry 276, 40389-40395.

Cahilly-Snyder, L., Yang-Feng, T., Francke, U., and George, D.L. (1987). Molecular analysis and chromosomal mapping of amplified genes isolated from a transformed mouse 3T3 cell line. Somatic cell and molecular genetics 13, 235-244.

Carlsson, P., and Mahlapuu, M. (2002). Forkhead transcription factors: key players in development and metabolism. Developmental biology 250, 1-23.

Carter, M.E., and Brunet, A. (2007). FOXO transcription factors. Curr Biol 17, R113-114.

Caspari, T. (2000). How to activate p53. Curr Biol 10, R315-317.

Castrillon, D.H., Miao, L., Kollipara, R., Horner, J.W., and DePinho, R.A. (2003). Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a. Science 301, 215-218.

Page 159: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 143 -

Catzavelos, C., Bhattacharya, N., Ung, Y.C., Wilson, J.A., Roncari, L., Sandhu, C., Shaw, P., Yeger, H., Morava-Protzner, I., Kapusta, L., Franssen, E., Pritchard, K.I., and Slingerland, J.M. (1997). Decreased levels of the cell-cycle inhibitor p27Kip1 protein: prognostic implications in primary breast cancer. Nature medicine 3, 227-230.

Chen, A., Mannen, H., and Li, S.S. (1998). Characterization of mouse ubiquitin-like SMT3A and SMT3B cDNAs and gene/pseudogenes. Biochemistry and molecular biology international 46, 1161-1174.

Chen, J., Lin, J., and Levine, A.J. (1995). Regulation of transcription functions of the p53 tumor suppressor by the mdm-2 oncogene. Molecular medicine (Cambridge, Mass 1, 142-152.

Chen, J., Marechal, V., and Levine, A.J. (1993). Mapping of the p53 and mdm-2 interaction domains. Molecular and cellular biology 13, 4107-4114.

Chen, J., Wu, X., Lin, J., and Levine, A.J. (1996). mdm-2 inhibits the G1 arrest and apoptosis functions of the p53 tumor suppressor protein. Molecular and cellular biology 16, 2445-2452.

Chen, J., Yusuf, I., Andersen, H.M., and Fruman, D.A. (2006). FOXO transcription factors cooperate with delta EF1 to activate growth suppressive genes in B lymphocytes. J Immunol 176, 2711-2721.

Chen, L., Marechal, V., Moreau, J., Levine, A.J., and Chen, J. (1997). Proteolytic cleavage of the mdm2 oncoprotein during apoptosis. The Journal of biological chemistry 272, 22966-22973.

Chevillard-Briet, M., Trouche, D., and Vandel, L. (2002). Control of CBP co-activating activity by arginine methylation. The EMBO journal 21, 5457-5466.

Chua, K.F., Mostoslavsky, R., Lombard, D.B., Pang, W.W., Saito, S., Franco, S., Kaushal, D., Cheng, H.L., Fischer, M.R., Stokes, N., Murphy, M.M., Appella, E., and Alt, F.W. (2005). Mammalian SIRT1 limits replicative life span in response to chronic genotoxic stress. Cell metabolism 2, 67-76.

Ciechanover, A., and Schwartz, A.L. (2002). Ubiquitin-mediated degradation of cellular proteins in health and disease. Hepatology (Baltimore, Md 35, 3-6.

Classon, M., Salama, S., Gorka, C., Mulloy, R., Braun, P., and Harlow, E. (2000). Combinatorial roles for pRB, p107, and p130 in E2F-mediated cell cycle control. Proceedings of the National Academy of Sciences of the United States of America 97, 10820-10825.

Page 160: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 144 -

Cohen, T., and Yao, T.P. (2004). AcK-knowledge reversible acetylation. Sci STKE 2004, pe42.

Czech, M.P. (2003). Insulin's expanding control of forkheads. Proceedings of the National Academy of Sciences of the United States of America 100, 11198-11200.

D'Andrea, A., and Pellman, D. (1998). Deubiquitinating enzymes: a new class of biological regulators. Critical reviews in biochemistry and molecular biology 33, 337-352.

Dai, M.S., Zeng, S.X., Jin, Y., Sun, X.X., David, L., and Lu, H. (2004). Ribosomal protein L23 activates p53 by inhibiting MDM2 function in response to ribosomal perturbation but not to translation inhibition. Molecular and cellular biology 24, 7654-7668.

Daitoku, H., Hatta, M., Matsuzaki, H., Aratani, S., Ohshima, T., Miyagishi, M., Nakajima, T., and Fukamizu, A. (2004). Silent information regulator 2 potentiates Foxo1-mediated transcription through its deacetylase activity. Proceedings of the National Academy of Sciences of the United States of America 101, 10042-10047.

Dansen, T.B., Kops, G.J., Denis, S., Jelluma, N., Wanders, R.J., Bos, J.L., Burgering, B.M., and Wirtz, K.W. (2004). Regulation of sterol carrier protein gene expression by the forkhead transcription factor FOXO3a. Journal of lipid research 45, 81-88.

Daujat, S., Neel, H., and Piette, J. (2001). MDM2: life without p53. Trends Genet 17, 459-464.

Dazard, J.E., Augias, D., Neel, H., Mils, V., Marechal, V., Basset-Seguin, N., and Piette, J. (1997). MDM-2 protein is expressed in different layers of normal human skin. Oncogene 14, 1123-1128.

De Baere, E., Dixon, M.J., Small, K.W., Jabs, E.W., Leroy, B.P., Devriendt, K., Gillerot, Y., Mortier, G., Meire, F., Van Maldergem, L., Courtens, W., Hjalgrim, H., Huang, S., Liebaers, I., Van Regemorter, N., Touraine, P., Praphanphoj, V., Verloes, A., Udar, N., Yellore, V., Chalukya, M., Yelchits, S., De Paepe, A., Kuttenn, F., Fellous, M., Veitia, R., and Messiaen, L. (2001). Spectrum of FOXL2 gene mutations in blepharophimosis-ptosis-epicanthus inversus (BPES) families demonstrates a genotype--phenotype correlation. Human molecular genetics 10, 1591-1600.

De Baere, E., Lemercier, B., Christin-Maitre, S., Durval, D., Messiaen, L., Fellous, M., and Veitia, R. (2002). FOXL2 mutation screening in a large panel of POF patients and XX males. Journal of medical genetics 39, e43.

de Ruijter, A.J., van Gennip, A.H., Caron, H.N., Kemp, S., and van Kuilenburg, A.B. (2003). Histone deacetylases (HDACs): characterization of the classical HDAC family. The Biochemical journal 370, 737-749.

Page 161: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 145 -

Deguchi, K., Ayton, P.M., Carapeti, M., Kutok, J.L., Snyder, C.S., Williams, I.R., Cross, N.C., Glass, C.K., Cleary, M.L., and Gilliland, D.G. (2003). MOZ-TIF2-induced acute myeloid leukemia requires the MOZ nucleosome binding motif and TIF2-mediated recruitment of CBP. Cancer cell 3, 259-271.

Desterro, J.M., Rodriguez, M.S., and Hay, R.T. (1998). SUMO-1 modification of IkappaBalpha inhibits NF-kappaB activation. Molecular cell 2, 233-239.

Epps, J.L., and Tanda, S. (1998). The Drosophila semushi mutation blocks nuclear import of bicoid during embryogenesis. Curr Biol 8, 1277-1280.

Essers, M.A., de Vries-Smits, L.M., Barker, N., Polderman, P.E., Burgering, B.M., and Korswagen, H.C. (2005). Functional interaction between beta-catenin and FOXO in oxidative stress signaling. Science 308, 1181-1184.

Essers, M.A., Weijzen, S., de Vries-Smits, A.M., Saarloos, I., de Ruiter, N.D., Bos, J.L., and Burgering, B.M. (2004). FOXO transcription factor activation by oxidative stress mediated by the small GTPase Ral and JNK. The EMBO journal 23, 4802-4812.

Fakharzadeh, S.S., Trusko, S.P., and George, D.L. (1991). Tumorigenic potential associated with enhanced expression of a gene that is amplified in a mouse tumor cell line. The EMBO journal 10, 1565-1569.

Fang, S., Jensen, J.P., Ludwig, R.L., Vousden, K.H., and Weissman, A.M. (2000). Mdm2 is a RING finger-dependent ubiquitin protein ligase for itself and p53. The Journal of biological chemistry 275, 8945-8951.

Feng, J., Tamaskovic, R., Yang, Z., Brazil, D.P., Merlo, A., Hess, D., and Hemmings, B.A. (2004). Stabilization of Mdm2 via decreased ubiquitination is mediated by protein kinase B/Akt-dependent phosphorylation. The Journal of biological chemistry 279, 35510-35517.

Fiddler, T.A., Smith, L., Tapscott, S.J., and Thayer, M.J. (1996). Amplification of MDM2 inhibits MyoD-mediated myogenesis. Molecular and cellular biology 16, 5048-5057.

Fornace, A.J., Jr., Nebert, D.W., Hollander, M.C., Luethy, J.D., Papathanasiou, M., Fargnoli, J., and Holbrook, N.J. (1989). Mammalian genes coordinately regulated by growth arrest signals and DNA-damaging agents. Molecular and cellular biology 9, 4196-4203.

Frank, J., Pignata, C., Panteleyev, A.A., Prowse, D.M., Baden, H., Weiner, L., Gaetaniello, L., Ahmad, W., Pozzi, N., Cserhalmi-Friedman, P.B., Aita, V.M., Uyttendaele, H., Gordon, D., Ott, J., Brissette, J.L., and Christiano, A.M. (1999). Exposing the human nude phenotype. Nature 398, 473-474.

Page 162: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 146 -

Fraser, A.G., Kamath, R.S., Zipperlen, P., Martinez-Campos, M., Sohrmann, M., and Ahringer, J. (2000). Functional genomic analysis of C. elegans chromosome I by systematic RNA interference. Nature 408, 325-330.

Freedman, D.A., and Levine, A.J. (1999). Regulation of the p53 protein by the MDM2 oncoprotein--thirty-eighth G.H.A. Clowes Memorial Award Lecture. Cancer Res 59, 1-7.

Fukuoka, M., Daitoku, H., Hatta, M., Matsuzaki, H., Umemura, S., and Fukamizu, A. (2003). Negative regulation of forkhead transcription factor AFX (Foxo4) by CBP-induced acetylation. International journal of molecular medicine 12, 503-508.

Furukawa-Hibi, Y., Yoshida-Araki, K., Ohta, T., Ikeda, K., and Motoyama, N. (2002). FOXO forkhead transcription factors induce G(2)-M checkpoint in response to oxidative stress. The Journal of biological chemistry 277, 26729-26732.

Furuyama, T., Kitayama, K., Yamashita, H., and Mori, N. (2003). Forkhead transcription factor FOXO1 (FKHR)-dependent induction of PDK4 gene expression in skeletal muscle during energy deprivation. The Biochemical journal 375, 365-371.

Galili, N., Davis, R.J., Fredericks, W.J., Mukhopadhyay, S., Rauscher, F.J., 3rd, Emanuel, B.S., Rovera, G., and Barr, F.G. (1993). Fusion of a fork head domain gene to PAX3 in the solid tumour alveolar rhabdomyosarcoma. Nature genetics 5, 230-235.

Ganguli, G., and Wasylyk, B. (2003). p53-independent functions of MDM2. Mol Cancer Res 1, 1027-1035.

Gao, N., Zhang, J., Rao, M.A., Case, T.C., Mirosevich, J., Wang, Y., Jin, R., Gupta, A., Rennie, P.S., and Matusik, R.J. (2003). The role of hepatocyte nuclear factor-3 alpha (Forkhead Box A1) and androgen receptor in transcriptional regulation of prostatic genes. Molecular endocrinology (Baltimore, Md 17, 1484-1507.

Gayther, S.A., Batley, S.J., Linger, L., Bannister, A., Thorpe, K., Chin, S.F., Daigo, Y., Russell, P., Wilson, A., Sowter, H.M., Delhanty, J.D., Ponder, B.A., Kouzarides, T., and Caldas, C. (2000). Mutations truncating the EP300 acetylase in human cancers. Nature genetics 24, 300-303.

Geyer, R.K., Yu, Z.K., and Maki, C.G. (2000). The MDM2 RING-finger domain is required to promote p53 nuclear export. Nature cell biology 2, 569-573.

Giannakou, M.E., Goss, M., Junger, M.A., Hafen, E., Leevers, S.J., and Partridge, L. (2004). Long-lived Drosophila with overexpressed dFOXO in adult fat body. Science 305, 361.

Giannakou, M.E., and Partridge, L. (2004). The interaction between FOXO and SIRT1: tipping the balance towards survival. Trends in cell biology 14, 408-412.

Page 163: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 147 -

Gillett, C.E., Smith, P., Peters, G., Lu, X., and Barnes, D.M. (1999). Cyclin-dependent kinase inhibitor p27Kip1 expression and interaction with other cell cycle-associated proteins in mammary carcinoma. The Journal of pathology 187, 200-206.

Gilley, J., Coffer, P.J., and Ham, J. (2003). FOXO transcription factors directly activate bim gene expression and promote apoptosis in sympathetic neurons. The Journal of cell biology 162, 613-622.

Girdwood, D., Bumpass, D., Vaughan, O.A., Thain, A., Anderson, L.A., Snowden, A.W., Garcia-Wilson, E., Perkins, N.D., and Hay, R.T. (2003). P300 transcriptional repression is mediated by SUMO modification. Molecular cell 11, 1043-1054.

Glauser, D.A., and Schlegel, W. (2007). The emerging role of FOXO transcription factors in pancreatic beta cells. The Journal of endocrinology 193, 195-207.

Goldberg, Z., Vogt Sionov, R., Berger, M., Zwang, Y., Perets, R., Van Etten, R.A., Oren, M., Taya, Y., and Haupt, Y. (2002). Tyrosine phosphorylation of Mdm2 by c-Abl: implications for p53 regulation. The EMBO journal 21, 3715-3727.

Gong, J.G., Costanzo, A., Yang, H.Q., Melino, G., Kaelin, W.G., Jr., Levrero, M., and Wang, J.Y. (1999). The tyrosine kinase c-Abl regulates p73 in apoptotic response to cisplatin-induced DNA damage. Nature 399, 806-809.

Goodman, R.H., and Smolik, S. (2000). CBP/p300 in cell growth, transformation, and development. Genes & development 14, 1553-1577.

Gotz, C., Kartarius, S., Scholtes, P., Nastainczyk, W., and Montenarh, M. (1999). Identification of a CK2 phosphorylation site in mdm2. European journal of biochemistry / FEBS 266, 493-501.

Granadino, B., Arias-de-la-Fuente, C., Perez-Sanchez, C., Parraga, M., Lopez-Fernandez, L.A., del Mazo, J., and Rey-Campos, J. (2000). Fhx (Foxj2) expression is activated during spermatogenesis and very early in embryonic development. Mechanisms of development 97, 157-160.

Gray, D.A., Inazawa, J., Gupta, K., Wong, A., Ueda, R., and Takahashi, T. (1995). Elevated expression of Unph, a proto-oncogene at 3p21.3, in human lung tumors. Oncogene 10, 2179-2183.

Greer, E.L., and Brunet, A. (2005). FOXO transcription factors at the interface between longevity and tumor suppression. Oncogene 24, 7410-7425.

Grossman, S.R., Perez, M., Kung, A.L., Joseph, M., Mansur, C., Xiao, Z.X., Kumar, S., Howley, P.M., and Livingston, D.M. (1998). p300/MDM2 complexes participate in MDM2-mediated p53 degradation. Molecular cell 2, 405-415.

Page 164: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 148 -

Guo, Y., Costa, R., Ramsey, H., Starnes, T., Vance, G., Robertson, K., Kelley, M., Reinbold, R., Scholer, H., and Hromas, R. (2002). The embryonic stem cell transcription factors Oct-4 and FoxD3 interact to regulate endodermal-specific promoter expression. Proceedings of the National Academy of Sciences of the United States of America 99, 3663-3667.

Gusterson, R.J., Jazrawi, E., Adcock, I.M., and Latchman, D.S. (2003). The transcriptional co-activators CREB-binding protein (CBP) and p300 play a critical role in cardiac hypertrophy that is dependent on their histone acetyltransferase activity. The Journal of biological chemistry 278, 6838-6847.

Hartwell, L.H., and Weinert, T.A. (1989). Checkpoints: controls that ensure the order of cell cycle events. Science 246, 629-634.

Hathaway, G.M., and Traugh, J.A. (1979). Cyclic nucleotide-independent protein kinases from rabbit reticulocytes. Purification of casein kinases. The Journal of biological chemistry 254, 762-768.

Hatini, V., Huh, S.O., Herzlinger, D., Soares, V.C., and Lai, E. (1996). Essential role of stromal mesenchyme in kidney morphogenesis revealed by targeted disruption of Winged Helix transcription factor BF-2. Genes & development 10, 1467-1478.

Hauck, L., Harms, C., Grothe, D., An, J., Gertz, K., Kronenberg, G., Dietz, R., Endres, M., and von Harsdorf, R. (2007). Critical role for FoxO3a-dependent regulation of p21CIP1/WAF1 in response to statin signaling in cardiac myocytes. Circulation research 100, 50-60.

Haupt, Y., Maya, R., Kazaz, A., and Oren, M. (1997). Mdm2 promotes the rapid degradation of p53. Nature 387, 296-299.

Hawke, T.J., Jiang, N., and Garry, D.J. (2003). Absence of p21CIP rescues myogenic progenitor cell proliferative and regenerative capacity in Foxk1 null mice. The Journal of biological chemistry 278, 4015-4020.

Hay, T.J., and Meek, D.W. (2000). Multiple sites of in vivo phosphorylation in the MDM2 oncoprotein cluster within two important functional domains. FEBS letters 478, 183-186.

Hayashi, T., Seki, M., Maeda, D., Wang, W., Kawabe, Y., Seki, T., Saitoh, H., Fukagawa, T., Yagi, H., and Enomoto, T. (2002). Ubc9 is essential for viability of higher eukaryotic cells. Experimental cell research 280, 212-221.

Henning, W., Rohaly, G., Kolzau, T., Knippschild, U., Maacke, H., and Deppert, W. (1997). MDM2 is a target of simian virus 40 in cellular transformation and during lytic infection. Journal of virology 71, 7609-7618.

Page 165: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 149 -

Hershko, A., and Ciechanover, A. (1998). The ubiquitin system. Annual review of biochemistry 67, 425-479.

Hicke, L. (2001). Protein regulation by monoubiquitin. Nat Rev Mol Cell Biol 2, 195-201.

Hillion, J., Le Coniat, M., Jonveaux, P., Berger, R., and Bernard, O.A. (1997). AF6q21, a novel partner of the MLL gene in t(6;11)(q21;q23), defines a forkhead transcriptional factor subfamily. Blood 90, 3714-3719.

Hoege, C., Pfander, B., Moldovan, G.L., Pyrowolakis, G., and Jentsch, S. (2002). RAD6-dependent DNA repair is linked to modification of PCNA by ubiquitin and SUMO. Nature 419, 135-141.

Hoekman, M.F., Jacobs, F.M., Smidt, M.P., and Burbach, J.P. (2006). Spatial and temporal expression of FoxO transcription factors in the developing and adult murine brain. Gene Expr Patterns 6, 134-140.

Holowaty, M.N., Sheng, Y., Nguyen, T., Arrowsmith, C., and Frappier, L. (2003). Protein interaction domains of the ubiquitin-specific protease, USP7/HAUSP. The Journal of biological chemistry 278, 47753-47761.

Holzenberger, M., Dupont, J., Ducos, B., Leneuve, P., Geloen, A., Even, P.C., Cervera, P., and Le Bouc, Y. (2003). IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 421, 182-187.

Honda, R., Tanaka, H., and Yasuda, H. (1997). Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS letters 420, 25-27.

Honda, R., and Yasuda, H. (1999). Association of p19(ARF) with Mdm2 inhibits ubiquitin ligase activity of Mdm2 for tumor suppressor p53. The EMBO journal 18, 22-27.

Honda, R., and Yasuda, H. (2000). Activity of MDM2, a ubiquitin ligase, toward p53 or itself is dependent on the RING finger domain of the ligase. Oncogene 19, 1473-1476.

Hong, H.K., Noveroske, J.K., Headon, D.J., Liu, T., Sy, M.S., Justice, M.J., and Chakravarti, A. (2001). The winged helix/forkhead transcription factor Foxq1 regulates differentiation of hair in satin mice. Genesis 29, 163-171.

Horne, M.C., Donaldson, K.L., Goolsby, G.L., Tran, D., Mulheisen, M., Hell, J.W., and Wahl, A.F. (1997). Cyclin G2 is up-regulated during growth inhibition and B cell antigen receptor-mediated cell cycle arrest. The Journal of biological chemistry 272, 12650-12661.

Horne, M.C., Goolsby, G.L., Donaldson, K.L., Tran, D., Neubauer, M., and Wahl, A.F. (1996). Cyclin G1 and cyclin G2 comprise a new family of cyclins with contrasting tissue-

Page 166: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 150 -

specific and cell cycle-regulated expression. The Journal of biological chemistry 271, 6050-6061.

Hosaka, T., Biggs, W.H., 3rd, Tieu, D., Boyer, A.D., Varki, N.M., Cavenee, W.K., and Arden, K.C. (2004). Disruption of forkhead transcription factor (FOXO) family members in mice reveals their functional diversification. Proceedings of the National Academy of Sciences of the United States of America 101, 2975-2980.

Howe, K., Williamson, J., Boddy, N., Sheer, D., Freemont, P., and Solomon, E. (1998). The ubiquitin-homology gene PIC1: characterization of mouse (Pic1) and human (UBL1) genes and pseudogenes. Genomics 47, 92-100.

Howitz, K.T., Bitterman, K.J., Cohen, H.Y., Lamming, D.W., Lavu, S., Wood, J.G., Zipkin, R.E., Chung, P., Kisielewski, A., Zhang, L.L., Scherer, B., and Sinclair, D.A. (2003). Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 425, 191-196.

Hu, M., Li, P., Li, M., Li, W., Yao, T., Wu, J.W., Gu, W., Cohen, R.E., and Shi, Y. (2002). Crystal structure of a UBP-family deubiquitinating enzyme in isolation and in complex with ubiquitin aldehyde. Cell 111, 1041-1054.

Hu, M.C., Lee, D.F., Xia, W., Golfman, L.S., Ou-Yang, F., Yang, J.Y., Zou, Y., Bao, S., Hanada, N., Saso, H., Kobayashi, R., and Hung, M.C. (2004). IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a. Cell 117, 225-237.

Huang, H., Regan, K.M., Lou, Z., Chen, J., and Tindall, D.J. (2006). CDK2-dependent phosphorylation of FOXO1 as an apoptotic response to DNA damage. Science 314, 294-297.

Huang, H., Regan, K.M., Wang, F., Wang, D., Smith, D.I., van Deursen, J.M., and Tindall, D.J. (2005). Skp2 inhibits FOXO1 in tumor suppression through ubiquitin-mediated degradation. Proceedings of the National Academy of Sciences of the United States of America 102, 1649-1654.

Huang, Y., Baker, R.T., and Fischer-Vize, J.A. (1995). Control of cell fate by a deubiquitinating enzyme encoded by the fat facets gene. Science 270, 1828-1831.

Huang, Y., and Fischer-Vize, J.A. (1996). Undifferentiated cells in the developing Drosophila eye influence facet assembly and require the Fat facets ubiquitin-specific protease. Development (Cambridge, England) 122, 3207-3216.

Hulander, M., Kiernan, A.E., Blomqvist, S.R., Carlsson, P., Samuelsson, E.J., Johansson, B.R., Steel, K.P., and Enerback, S. (2003). Lack of pendrin expression leads to deafness and expansion of the endolymphatic compartment in inner ears of Foxi1 null mutant mice. Development (Cambridge, England) 130, 2013-2025.

Page 167: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 151 -

Hurst, J.A., Baraitser, M., Auger, E., Graham, F., and Norell, S. (1990). An extended family with a dominantly inherited speech disorder. Developmental medicine and child neurology 32, 352-355.

Hwangbo, D.S., Gershman, B., Tu, M.P., Palmer, M., and Tatar, M. (2004). Drosophila dFOXO controls lifespan and regulates insulin signalling in brain and fat body. Nature 429, 562-566.

Imai, S., Johnson, F.B., Marciniak, R.A., McVey, M., Park, P.U., and Guarente, L. (2000). Sir2: an NAD-dependent histone deacetylase that connects chromatin silencing, metabolism, and aging. Cold Spring Harbor symposia on quantitative biology 65, 297-302.

Ito, A., Kawaguchi, Y., Lai, C.H., Kovacs, J.J., Higashimoto, Y., Appella, E., and Yao, T.P. (2002). MDM2-HDAC1-mediated deacetylation of p53 is required for its degradation. The EMBO journal 21, 6236-6245.

Jacobs, F.M., van der Heide, L.P., Wijchers, P.J., Burbach, J.P., Hoekman, M.F., and Smidt, M.P. (2003). FoxO6, a novel member of the FoxO class of transcription factors with distinct shuttling dynamics. The Journal of biological chemistry 278, 35959-35967.

Jensen, D.E., Proctor, M., Marquis, S.T., Gardner, H.P., Ha, S.I., Chodosh, L.A., Ishov, A.M., Tommerup, N., Vissing, H., Sekido, Y., Minna, J., Borodovsky, A., Schultz, D.C., Wilkinson, K.D., Maul, G.G., Barlev, N., Berger, S.L., Prendergast, G.C., and Rauscher, F.J., 3rd (1998). BAP1: a novel ubiquitin hydrolase which binds to the BRCA1 RING finger and enhances BRCA1-mediated cell growth suppression. Oncogene 16, 1097-1112.

Jeong, J., Juhn, K., Lee, H., Kim, S.H., Min, B.H., Lee, K.M., Cho, M.H., Park, G.H., and Lee, K.H. (2007). SIRT1 promotes DNA repair activity and deacetylation of Ku70. Experimental & molecular medicine 39, 8-13.

Jeong, J.W., Bae, M.K., Ahn, M.Y., Kim, S.H., Sohn, T.K., Bae, M.H., Yoo, M.A., Song, E.J., Lee, K.J., and Kim, K.W. (2002). Regulation and destabilization of HIF-1alpha by ARD1-mediated acetylation. Cell 111, 709-720.

Jiang, M., Shao, Z.M., Wu, J., Lu, J.S., Yu, L.M., Yuan, J.D., Han, Q.X., Shen, Z.Z., and Fontana, J.A. (1997). p21/waf1/cip1 and mdm-2 expression in breast carcinoma patients as related to prognosis. International journal of cancer 74, 529-534.

Jin, S., and Levine, A.J. (2001). The p53 functional circuit. Journal of cell science 114, 4139-4140.

Joanisse, D.R., Inaguma, Y., and Tanguay, R.M. (1998). Cloning and developmental expression of a nuclear ubiquitin-conjugating enzyme (DmUbc9) that interacts with small

Page 168: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 152 -

heat shock proteins in Drosophila melanogaster. Biochemical and biophysical research communications 244, 102-109.

Johnson, E.S. (2004). Protein modification by SUMO. Annual review of biochemistry 73, 355-382.

Jones, D., Crowe, E., Stevens, T.A., and Candido, E.P. (2002). Functional and phylogenetic analysis of the ubiquitylation system in Caenorhabditis elegans: ubiquitin-conjugating enzymes, ubiquitin-activating enzymes, and ubiquitin-like proteins. Genome biology 3, RESEARCH0002.

Jones, S.N., Roe, A.E., Donehower, L.A., and Bradley, A. (1995). Rescue of embryonic lethality in Mdm2-deficient mice by absence of p53. Nature 378, 206-208.

Junger, M.A., Rintelen, F., Stocker, H., Wasserman, J.D., Vegh, M., Radimerski, T., Greenberg, M.E., and Hafen, E. (2003). The Drosophila forkhead transcription factor FOXO mediates the reduction in cell number associated with reduced insulin signaling. Journal of biology 2, 20.

Juven-Gershon, T., Shifman, O., Unger, T., Elkeles, A., Haupt, Y., and Oren, M. (1998). The Mdm2 oncoprotein interacts with the cell fate regulator Numb. Molecular and cellular biology 18, 3974-3982.

Kadoya, T., Yamamoto, H., Suzuki, T., Yukita, A., Fukui, A., Michiue, T., Asahara, T., Tanaka, K., Asashima, M., and Kikuchi, A. (2002). Desumoylation activity of Axam, a novel Axin-binding protein, is involved in downregulation of beta-catenin. Molecular and cellular biology 22, 3803-3819.

Kaeberlein, M., McVey, M., and Guarente, L. (1999). The SIR2/3/4 complex and SIR2 alone promote longevity in Saccharomyces cerevisiae by two different mechanisms. Genes & development 13, 2570-2580.

Kaestner, K.H. (2000). The hepatocyte nuclear factor 3 (HNF3 or FOXA) family in metabolism. Trends in endocrinology and metabolism: TEM 11, 281-285.

Kaestner, K.H., Hiemisch, H., Luckow, B., and Schutz, G. (1994). The HNF-3 gene family of transcription factors in mice: gene structure, cDNA sequence, and mRNA distribution. Genomics 20, 377-385.

Kaestner, K.H., Knochel, W., and Martinez, D.E. (2000). Unified nomenclature for the winged helix/forkhead transcription factors. Genes & development 14, 142-146.

Kaestner, K.H., Silberg, D.G., Traber, P.G., and Schutz, G. (1997). The mesenchymal winged helix transcription factor Fkh6 is required for the control of gastrointestinal proliferation and differentiation. Genes & development 11, 1583-1595.

Page 169: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 153 -

Kajihara, T., Jones, M., Fusi, L., Takano, M., Feroze-Zaidi, F., Pirianov, G., Mehmet, H., Ishihara, O., Higham, J.M., Lam, E.W., and Brosens, J.J. (2006). Differential expression of FOXO1 and FOXO3a confers resistance to oxidative cell death upon endometrial decidualization. Molecular endocrinology (Baltimore, Md 20, 2444-2455.

Kalinichenko, V.V., Lim, L., Stolz, D.B., Shin, B., Rausa, F.M., Clark, J., Whitsett, J.A., Watkins, S.C., and Costa, R.H. (2001). Defects in pulmonary vasculature and perinatal lung hemorrhage in mice heterozygous null for the Forkhead Box f1 transcription factor. Developmental biology 235, 489-506.

Kalinichenko, V.V., Zhou, Y., Shin, B., Stolz, D.B., Watkins, S.C., Whitsett, J.A., and Costa, R.H. (2002). Wild-type levels of the mouse Forkhead Box f1 gene are essential for lung repair. American journal of physiology 282, L1253-1265.

Kamei, Y., Mizukami, J., Miura, S., Suzuki, M., Takahashi, N., Kawada, T., Taniguchi, T., and Ezaki, O. (2003). A forkhead transcription factor FKHR up-regulates lipoprotein lipase expression in skeletal muscle. FEBS letters 536, 232-236.

Kamitani, T., Kito, K., Nguyen, H.P., Fukuda-Kamitani, T., and Yeh, E.T. (1998). Characterization of a second member of the sentrin family of ubiquitin-like proteins. The Journal of biological chemistry 273, 11349-11353.

Katoh, M., and Katoh, M. (2004a). Germ-line mutation of Foxn5 gene in mouse lineage. International journal of molecular medicine 14, 463-467.

Katoh, M., and Katoh, M. (2004b). Human FOX gene family (Review). International journal of oncology 25, 1495-1500.

Katoh, M., and Katoh, M. (2004c). Identification and characterization of human FOXN5 and rat Foxn5 genes in silico. International journal of oncology 24, 1339-1344.

Katoh, M., and Katoh, M. (2004d). Identification and characterization of human FOXN6, mouse Foxn6, and rat Foxn6 genes in silico. International journal of oncology 25, 219-223.

Kenyon, C., Chang, J., Gensch, E., Rudner, A., and Tabtiang, R. (1993). A C. elegans mutant that lives twice as long as wild type. Nature 366, 461-464.

Kim, K.I., Baek, S.H., Jeon, Y.J., Nishimori, S., Suzuki, T., Uchida, S., Shimbara, N., Saitoh, H., Tanaka, K., and Chung, C.H. (2000). A new SUMO-1-specific protease, SUSP1, that is highly expressed in reproductive organs. The Journal of biological chemistry 275, 14102-14106.

Page 170: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 154 -

Kimura, K.D., Tissenbaum, H.A., Liu, Y., and Ruvkun, G. (1997). daf-2, an insulin receptor-like gene that regulates longevity and diapause in Caenorhabditis elegans. Science 277, 942-946.

Kirkwood, T.B., and Austad, S.N. (2000). Why do we age? Nature 408, 233-238.

Kitamura, T., Nakae, J., Kitamura, Y., Kido, Y., Biggs, W.H., 3rd, Wright, C.V., White, M.F., Arden, K.C., and Accili, D. (2002). The forkhead transcription factor Foxo1 links insulin signaling to Pdx1 regulation of pancreatic beta cell growth. The Journal of clinical investigation 110, 1839-1847.

Kitamura, Y.I., Kitamura, T., Kruse, J.P., Raum, J.C., Stein, R., Gu, W., and Accili, D. (2005). FoxO1 protects against pancreatic beta cell failure through NeuroD and MafA induction. Cell metabolism 2, 153-163.

Kloetzli, J.M., Fontaine-Glover, I.A., Brown, E.R., Kuo, M., and Labosky, P.A. (2001). The winged helix gene, Foxb1, controls development of mammary glands and regions of the CNS that regulate the milk-ejection reflex. Genesis 29, 60-71.

Kobayashi, T., and Cohen, P. (1999). Activation of serum- and glucocorticoid-regulated protein kinase by agonists that activate phosphatidylinositide 3-kinase is mediated by 3-phosphoinositide-dependent protein kinase-1 (PDK1) and PDK2. The Biochemical journal 339 ( Pt 2), 319-328.

Kops, G.J., Dansen, T.B., Polderman, P.E., Saarloos, I., Wirtz, K.W., Coffer, P.J., Huang, T.T., Bos, J.L., Medema, R.H., and Burgering, B.M. (2002). Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature 419, 316-321.

Korver, W., Roose, J., and Clevers, H. (1997). The winged-helix transcription factor Trident is expressed in cycling cells. Nucleic acids research 25, 1715-1719.

Kramer, J.M., Davidge, J.T., Lockyer, J.M., and Staveley, B.E. (2003). Expression of Drosophila FOXO regulates growth and can phenocopy starvation. BMC developmental biology 3, 5.

Kubbutat, M.H., Jones, S.N., and Vousden, K.H. (1997). Regulation of p53 stability by Mdm2. Nature 387, 299-303.

Kume, T., Deng, K., and Hogan, B.L. (2000). Murine forkhead/winged helix genes Foxc1 (Mf1) and Foxc2 (Mfh1) are required for the early organogenesis of the kidney and urinary tract. Development (Cambridge, England) 127, 1387-1395.

Page 171: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 155 -

Kume, T., Deng, K.Y., Winfrey, V., Gould, D.B., Walter, M.A., and Hogan, B.L. (1998). The forkhead/winged helix gene Mf1 is disrupted in the pleiotropic mouse mutation congenital hydrocephalus. Cell 93, 985-996.

Kurepa, J., Walker, J.M., Smalle, J., Gosink, M.M., Davis, S.J., Durham, T.L., Sung, D.Y., and Vierstra, R.D. (2003). The small ubiquitin-like modifier (SUMO) protein modification system in Arabidopsis. Accumulation of SUMO1 and -2 conjugates is increased by stress. The Journal of biological chemistry 278, 6862-6872.

Kussie, P.H., Gorina, S., Marechal, V., Elenbaas, B., Moreau, J., Levine, A.J., and Pavletich, N.P. (1996). Structure of the MDM2 oncoprotein bound to the p53 tumor suppressor transactivation domain. Science 274, 948-953.

Kwok, R.P., Liu, X.T., and Smith, G.D. (2006). Distribution of co-activators CBP and p300 during mouse oocyte and embryo development. Molecular reproduction and development 73, 885-894.

Kwon, H.S., Huang, B., Unterman, T.G., and Harris, R.A. (2004). Protein kinase B-alpha inhibits human pyruvate dehydrogenase kinase-4 gene induction by dexamethasone through inactivation of FOXO transcription factors. Diabetes 53, 899-910.

Kyoung Kim, H., Kyoung Kim, Y., Song, I.H., Baek, S.H., Lee, S.R., Hye Kim, J., and Kim, J.R. (2005). Down-regulation of a forkhead transcription factor, FOXO3a, accelerates cellular senescence in human dermal fibroblasts. J Gerontol A Biol Sci Med Sci 60, 4-9.

Lai, C.S., Fisher, S.E., Hurst, J.A., Vargha-Khadem, F., and Monaco, A.P. (2001). A forkhead-domain gene is mutated in a severe speech and language disorder. Nature 413, 519-523.

Lam, E.W., Francis, R.E., and Petkovic, M. (2006). FOXO transcription factors: key regulators of cell fate. Biochemical Society transactions 34, 722-726.

Landgren, H., and Carlsson, P. (2004). FoxJ3, a novel mammalian forkhead gene expressed in neuroectoderm, neural crest, and myotome. Dev Dyn 231, 396-401.

Langley, E., Pearson, M., Faretta, M., Bauer, U.M., Frye, R.A., Minucci, S., Pelicci, P.G., and Kouzarides, T. (2002). Human SIR2 deacetylates p53 and antagonizes PML/p53-induced cellular senescence. The EMBO journal 21, 2383-2396.

Lawlor, M.A., and Rotwein, P. (2000). Insulin-like growth factor-mediated muscle cell survival: central roles for Akt and cyclin-dependent kinase inhibitor p21. Molecular and cellular biology 20, 8983-8995.

Page 172: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 156 -

Lee, P.S., Chang, C., Liu, D., and Derynck, R. (2003). Sumoylation of Smad4, the common Smad mediator of transforming growth factor-beta family signaling. The Journal of biological chemistry 278, 27853-27863.

Lee, T.A., and Tyers, M. (2001). Ubiquitin junction, what's your function? Genome biology 2, REPORTS4025.

Lehmann, O.J., Sowden, J.C., Carlsson, P., Jordan, T., and Bhattacharya, S.S. (2003). Fox's in development and disease. Trends Genet 19, 339-344.

Lehtinen, M.K., Yuan, Z., Boag, P.R., Yang, Y., Villen, J., Becker, E.B., DiBacco, S., de la Iglesia, N., Gygi, S., Blackwell, T.K., and Bonni, A. (2006). A conserved MST-FOXO signaling pathway mediates oxidative-stress responses and extends life span. Cell 125, 987-1001.

Leibiger, I.B., and Berggren, P.O. (2006). Sirt1: a metabolic master switch that modulates lifespan. Nature medicine 12, 34-36; discussion 36.

Lengner, C.J., Steinman, H.A., Gagnon, J., Smith, T.W., Henderson, J.E., Kream, B.E., Stein, G.S., Lian, J.B., and Jones, S.N. (2006). Osteoblast differentiation and skeletal development are regulated by Mdm2-p53 signaling. The Journal of cell biology 172, 909-921.

Li, C., Lusis, A.J., Sparkes, R., Tran, S.M., and Gaynor, R. (1992). Characterization and chromosomal mapping of the gene encoding the cellular DNA binding protein HTLF. Genomics 13, 658-664.

Li, M., Brooks, C.L., Wu-Baer, F., Chen, D., Baer, R., and Gu, W. (2003a). Mono- versus polyubiquitination: differential control of p53 fate by Mdm2. Science 302, 1972-1975.

Li, M., Luo, J., Brooks, C.L., and Gu, W. (2002). Acetylation of p53 inhibits its ubiquitination by Mdm2. The Journal of biological chemistry 277, 50607-50611.

Li, M., Zhang, Z., Hill, D.L., Chen, X., Wang, H., and Zhang, R. (2005). Genistein, a dietary isoflavone, down-regulates the MDM2 oncogene at both transcriptional and posttranslational levels. Cancer research 65, 8200-8208.

Li, P., Lee, H., Guo, S., Unterman, T.G., Jenster, G., and Bai, W. (2003b). AKT-independent protection of prostate cancer cells from apoptosis mediated through complex formation between the androgen receptor and FKHR. Molecular and cellular biology 23, 104-118.

Li, P., Nicosia, S.V., and Bai, W. (2001). Antagonism between PTEN/MMAC1/TEP-1 and androgen receptor in growth and apoptosis of prostatic cancer cells. The Journal of biological chemistry 276, 20444-20450.

Page 173: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 157 -

Li, S., Mo, Z., Yang, X., Price, S.M., Shen, M.M., and Xiang, M. (2004a). Foxn4 controls the genesis of amacrine and horizontal cells by retinal progenitors. Neuron 43, 795-807.

Li, S., Zhou, D., Lu, M.M., and Morrisey, E.E. (2004b). Advanced cardiac morphogenesis does not require heart tube fusion. Science 305, 1619-1622.

Lin, H.K., Wang, L., Hu, Y.C., Altuwaijri, S., and Chang, C. (2002a). Phosphorylation-dependent ubiquitylation and degradation of androgen receptor by Akt require Mdm2 E3 ligase. The EMBO journal 21, 4037-4048.

Lin, J., Chen, J., Elenbaas, B., and Levine, A.J. (1994). Several hydrophobic amino acids in the p53 amino-terminal domain are required for transcriptional activation, binding to mdm-2 and the adenovirus 5 E1B 55-kD protein. Genes & development 8, 1235-1246.

Lin, K., Hsin, H., Libina, N., and Kenyon, C. (2001). Regulation of the Caenorhabditis elegans longevity protein DAF-16 by insulin/IGF-1 and germline signaling. Nature genetics 28, 139-145.

Lin, L., Miller, C.T., Contreras, J.I., Prescott, M.S., Dagenais, S.L., Wu, R., Yee, J., Orringer, M.B., Misek, D.E., Hanash, S.M., Glover, T.W., and Beer, D.G. (2002b). The hepatocyte nuclear factor 3 alpha gene, HNF3alpha (FOXA1), on chromosome band 14q13 is amplified and overexpressed in esophageal and lung adenocarcinomas. Cancer research 62, 5273-5279.

Lin, X., Liang, M., Liang, Y.Y., Brunicardi, F.C., and Feng, X.H. (2003a). SUMO-1/Ubc9 promotes nuclear accumulation and metabolic stability of tumor suppressor Smad4. The Journal of biological chemistry 278, 31043-31048.

Lin, X., Liang, M., Liang, Y.Y., Brunicardi, F.C., Melchior, F., and Feng, X.H. (2003b). Activation of transforming growth factor-beta signaling by SUMO-1 modification of tumor suppressor Smad4/DPC4. The Journal of biological chemistry 278, 18714-18719.

Lohrum, M.A., Ashcroft, M., Kubbutat, M.H., and Vousden, K.H. (2000). Identification of a cryptic nucleolar-localization signal in MDM2. Nature cell biology 2, 179-181.

Lohrum, M.A., Ludwig, R.L., Kubbutat, M.H., Hanlon, M., and Vousden, K.H. (2003). Regulation of HDM2 activity by the ribosomal protein L11. Cancer cell 3, 577-587.

Lois, L.M., Lima, C.D., and Chua, N.H. (2003). Small ubiquitin-like modifier modulates abscisic acid signaling in Arabidopsis. The Plant cell 15, 1347-1359.

Lundgren, K., Montes de Oca Luna, R., McNeill, Y.B., Emerick, E.P., Spencer, B., Barfield, C.R., Lozano, G., Rosenberg, M.P., and Finlay, C.A. (1997). Targeted

Page 174: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 158 -

expression of MDM2 uncouples S phase from mitosis and inhibits mammary gland development independent of p53. Genes & development 11, 714-725.

Luo, J., Nikolaev, A.Y., Imai, S., Chen, D., Su, F., Shiloh, A., Guarente, L., and Gu, W. (2001). Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell 107, 137-148.

Macri, E., and Loda, M. (1998). Role of p27 in prostate carcinogenesis. Cancer metastasis reviews 17, 337-344.

Mahmud, D.L., M, G.A., Deb, D.K., Platanias, L.C., Uddin, S., and Wickrema, A. (2002). Phosphorylation of forkhead transcription factors by erythropoietin and stem cell factor prevents acetylation and their interaction with coactivator p300 in erythroid progenitor cells. Oncogene 21, 1556-1562.

Mannen, H., Tseng, H.M., Cho, C.L., and Li, S.S. (1996). Cloning and expression of human homolog HSMT3 to yeast SMT3 suppressor of MIF2 mutations in a centromere protein gene. Biochemical and biophysical research communications 222, 178-180.

Marchenko, N.D., Wolff, S., Erster, S., Becker, K., and Moll, U.M. (2007). Monoubiquitylation promotes mitochondrial p53 translocation. The EMBO journal 26, 923-934.

Marechal, V., Elenbaas, B., Piette, J., Nicolas, J.C., and Levine, A.J. (1994). The ribosomal L5 protein is associated with mdm-2 and mdm-2-p53 complexes. Molecular and cellular biology 14, 7414-7420.

Martinez-Gac, L., Marques, M., Garcia, Z., Campanero, M.R., and Carrera, A.C. (2004). Control of cyclin G2 mRNA expression by forkhead transcription factors: novel mechanism for cell cycle control by phosphoinositide 3-kinase and forkhead. Molecular and cellular biology 24, 2181-2189.

Martinez, S.C., Cras-Meneur, C., Bernal-Mizrachi, E., and Permutt, M.A. (2006). Glucose regulates Foxo1 through insulin receptor signaling in the pancreatic islet beta-cell. Diabetes 55, 1581-1591.

Matsuzaki, H., Daitoku, H., Hatta, M., Aoyama, H., Yoshimochi, K., and Fukamizu, A. (2005). Acetylation of Foxo1 alters its DNA-binding ability and sensitivity to phosphorylation. Proceedings of the National Academy of Sciences of the United States of America 102, 11278-11283.

Matsuzaki, H., Daitoku, H., Hatta, M., Tanaka, K., and Fukamizu, A. (2003). Insulin-induced phosphorylation of FKHR (Foxo1) targets to proteasomal degradation. Proceedings of the National Academy of Sciences of the United States of America 100, 11285-11290.

Page 175: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 159 -

Maya, R., Balass, M., Kim, S.T., Shkedy, D., Leal, J.F., Shifman, O., Moas, M., Buschmann, T., Ronai, Z., Shiloh, Y., Kastan, M.B., Katzir, E., and Oren, M. (2001). ATM-dependent phosphorylation of Mdm2 on serine 395: role in p53 activation by DNA damage. Genes & development 15, 1067-1077.

Mayo, L.D., and Donner, D.B. (2001). A phosphatidylinositol 3-kinase/Akt pathway promotes translocation of Mdm2 from the cytoplasm to the nucleus. Proceedings of the National Academy of Sciences of the United States of America 98, 11598-11603.

Mayo, L.D., Turchi, J.J., and Berberich, S.J. (1997). Mdm-2 phosphorylation by DNA-dependent protein kinase prevents interaction with p53. Cancer research 57, 5013-5016.

Mazet, F., Yu, J.K., Liberles, D.A., Holland, L.Z., and Shimeld, S.M. (2003). Phylogenetic relationships of the Fox (Forkhead) gene family in the Bilateria. Gene 316, 79-89.

McBurney, M.W., Yang, X., Jardine, K., Bieman, M., Th'ng, J., and Lemieux, M. (2003). The absence of SIR2alpha protein has no effect on global gene silencing in mouse embryonic stem cells. Mol Cancer Res 1, 402-409.

McCoy, M.A., Gesell, J.J., Senior, M.M., and Wyss, D.F. (2003). Flexible lid to the p53-binding domain of human Mdm2: implications for p53 regulation. Proceedings of the National Academy of Sciences of the United States of America 100, 1645-1648.

Medema, R.H., Kops, G.J., Bos, J.L., and Burgering, B.M. (2000). AFX-like Forkhead transcription factors mediate cell-cycle regulation by Ras and PKB through p27kip1. Nature 404, 782-787.

Meek, D.W. (1994). Post-translational modification of p53. Seminars in cancer biology 5, 203-210.

Mihara, M., Erster, S., Zaika, A., Petrenko, O., Chittenden, T., Pancoska, P., and Moll, U.M. (2003). p53 has a direct apoptogenic role at the mitochondria. Molecular cell 11, 577-590.

Mimnaugh, E.G., Chen, H.Y., Davie, J.R., Celis, J.E., and Neckers, L. (1997). Rapid deubiquitination of nucleosomal histones in human tumor cells caused by proteasome inhibitors and stress response inducers: effects on replication, transcription, translation, and the cellular stress response. Biochemistry 36, 14418-14429.

Minoretti, P., Arra, M., Emanuele, E., Olivieri, V., Aldeghi, A., Politi, P., Martinelli, V., Pesenti, S., and Falcone, C. (2007). A W148R mutation in the human FOXD4 gene segregating with dilated cardiomyopathy, obsessive-compulsive disorder, and suicidality. International journal of molecular medicine 19, 369-372.

Page 176: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 160 -

Miwa, S., Uchida, C., Kitagawa, K., Hattori, T., Oda, T., Sugimura, H., Yasuda, H., Nakamura, H., Chida, K., and Kitagawa, M. (2006). Mdm2-mediated pRB downregulation is involved in carcinogenesis in a p53-independent manner. Biochemical and biophysical research communications 340, 54-61.

Miyauchi, H., Minamino, T., Tateno, K., Kunieda, T., Toko, H., and Komuro, I. (2004). Akt negatively regulates the in vitro lifespan of human endothelial cells via a p53/p21-dependent pathway. The EMBO journal 23, 212-220.

Moazed, D., and Johnson, D. (1996). A deubiquitinating enzyme interacts with SIR4 and regulates silencing in S. cerevisiae. Cell 86, 667-677.

Modur, V., Nagarajan, R., Evers, B.M., and Milbrandt, J. (2002). FOXO proteins regulate tumor necrosis factor-related apoptosis inducing ligand expression. Implications for PTEN mutation in prostate cancer. The Journal of biological chemistry 277, 47928-47937.

Momand, J., Zambetti, G.P., Olson, D.C., George, D., and Levine, A.J. (1992). The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell 69, 1237-1245.

Montes de Oca Luna, R., Wagner, D.S., and Lozano, G. (1995). Rescue of early embryonic lethality in mdm2-deficient mice by deletion of p53. Nature 378, 203-206.

Mossessova, E., and Lima, C.D. (2000). Ulp1-SUMO crystal structure and genetic analysis reveal conserved interactions and a regulatory element essential for cell growth in yeast. Molecular cell 5, 865-876.

Motta, M.C., Divecha, N., Lemieux, M., Kamel, C., Chen, D., Gu, W., Bultsma, Y., McBurney, M., and Guarente, L. (2004). Mammalian SIRT1 represses forkhead transcription factors. Cell 116, 551-563.

Muraoka, M., Konishi, M., Kikuchi-Yanoshita, R., Tanaka, K., Shitara, N., Chong, J.M., Iwama, T., and Miyaki, M. (1996). p300 gene alterations in colorectal and gastric carcinomas. Oncogene 12, 1565-1569.

Nadal, A., Marrero, P.F., and Haro, D. (2002). Down-regulation of the mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase gene by insulin: the role of the forkhead transcription factor FKHRL1. The Biochemical journal 366, 289-297.

Nakae, J., Biggs, W.H., 3rd, Kitamura, T., Cavenee, W.K., Wright, C.V., Arden, K.C., and Accili, D. (2002). Regulation of insulin action and pancreatic beta-cell function by mutated alleles of the gene encoding forkhead transcription factor Foxo1. Nature genetics 32, 245-253.

Page 177: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 161 -

Nakae, J., Cao, Y., Daitoku, H., Fukamizu, A., Ogawa, W., Yano, Y., and Hayashi, Y. (2006). The LXXLL motif of murine forkhead transcription factor FoxO1 mediates Sirt1-dependent transcriptional activity. The Journal of clinical investigation 116, 2473-2483.

Nakae, J., Kitamura, T., Kitamura, Y., Biggs, W.H., 3rd, Arden, K.C., and Accili, D. (2003). The forkhead transcription factor Foxo1 regulates adipocyte differentiation. Developmental cell 4, 119-129.

Nakae, J., Kitamura, T., Silver, D.L., and Accili, D. (2001). The forkhead transcription factor Foxo1 (Fkhr) confers insulin sensitivity onto glucose-6-phosphatase expression. The Journal of clinical investigation 108, 1359-1367.

Nakamura, N., Ramaswamy, S., Vazquez, F., Signoretti, S., Loda, M., and Sellers, W.R. (2000). Forkhead transcription factors are critical effectors of cell death and cell cycle arrest downstream of PTEN. Molecular and cellular biology 20, 8969-8982.

Nakamura, T., Furukawa, Y., Nakagawa, H., Tsunoda, T., Ohigashi, H., Murata, K., Ishikawa, O., Ohgaki, K., Kashimura, N., Miyamoto, M., Hirano, S., Kondo, S., Katoh, H., Nakamura, Y., and Katagiri, T. (2004). Genome-wide cDNA microarray analysis of gene expression profiles in pancreatic cancers using populations of tumor cells and normal ductal epithelial cells selected for purity by laser microdissection. Oncogene 23, 2385-2400.

Ng, I.O., Lam, K.Y., Ng, M., and Regezi, J.A. (1999). Expression of p21/waf1 in oral squamous cell carcinomas--correlation with p53 and mdm2 and cellular proliferation index. Oral oncology 35, 63-69.

Nishida, T., Kaneko, F., Kitagawa, M., and Yasuda, H. (2001). Characterization of a novel mammalian SUMO-1/Smt3-specific isopeptidase, a homologue of rat axam, which is an axin-binding protein promoting beta-catenin degradation. The Journal of biological chemistry 276, 39060-39066.

Nishida, T., Tanaka, H., and Yasuda, H. (2000). A novel mammalian Smt3-specific isopeptidase 1 (SMT3IP1) localized in the nucleolus at interphase. European journal of biochemistry / FEBS 267, 6423-6427.

O'Connor, L., Strasser, A., O'Reilly, L.A., Hausmann, G., Adams, J.M., Cory, S., and Huang, D.C. (1998). Bim: a novel member of the Bcl-2 family that promotes apoptosis. The EMBO journal 17, 384-395.

Ogg, S., Paradis, S., Gottlieb, S., Patterson, G.I., Lee, L., Tissenbaum, H.A., and Ruvkun, G. (1997). The Fork head transcription factor DAF-16 transduces insulin-like metabolic and longevity signals in C. elegans. Nature 389, 994-999.

Page 178: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 162 -

Ogryzko, V.V., Schiltz, R.L., Russanova, V., Howard, B.H., and Nakatani, Y. (1996). The transcriptional coactivators p300 and CBP are histone acetyltransferases. Cell 87, 953-959.

Oliner, J.D., Kinzler, K.W., Meltzer, P.S., George, D.L., and Vogelstein, B. (1992). Amplification of a gene encoding a p53-associated protein in human sarcomas. Nature 358, 80-83.

Olson, D.C., Marechal, V., Momand, J., Chen, J., Romocki, C., and Levine, A.J. (1993). Identification and characterization of multiple mdm-2 proteins and mdm-2-p53 protein complexes. Oncogene 8, 2353-2360.

Onuma, H., Vander Kooi, B.T., Boustead, J.N., Oeser, J.K., and O'Brien, R.M. (2006). Correlation between FOXO1a (FKHR) and FOXO3a (FKHRL1) binding and the inhibition of basal glucose-6-phosphatase catalytic subunit gene transcription by insulin. Molecular endocrinology (Baltimore, Md 20, 2831-2847.

Ortiz, L., Zannini, M., Di Lauro, R., and Santisteban, P. (1997). Transcriptional control of the forkhead thyroid transcription factor TTF-2 by thyrotropin, insulin, and insulin-like growth factor I. The Journal of biological chemistry 272, 23334-23339.

Pan, Y., and Chen, J. (2003). MDM2 promotes ubiquitination and degradation of MDMX. Molecular and cellular biology 23, 5113-5121.

Papa, F.R., and Hochstrasser, M. (1993). The yeast DOA4 gene encodes a deubiquitinating enzyme related to a product of the human tre-2 oncogene. Nature 366, 313-319.

Papathanasiou, M.A., Kerr, N.C., Robbins, J.H., McBride, O.W., Alamo, I., Jr., Barrett, S.F., Hickson, I.D., and Fornace, A.J., Jr. (1991). Induction by ionizing radiation of the gadd45 gene in cultured human cells: lack of mediation by protein kinase C. Molecular and cellular biology 11, 1009-1016.

Parry, P., Wei, Y., and Evans, G. (1994). Cloning and characterization of the t(X;11) breakpoint from a leukemic cell line identify a new member of the forkhead gene family. Genes, chromosomes & cancer 11, 79-84.

Pauley, S., Lai, E., and Fritzsch, B. (2006). Foxg1 is required for morphogenesis and histogenesis of the mammalian inner ear. Dev Dyn 235, 2470-2482.

Pelletier, G.J., Brody, S.L., Liapis, H., White, R.A., and Hackett, B.P. (1998). A human forkhead/winged-helix transcription factor expressed in developing pulmonary and renal epithelium. The American journal of physiology 274, L351-359.

Page 179: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 163 -

Peng, Y., Chen, L., Li, C., Lu, W., and Chen, J. (2001). Inhibition of MDM2 by hsp90 contributes to mutant p53 stabilization. The Journal of biological chemistry 276, 40583-40590.

Perrot, V., and Rechler, M.M. (2005). The coactivator p300 directly acetylates the forkhead transcription factor Foxo1 and stimulates Foxo1-induced transcription. Molecular endocrinology (Baltimore, Md 19, 2283-2298.

Perry, M.E., Piette, J., Zawadzki, J.A., Harvey, D., and Levine, A.J. (1993). The mdm-2 gene is induced in response to UV light in a p53-dependent manner. Proceedings of the National Academy of Sciences of the United States of America 90, 11623-11627.

Picard, F., Kurtev, M., Chung, N., Topark-Ngarm, A., Senawong, T., Machado De Oliveira, R., Leid, M., McBurney, M.W., and Guarente, L. (2004). Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-gamma. Nature 429, 771-776.

Pickart, C.M. (2001). Mechanisms underlying ubiquitination. Annual review of biochemistry 70, 503-533.

Picksley, S.M., Vojtesek, B., Sparks, A., and Lane, D.P. (1994). Immunochemical analysis of the interaction of p53 with MDM2;--fine mapping of the MDM2 binding site on p53 using synthetic peptides. Oncogene 9, 2523-2529.

Plas, D.R., and Thompson, C.B. (2003). Akt activation promotes degradation of tuberin and FOXO3a via the proteasome. The Journal of biological chemistry 278, 12361-12366.

Pochampally, R., Fodera, B., Chen, L., Lu, W., and Chen, J. (1999). Activation of an MDM2-specific caspase by p53 in the absence of apoptosis. The Journal of biological chemistry 274, 15271-15277.

Polesskaya, A., Duquet, A., Naguibneva, I., Weise, C., Vervisch, A., Bengal, E., Hucho, F., Robin, P., and Harel-Bellan, A. (2000). CREB-binding protein/p300 activates MyoD by acetylation. The Journal of biological chemistry 275, 34359-34364.

Potente, M., Urbich, C., Sasaki, K., Hofmann, W.K., Heeschen, C., Aicher, A., Kollipara, R., DePinho, R.A., Zeiher, A.M., and Dimmeler, S. (2005). Involvement of Foxo transcription factors in angiogenesis and postnatal neovascularization. The Journal of clinical investigation 115, 2382-2392.

Puig, O., Marr, M.T., Ruhf, M.L., and Tjian, R. (2003). Control of cell number by Drosophila FOXO: downstream and feedback regulation of the insulin receptor pathway. Genes & development 17, 2006-2020.

Page 180: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 164 -

Ramaswamy, S., Nakamura, N., Sansal, I., Bergeron, L., and Sellers, W.R. (2002). A novel mechanism of gene regulation and tumor suppression by the transcription factor FKHR. Cancer cell 2, 81-91.

Rena, G., Woods, Y.L., Prescott, A.R., Peggie, M., Unterman, T.G., Williams, M.R., and Cohen, P. (2002). Two novel phosphorylation sites on FKHR that are critical for its nuclear exclusion. The EMBO journal 21, 2263-2271.

Robzyk, K., Recht, J., and Osley, M.A. (2000). Rad6-dependent ubiquitination of histone H2B in yeast. Science 287, 501-504.

Rogina, B., and Helfand, S.L. (2004). Sir2 mediates longevity in the fly through a pathway related to calorie restriction. Proceedings of the National Academy of Sciences of the United States of America 101, 15998-16003.

Ross, S., Best, J.L., Zon, L.I., and Gill, G. (2002). SUMO-1 modification represses Sp3 transcriptional activation and modulates its subnuclear localization. Molecular cell 10, 831-842.

Sakaguchi, K., Herrera, J.E., Saito, S., Miki, T., Bustin, M., Vassilev, A., Anderson, C.W., and Appella, E. (1998). DNA damage activates p53 through a phosphorylation-acetylation cascade. Genes & development 12, 2831-2841.

Samuel, V.T., Choi, C.S., Phillips, T.G., Romanelli, A.J., Geisler, J.G., Bhanot, S., McKay, R., Monia, B., Shutter, J.R., Lindberg, R.A., Shulman, G.I., and Veniant, M.M. (2006). Targeting foxo1 in mice using antisense oligonucleotide improves hepatic and peripheral insulin action. Diabetes 55, 2042-2050.

Sandri, M., Sandri, C., Gilbert, A., Skurk, C., Calabria, E., Picard, A., Walsh, K., Schiaffino, S., Lecker, S.H., and Goldberg, A.L. (2004). Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117, 399-412.

Saucedo, L.J., Myers, C.D., and Perry, M.E. (1999). Multiple murine double minute gene 2 (MDM2) proteins are induced by ultraviolet light. The Journal of biological chemistry 274, 8161-8168.

Schmidt, M., Fernandez de Mattos, S., van der Horst, A., Klompmaker, R., Kops, G.J., Lam, E.W., Burgering, B.M., and Medema, R.H. (2002). Cell cycle inhibition by FoxO forkhead transcription factors involves downregulation of cyclin D. Molecular and cellular biology 22, 7842-7852.

Scott, K.L., and Plon, S.E. (2005). CHES1/FOXN3 interacts with Ski-interacting protein and acts as a transcriptional repressor. Gene 359, 119-126.

Page 181: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 165 -

Seoane, J., Le, H.V., Shen, L., Anderson, S.A., and Massague, J. (2004). Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell 117, 211-223.

Shen, Z., Pardington-Purtymun, P.E., Comeaux, J.C., Moyzis, R.K., and Chen, D.J. (1996). UBL1, a human ubiquitin-like protein associating with human RAD51/RAD52 proteins. Genomics 36, 271-279.

Siliciano, J.D., Canman, C.E., Taya, Y., Sakaguchi, K., Appella, E., and Kastan, M.B. (1997). DNA damage induces phosphorylation of the amino terminus of p53. Genes & development 11, 3471-3481.

Sionov, R.V., Moallem, E., Berger, M., Kazaz, A., Gerlitz, O., Ben-Neriah, Y., Oren, M., and Haupt, Y. (1999). c-Abl neutralizes the inhibitory effect of Mdm2 on p53. The Journal of biological chemistry 274, 8371-8374.

Sorensen, P.H., Lynch, J.C., Qualman, S.J., Tirabosco, R., Lim, J.F., Maurer, H.M., Bridge, J.A., Crist, W.M., Triche, T.J., and Barr, F.G. (2002). PAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicators in alveolar rhabdomyosarcoma: a report from the children's oncology group. J Clin Oncol 20, 2672-2679.

Stahl, M., Dijkers, P.F., Kops, G.J., Lens, S.M., Coffer, P.J., Burgering, B.M., and Medema, R.H. (2002). The forkhead transcription factor FoxO regulates transcription of p27Kip1 and Bim in response to IL-2. J Immunol 168, 5024-5031.

Stein, J.P., Ginsberg, D.A., Grossfeld, G.D., Chatterjee, S.J., Esrig, D., Dickinson, M.G., Groshen, S., Taylor, C.R., Jones, P.A., Skinner, D.G., and Cote, R.J. (1998). Effect of p21WAF1/CIP1 expression on tumor progression in bladder cancer. Journal of the National Cancer Institute 90, 1072-1079.

Storz, P. (2006). Reactive oxygen species-mediated mitochondria-to-nucleus signaling: a key to aging and radical-caused diseases. Sci STKE 2006, re3.

Suliman, A., Lam, A., Datta, R., and Srivastava, R.K. (2001). Intracellular mechanisms of TRAIL: apoptosis through mitochondrial-dependent and -independent pathways. Oncogene 20, 2122-2133.

Sun, P., Dong, P., Dai, K., Hannon, G.J., and Beach, D. (1998). p53-independent role of MDM2 in TGF-beta1 resistance. Science 282, 2270-2272.

Sunters, A., Fernandez de Mattos, S., Stahl, M., Brosens, J.J., Zoumpoulidou, G., Saunders, C.A., Coffer, P.J., Medema, R.H., Coombes, R.C., and Lam, E.W. (2003). FoxO3a transcriptional regulation of Bim controls apoptosis in paclitaxel-treated breast cancer cell lines. The Journal of biological chemistry 278, 49795-49805.

Page 182: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 166 -

Suri-Payer, E., and Fritzsching, B. (2006). Regulatory T cells in experimental autoimmune disease. Springer seminars in immunopathology 28, 3-16.

Takata, T., and Ishikawa, F. (2003). Human Sir2-related protein SIRT1 associates with the bHLH repressors HES1 and HEY2 and is involved in HES1- and HEY2-mediated transcriptional repression. Biochemical and biophysical research communications 301, 250-257.

Tan, P., Cady, B., Wanner, M., Worland, P., Cukor, B., Magi-Galluzzi, C., Lavin, P., Draetta, G., Pagano, M., and Loda, M. (1997). The cell cycle inhibitor p27 is an independent prognostic marker in small (T1a,b) invasive breast carcinomas. Cancer research 57, 1259-1263.

Tao, W., and Levine, A.J. (1999). P19(ARF) stabilizes p53 by blocking nucleo-cytoplasmic shuttling of Mdm2. Proceedings of the National Academy of Sciences of the United States of America 96, 6937-6941.

Tatar, M., Kopelman, A., Epstein, D., Tu, M.P., Yin, C.M., and Garofalo, R.S. (2001). A mutant Drosophila insulin receptor homolog that extends life-span and impairs neuroendocrine function. Science 292, 107-110.

Thisse, C., Neel, H., Thisse, B., Daujat, S., and Piette, J. (2000). The Mdm2 gene of zebrafish (Danio rerio): preferential expression during development of neural and muscular tissues, and absence of tumor formation after overexpression of its cDNA during early embryogenesis. Differentiation; research in biological diversity 66, 61-70.

Thomas, J.H. (1993). Chemosensory regulation of development in C. elegans. Bioessays 15, 791-797.

Thompson, P.R., Wang, D., Wang, L., Fulco, M., Pediconi, N., Zhang, D., An, W., Ge, Q., Roeder, R.G., Wong, J., Levrero, M., Sartorelli, V., Cotter, R.J., and Cole, P.A. (2004). Regulation of the p300 HAT domain via a novel activation loop. Nature structural & molecular biology 11, 308-315.

Tissenbaum, H.A., and Guarente, L. (2001). Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans. Nature 410, 227-230.

Tomaru, Y., Kondo, S., Suzuki, M., and Hayashizaki, Y. (2003). A comprehensive search for HNF-3alpha-regulated genes in mouse hepatoma cells by 60K cDNA microarray and chromatin immunoprecipitation/PCR analysis. Biochemical and biophysical research communications 310, 667-674.

Tothova, Z., Kollipara, R., Huntly, B.J., Lee, B.H., Castrillon, D.H., Cullen, D.E., McDowell, E.P., Lazo-Kallanian, S., Williams, I.R., Sears, C., Armstrong, S.A., Passegue, E., DePinho, R.A., and Gilliland, D.G. (2007). FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128, 325-339.

Page 183: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 167 -

Tran, H., Brunet, A., Grenier, J.M., Datta, S.R., Fornace, A.J., Jr., DiStefano, P.S., Chiang, L.W., and Greenberg, M.E. (2002). DNA repair pathway stimulated by the forkhead transcription factor FOXO3a through the Gadd45 protein. Science 296, 530-534.

Tsuchida, A., Yamauchi, T., Ito, Y., Hada, Y., Maki, T., Takekawa, S., Kamon, J., Kobayashi, M., Suzuki, R., Hara, K., Kubota, N., Terauchi, Y., Froguel, P., Nakae, J., Kasuga, M., Accili, D., Tobe, K., Ueki, K., Nagai, R., and Kadowaki, T. (2004). Insulin/Foxo1 pathway regulates expression levels of adiponectin receptors and adiponectin sensitivity. The Journal of biological chemistry 279, 30817-30822.

Uchida, C., Miwa, S., Kitagawa, K., Hattori, T., Isobe, T., Otani, S., Oda, T., Sugimura, H., Kamijo, T., Ookawa, K., Yasuda, H., and Kitagawa, M. (2005). Enhanced Mdm2 activity inhibits pRB function via ubiquitin-dependent degradation. The EMBO journal 24, 160-169.

Urbich, C., Knau, A., Fichtlscherer, S., Walter, D.H., Bruhl, T., Potente, M., Hofmann, W.K., de Vos, S., Zeiher, A.M., and Dimmeler, S. (2005). FOXO-dependent expression of the proapoptotic protein Bim: pivotal role for apoptosis signaling in endothelial progenitor cells. Faseb J 19, 974-976.

Valleix, S., Niel, F., Nedelec, B., Algros, M.P., Schwartz, C., Delbosc, B., Delpech, M., and Kantelip, B. (2006). Homozygous nonsense mutation in the FOXE3 gene as a cause of congenital primary aphakia in humans. American journal of human genetics 79, 358-364.

Van Der Heide, L.P., Hoekman, M.F., and Smidt, M.P. (2004). The ins and outs of FoxO shuttling: mechanisms of FoxO translocation and transcriptional regulation. The Biochemical journal 380, 297-309.

van der Heide, L.P., and Smidt, M.P. (2005). Regulation of FoxO activity by CBP/p300-mediated acetylation. Trends in biochemical sciences 30, 81-86.

van der Horst, A., de Vries-Smits, A.M., Brenkman, A.B., van Triest, M.H., van den Broek, N., Colland, F., Maurice, M.M., and Burgering, B.M. (2006). FOXO4 transcriptional activity is regulated by monoubiquitination and USP7/HAUSP. Nature cell biology 8, 1064-1073.

van der Horst, A., Tertoolen, L.G., de Vries-Smits, L.M., Frye, R.A., Medema, R.H., and Burgering, B.M. (2004). FOXO4 is acetylated upon peroxide stress and deacetylated by the longevity protein hSir2(SIRT1). The Journal of biological chemistry 279, 28873-28879.

Vaziri, H., Dessain, S.K., Ng Eaton, E., Imai, S.I., Frye, R.A., Pandita, T.K., Guarente, L., and Weinberg, R.A. (2001). hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell 107, 149-159.

Page 184: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 168 -

Voelter-Mahlknecht, S., and Mahlknecht, U. (2006). Cloning, chromosomal characterization and mapping of the NAD-dependent histone deacetylases gene sirtuin 1. International journal of molecular medicine 17, 59-67.

Voges, D., Zwickl, P., and Baumeister, W. (1999). The 26S proteasome: a molecular machine designed for controlled proteolysis. Annual review of biochemistry 68, 1015-1068.

Vousden, K.H., and Lu, X. (2002). Live or let die: the cell's response to p53. Nature reviews 2, 594-604.

Wang, C., Fu, M., Angeletti, R.H., Siconolfi-Baez, L., Reutens, A.T., Albanese, C., Lisanti, M.P., Katzenellenbogen, B.S., Kato, S., Hopp, T., Fuqua, S.A., Lopez, G.N., Kushner, P.J., and Pestell, R.G. (2001). Direct acetylation of the estrogen receptor alpha hinge region by p300 regulates transactivation and hormone sensitivity. The Journal of biological chemistry 276, 18375-18383.

Wang, M.C., Bohmann, D., and Jasper, H. (2005). JNK extends life span and limits growth by antagonizing cellular and organism-wide responses to insulin signaling. Cell 121, 115-125.

Wang, X.W., Zhan, Q., Coursen, J.D., Khan, M.A., Kontny, H.U., Yu, L., Hollander, M.C., O'Connor, P.M., Fornace, A.J., Jr., and Harris, C.C. (1999). GADD45 induction of a G2/M cell cycle checkpoint. Proceedings of the National Academy of Sciences of the United States of America 96, 3706-3711.

Weber, J.D., Taylor, L.J., Roussel, M.F., Sherr, C.J., and Bar-Sagi, D. (1999). Nucleolar Arf sequesters Mdm2 and activates p53. Nature cell biology 1, 20-26.

Wessells, R.J., Fitzgerald, E., Cypser, J.R., Tatar, M., and Bodmer, R. (2004). Insulin regulation of heart function in aging fruit flies. Nature genetics 36, 1275-1281.

Wilkinson, K.D. (1997). Regulation of ubiquitin-dependent processes by deubiquitinating enzymes. Faseb J 11, 1245-1256.

Wilkinson, K.D., Tashayev, V.L., O'Connor, L.B., Larsen, C.N., Kasperek, E., and Pickart, C.M. (1995). Metabolism of the polyubiquitin degradation signal: structure, mechanism, and role of isopeptidase T. Biochemistry 34, 14535-14546.

Wilm, B., James, R.G., Schultheiss, T.M., and Hogan, B.L. (2004). The forkhead genes, Foxc1 and Foxc2, regulate paraxial versus intermediate mesoderm cell fate. Developmental biology 271, 176-189.

Page 185: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 169 -

Wood, J.G., Rogina, B., Lavu, S., Howitz, K., Helfand, S.L., Tatar, M., and Sinclair, D. (2004). Sirtuin activators mimic caloric restriction and delay ageing in metazoans. Nature 430, 686-689.

Woods, Y.L., and Rena, G. (2002). Effect of multiple phosphorylation events on the transcription factors FKHR, FKHRL1 and AFX. Biochemical Society transactions 30, 391-397.

Wu, J., Shen, Z.Z., Lu, J.S., Jiang, M., Han, Q.X., Fontana, J.A., Barsky, S.H., and Shao, Z.M. (1999). Prognostic role of p27Kip1 and apoptosis in human breast cancer. British journal of cancer 79, 1572-1578.

Xiao, Z.X., Chen, J., Levine, A.J., Modjtahedi, N., Xing, J., Sellers, W.R., and Livingston, D.M. (1995). Interaction between the retinoblastoma protein and the oncoprotein MDM2. Nature 375, 694-698.

Yaciuk, P., and Moran, E. (1991). Analysis with specific polyclonal antiserum indicates that the E1A-associated 300-kDa product is a stable nuclear phosphoprotein that undergoes cell cycle phase-specific modification. Molecular and cellular biology 11, 5389-5397.

Yadav, N., Lee, J., Kim, J., Shen, J., Hu, M.C., Aldaz, C.M., and Bedford, M.T. (2003). Specific protein methylation defects and gene expression perturbations in coactivator-associated arginine methyltransferase 1-deficient mice. Proceedings of the National Academy of Sciences of the United States of America 100, 6464-6468.

Yamamoto, M., Meno, C., Sakai, Y., Shiratori, H., Mochida, K., Ikawa, Y., Saijoh, Y., and Hamada, H. (2001). The transcription factor FoxH1 (FAST) mediates Nodal signaling during anterior-posterior patterning and node formation in the mouse. Genes & development 15, 1242-1256.

Yamamura, Y., Lee, W.L., Inoue, K., Ida, H., and Ito, Y. (2006). RUNX3 cooperates with FoxO3a to induce apoptosis in gastric cancer cells. The Journal of biological chemistry 281, 5267-5276.

Yan, J., Xu, L., Crawford, G., Wang, Z., and Burgess, S.M. (2006). The forkhead transcription factor FoxI1 remains bound to condensed mitotic chromosomes and stably remodels chromatin structure. Molecular and cellular biology 26, 155-168.

Yang, Y., Hou, H., Haller, E.M., Nicosia, S.V., and Bai, W. (2005). Suppression of FOXO1 activity by FHL2 through SIRT1-mediated deacetylation. The EMBO journal 24, 1021-1032.

Yao, K.M., Sha, M., Lu, Z., and Wong, G.G. (1997). Molecular analysis of a novel winged helix protein, WIN. Expression pattern, DNA binding property, and alternative

Page 186: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 170 -

splicing within the DNA binding domain. The Journal of biological chemistry 272, 19827-19836.

Yao, T.P., Oh, S.P., Fuchs, M., Zhou, N.D., Ch'ng, L.E., Newsome, D., Bronson, R.T., Li, E., Livingston, D.M., and Eckner, R. (1998). Gene dosage-dependent embryonic development and proliferation defects in mice lacking the transcriptional integrator p300. Cell 93, 361-372.

Ye, H., Kelly, T.F., Samadani, U., Lim, L., Rubio, S., Overdier, D.G., Roebuck, K.A., and Costa, R.H. (1997). Hepatocyte nuclear factor 3/fork head homolog 11 is expressed in proliferating epithelial and mesenchymal cells of embryonic and adult tissues. Molecular and cellular biology 17, 1626-1641.

Yogosawa, S., Miyauchi, Y., Honda, R., Tanaka, H., and Yasuda, H. (2003). Mammalian Numb is a target protein of Mdm2, ubiquitin ligase. Biochemical and biophysical research communications 302, 869-872.

You, H., Jang, Y., You-Ten, A.I., Okada, H., Liepa, J., Wakeham, A., Zaugg, K., and Mak, T.W. (2004a). p53-dependent inhibition of FKHRL1 in response to DNA damage through protein kinase SGK1. Proceedings of the National Academy of Sciences of the United States of America 101, 14057-14062.

You, H., Pellegrini, M., Tsuchihara, K., Yamamoto, K., Hacker, G., Erlacher, M., Villunger, A., and Mak, T.W. (2006a). FOXO3a-dependent regulation of Puma in response to cytokine/growth factor withdrawal. The Journal of experimental medicine 203, 1657-1663.

You, H., Yamamoto, K., and Mak, T.W. (2006b). Regulation of transactivation-independent proapoptotic activity of p53 by FOXO3a. Proceedings of the National Academy of Sciences of the United States of America 103, 9051-9056.

You, Y., Huang, T., Richer, E.J., Schmidt, J.E., Zabner, J., Borok, Z., and Brody, S.L. (2004b). Role of f-box factor foxj1 in differentiation of ciliated airway epithelial cells. American journal of physiology 286, L650-657.

Yu, J.K., Holland, N.D., and Holland, L.Z. (2002). An amphioxus winged helix/forkhead gene, AmphiFoxD: insights into vertebrate neural crest evolution. Dev Dyn 225, 289-297.

Yu, Z.K., Geyer, R.K., and Maki, C.G. (2000). MDM2-dependent ubiquitination of nuclear and cytoplasmic P53. Oncogene 19, 5892-5897.

Yuan, Z.M., Huang, Y., Ishiko, T., Kharbanda, S., Weichselbaum, R., and Kufe, D. (1997). Regulation of DNA damage-induced apoptosis by the c-Abl tyrosine kinase. Proceedings of the National Academy of Sciences of the United States of America 94, 1437-1440.

Page 187: Regulation of FOXO stability and activity by MDM2 E3 ligase

- 171 -

Yuan, Z.M., Huang, Y., Whang, Y., Sawyers, C., Weichselbaum, R., Kharbanda, S., and Kufe, D. (1996). Role for c-Abl tyrosine kinase in growth arrest response to DNA damage. Nature 382, 272-274.

Zannini, M., Avantaggiato, V., Biffali, E., Arnone, M.I., Sato, K., Pischetola, M., Taylor, B.A., Phillips, S.J., Simeone, A., and Di Lauro, R. (1997). TTF-2, a new forkhead protein, shows a temporal expression in the developing thyroid which is consistent with a role in controlling the onset of differentiation. The EMBO journal 16, 3185-3197.

Zhang, Y., Wolf, G.W., Bhat, K., Jin, A., Allio, T., Burkhart, W.A., and Xiong, Y. (2003). Ribosomal protein L11 negatively regulates oncoprotein MDM2 and mediates a p53-dependent ribosomal-stress checkpoint pathway. Molecular and cellular biology 23, 8902-8912.

Zhang, Y., and Xiong, Y. (2001). Control of p53 ubiquitination and nuclear export by MDM2 and ARF. Cell Growth Differ 12, 175-186.

Zhao, Y.Y., Gao, X.P., Zhao, Y.D., Mirza, M.K., Frey, R.S., Kalinichenko, V.V., Wang, I.C., Costa, R.H., and Malik, A.B. (2006). Endothelial cell-restricted disruption of FoxM1 impairs endothelial repair following LPS-induced vascular injury. The Journal of clinical investigation 116, 2333-2343.

Zhou, B.P., Liao, Y., Xia, W., Zou, Y., Spohn, B., and Hung, M.C. (2001). HER-2/neu induces p53 ubiquitination via Akt-mediated MDM2 phosphorylation. Nature cell biology 3, 973-982.

Zhou, Y., Kato, H., Asanoma, K., Kondo, H., Arima, T., Kato, K., Matsuda, T., and Wake, N. (2002). Identification of FOXC1 as a TGF-beta1 responsive gene and its involvement in negative regulation of cell growth. Genomics 80, 465-472.

Zhu, W., Bijur, G.N., Styles, N.A., and Li, X. (2004). Regulation of FOXO3a by brain-derived neurotrophic factor in differentiated human SH-SY5Y neuroblastoma cells. Brain research 126, 45-56.

Zhu, Y., Carroll, M., Papa, F.R., Hochstrasser, M., and D'Andrea, A.D. (1996). DUB-1, a deubiquitinating enzyme with growth-suppressing activity. Proceedings of the National Academy of Sciences of the United States of America 93, 3275-3279.

Page 188: Regulation of FOXO stability and activity by MDM2 E3 ligase

About the author

Wei Fu received her Bachelor’s degree in Biology from Shaanxi Normal

University in China in 1994 and her Master’s degree in Medical Microbiology and

Immunology from Xi’an Medical University in 1997. During her graduate year she worked

in the laboratory of Dr. Yonglie Chu and completed her thesis was on the molecular

biology of the human papillomavirus in cervical cancer. Wei worked also as a research

assistant in the Molecular Center of the First Clinical Hospital of Xi’an Jiaotong

Univeristy where she carried out both basic and clinical study. Wei started her Graduate

School studies in the fall of 2001 at University of South Florida, Tampa, Florida, and

persued her Ph.D thesis work under the tutelage of Dr. Wenlong Bai studying the

regulation of FOXO in stress response and tumorigenesis. She successfully defended

her doctoral dissertation in November 2007 at the University of South Florida. Wei is a

member of the Amercian Cancer Association and the American Heart Association.