characterization of the e3 ubiquitin ligase pirh2 · the p53 gene encodes a transcription factor...

133
Characterization of the E3 Ubiquitin Ligase Pirh2 by Elizabeth Tai A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Graduate Department of Medical Biophysics University of Toronto © Copyright by Elizabeth Tai (2010)

Upload: others

Post on 07-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

Characterization of the E3 Ubiquitin Ligase Pirh2

by

Elizabeth Tai

A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy

Graduate Department of Medical Biophysics University of Toronto

© Copyright by Elizabeth Tai (2010)

Page 2: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

II

Characterization of the E3 Ubiquitin Ligase Pirh2

Elizabeth Tai

Doctor of Philosophy

Department of Medical Biophysics University of Toronto

2010

Abstract

The p53 tumour suppressor gene is inactivated by mutation in over 50% of all human

cancers. The p53 protein is activated and stabilized through several post-translational

modifications in response to various stresses and promotes cell cycle arrest and apoptosis.

Thus, regulation of p53 is critical for normal cellular function. Pirh2 is a p53-regulated

gene recently identified in our laboratory which encodes an E3 RING-finger ubiquitin

ligase that binds to p53 and negatively regulates p53 by targeting it for ubiquitin-

mediated proteolysis. Pirh2 is similar to another well-characterized E3 RING finger

ubiquitin ligase, Mdm2, which also participates in a similar negative feedback loop with

p53. At least seven E3 ubiquitin ligases are known to target p53 for degradation and the

reason for this functional redundancy is unclear. The purpose of this study is to

characterize Pirh2 activity.

This study has two aims the first is to identify additional interacting proteins for

Pirh2, and the second is to delineate Pirh2 regulation of p53. Using several tandem

affinity purification strategies and a GST-pull down approach, we have identified PKC

as a candidate interacting protein.

Page 3: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

The second aim is to further characterize Pirh2 regulation of p53. Splenocytes and

thymocytes from Pirh2-/- mice demonstrate a subtle increase in total p53 levels after

irradiation when compared to wild-type controls. Phosphoserine 15 p53 levels are

significantly higher in splenocytes and thymocytes from Pirh2 -/- mice relative to wild-

type counterparts. Cells stably transfected with Pirh2 have decreased levels of

phosphoserine 15 p53 and decreased induction of p21 relative to vector control and

Mdm2 expressing cells.

The stability of the p53 protein is primarily regulated through ubiquitin mediated

proteolysis, and there are multiple ubiquitin ligases targeting p53 for degradation. Here

we are able to address the question of functional redundancy by indicating that Pirh2 can

target serine 15 phosphorylated p53 which is reported to not be regulated by Mdm2.

III

Page 4: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

Acknowledgements

This thesis arose from many years of work previously completed in the

Benchimol lab. During my time working on the project, I was fortunate to receive

several contributions from many individuals; without their assistance, I would not have

been able to complete this thesis. I would now like to take the time to humbly

acknowledge those individuals.

I would like to thank my supervisor Dr. Sam Benchimol for his advice, guidance

and patience in helping me to develop scientific skills; it has been a fantastic learning

experience. Above all, he has instilled in me a strong sense of integrity, and enthusiasm

in the pursuit of science.

I would also like to acknowledge my committee members Dr. Cheryl Arrowsmith,

Dr. Razqallah Hakem, and Dr. Linda Penn. Their advice and support have been

instrumental in the development of my project and in my development as a researcher.

I would also like to thank the past and present members of the Benchimol lab for

useful and critical discussion. I would like to thank Weili Ma for her unwavering support

and for sharing her extensive technical expertise. I would also like to acknowledge Dr.

Keith Wheaton, Dr. Wissam Assaily, Dr. Christina Berube and Dr. Jenny Ho for

encouragement and scientific discussion. They are my scientific role models, and set a

high standard for which I strived to achieve.

I would also like to thank Dr. Yi Sheng, Shili Duan, and Dr. Anne Hakem for

sharing their technical expertise.

I would like to thank my mother, father, brother and friends for their steadfast

support. Their encouragement has been instrumental to the completion of my thesis.

IV

Page 5: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

Table of Contents

CHAPTER 1: Introduction...............................................................................................1 The p53 Network .................................................................................................................2

p53 as a tumour suppressor......................................................................................2 p53-dependent cell cycle arrest................................................................................3 p53-dependent apoptosis..........................................................................................5 Choice of response...................................................................................................7 Mouse models of p53 tumour suppression ..............................................................8 Regulation of p53 function ....................................................................................11 Phosphorylation of p53 at serine 15.......................................................................16

Ubiquitination ....................................................................................................................17 Ubiquitin ................................................................................................................17 The ubiquitination mechanism...............................................................................18 Ubiquitination and signalling.................................................................................24 Ubiquitination and protein degradation .................................................................26 Deubiquitination ....................................................................................................28

Ubiquitination and regulation of p53.................................................................................30 Pirh2.......................................................................................................................35

Thesis Hypothesis ..............................................................................................................37 CHAPTER 2: Identification of Pirh2 Interacting Proteins .........................................38

Abstract ..................................................................................................................39 Introduction............................................................................................................39 Experimental Procedures .......................................................................................41

Cell Culture................................................................................................41 Plasmids .....................................................................................................41 Generation of Clones Expressing pCMV TAP hPirh2 ..............................42 Generation of Clones Expressing pBabe neo Pirh2-SPA ..........................42 Generation of Clones Expressing pBabe puro Pirh2-HF...........................42 Ubiquitination Assay .................................................................................43 Coimmunoprecipitation .............................................................................43 Western blots .............................................................................................44 TAP Purification ........................................................................................44 SPA Purification ........................................................................................45 His-Flag Purification..................................................................................46 GST-pull down...........................................................................................47

Results....................................................................................................................47 Generation and characterization of TAP-hPirh2 clones ............................47 Purification of TAP and TAP-hPirh2 ........................................................51 Generation of clones and purification of Pirh2-SPA .................................51 Generation of clones and purification of Pirh2-HF ...................................54 Identification of Pirh2 interactors using GST-pull down ..........................56

Discussion..............................................................................................................57

V

Page 6: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

CHAPTER 3: Pirh2 targets phosphoserine 15 p53 for ubiquitination......................61

Abstract ..................................................................................................................62 Introduction............................................................................................................62 Experimental Procedure.........................................................................................64

Cell Culture................................................................................................64 Splenocyte and Thymocyte Culture Preparation .......................................65 Preparation of MCF-7 p53shRNA Cells....................................................65 Western Blotting ........................................................................................66 Immunohistochemistry ..............................................................................66 Coimmunoprecipitation .............................................................................66 Ubiquitination Assay .................................................................................67 Flow Cytometry .........................................................................................67

Results....................................................................................................................68 Pirh2-/- Tissues Show Elevated Levels of p53 After DNA Damage.........68 Pirh2-/- Tissues Show Elevated Levels of Phosphoserine 15 p53

After DNA Damage .......................................................................68 Apoptotic Rate is Increased in Pirh2-/- Mice ............................................70 Pirh2 Can Regulate p53 Mutants for Serine 15 .........................................73 Cells Overexpressing Pirh2 Decrease Levels of Phosphoserine 15 p53

and p21, But Does Not Affect Cell Cycle Arrest...........................73 Pirh2 Coimmunoprecipitates with Phosphoserine 15 p53 and promotes

Phosphoserine 15 p53 Ubiquitination............................................78 Discussion..........................................................................................................................80 CHAPTER 4: Summary and Future Directions ..........................................................84

I. Summary of Findings: Identification of Pirh2 Interactors..................................85 Future Directions: Identification of Pirh2 Interactors............................................85

Identification of Pirh2 Cofactors and Substrates .......................................85 Characterization of the Interactors.............................................................90 Identification of the Pirh2 Degron .............................................................92

II. Summary of Findings: Pirh2 Regulation of Phosphoserine 15 p53 ..................93 Future Directions ...................................................................................................96

Dissecting the Functional Redundancy of p53 Ubiquitin Ligases.............96 Non-Proteolytic Ubiquitination of p53 ......................................................98 Pirh2-Mediated Ubiquitination of p53.......................................................99

III. Pirh2 and Cancer............................................................................................101 REFERENCES...............................................................................................................105

VI

Page 7: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

List of Illustrations Figure 1.1 Post-Translational Modifications of p53 .........................................................12 Figure 1.2 Ubiquitin Linkage Forms ................................................................................19 Figure 1.3 Ubiquitination Reaction Mechanism...............................................................21 Figure 1.4 Ubiquitination in the NF-κB Pathway.............................................................25 Figure 2.1 Generation of TAP-hPirh2 clones in H1299 cells ...........................................48 Figure 2.2 Functional Assays for TAP-hPirh2 Activity ...................................................50 Figure 2.3 Purification of TAP and TAP-hPirh2 ..............................................................52 Figure 2.4 Generation and Purification of Pirh2-SPA Clones..........................................53 Figure 2.5 Generation and Purification of Pirh2-HF Clones ............................................55 Figure 2.6 GST-pull Down to Identify Novel Pirh2 Interacting Proteins.........................58 Figure 3.1 Elevated p53 Levels in Pirh2-/- Tissues..........................................................69 Figure 3.2 Elevated Levels of Phosphoserine 15 p53 in Pirh2-/- Mice ............................71 Figure 3.3 Elevated Levels of Apoptosis in Pirh2-/- Mice ...............................................72 Figure 3.4 Mdm2 and Pirh2 Can Regulate p53 Serine 15 Mutant....................................74 Figure 3.5 Decreased Levels of Phosphoserine 15 p53 and p21

in Pirh2 Overexpressing Cells ...................................................................76 Figure 3.6 Overexpression of Pirh2 Does Not Decrease S-phase

After DNA Damage ...................................................................................77 Figure 3.7 Pirh2 Interacts with and Ubiquitinates Phosphoserine 15 p53........................79 Figure 4.1 Model for Direct Pirh2 Regulation of p53 ......................................................95

VII

Page 8: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

List of Abbreviations

ADP – Adenosine Diphosphate ATP – Adenosine Triphosphate BrdU – Bromodeoxyuridine BrdU-FITC – Bromodeoxyuridine – Fluorescein BSA – Bovine Serum Albumin CBP – Calmodulin Binding Peptide cIAP – Cellular Inhibitor of Apoptosis cICAT – cleavable Isotope-Coded Affinity Tags DNA – Deoxyribose Nucleic Acid DTT – Dithiothreitol EDTA – Ethylenediaminetetraacetic acid EGFR – Epidermal Growth Factor Receptor EGTA – Ethylene glycol tetraacetic acid EMSA – Electrophoretic Mobility Shift Assay ERAD – Endoplasmic Reticulum Associated Protein Degradation FACS – Fluorescence Activated Cell Sorting FCS – Fetal Calf Serum GST – Glutathione – S Transferase H&E – Hematoxylin & Eosin HA – Hemaglutinnin HECT – Homologous to E6 – Associated Protein Carboxy-Terminus HEPES – 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid HPV – Human Papilloma Virus IgG – Immunoglobulin G IgH – Immunoglobulin Heavy chain IHC – Immunohistochemistry IR – Ionizing Radiation ITRAQ – Isobaric Tags for Relative and Absolute Quantification LC – MS/MS – Liquid Chromatography – Mass Spectrometry/ Mass Spectrometry LFS – Li-Fraumeni Syndrome MALDI - MS/MS – Matrix Laser Desorption Ionization – Mass Spectrometry/Mass

Spectrometry MEFs – Murine Embryonic Fibroblasts MJD – Machado-Joseph Disease mRNA – messenger Ribonucleic Acid NAC – N-Acetyl Cysteine NP-40 – Nonyl phenoxylpolyethoxylethanol OTU – Otubain proteases PAGE – Polyacrylamide Gel Electrophresis PBS – Phosphate Buffered Saline PCR – Polymerase Chain Reaction PKC – Protein Kinase C RING – Really Interesting New Gene RNAi – Ribonucleic Acid interference

VIII

Page 9: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

IX

RPMI – Roswell Park Memorial Institute SDS – Sodium Dodecyl Sulfate SNP – Single Nucleotide Polymorphism SPA – Sequential Peptide Affinity TAP – Tandem Affinity Purification TEV – Tobacco Etch Virus Tris-HCl – Tris(hydroxymethyl)aminomethane – Hydrogen Chloride ts – temperature sensitive TUNEL – Terminal deoxynucleotidyl transferase dUTP nick end labeling UCH – Ubiquitin C-terminal hydrolase USP – Ubiquitin Specific Protease UV – Ultraviolet Radiation

Page 10: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

CHAPTER 1

Introduction

1

Page 11: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

2

The p53 Network

p53 as a Tumour Suppressor

p53 was initially characterized as a protein that interacts with the large T antigen

from the SV40 virus (Lane and Crawford, 1979; Linzer and Levine, 1979). Early

research suggested that p53 was involved in promoting cell division as p53 could

cooperate with oncogenic ras to transform rat embryo fibroblasts (Eliyahu et al., 1984;

Jenkins et al., 1984; Parada et al., 1984). However, it was later determined that wild-type

p53 actually suppresses cell transformation, and the previously characterized oncogenic

properties of p53 were due to mutations in the gene (Finlay et al., 1989; Hinds et al.,

1989). In fact, rearrangements and point mutations in the p53 gene were observed in the

majority of murine erythroleukemic cell lines (Ben David et al., 1988; Mowat et al., 1985;

Munroe et al., 1990). In vivo, the p53 gene is mutated in over 50% of all human cancers,

and is one of the most common mutations in cancer (Vousden and Lu, 2002). Germline

mutations of p53 have been found, and humans born with one mutated copy of the p53

gene develop Li-Fraumeni syndrome (LFS). Patients with LFS have an increased number

and earlier onset of cancers (Malkin et al., 1990). These results suggested that the wild-

type p53 protein functions as a tumour suppressor, and that mutation of the protein

promotes oncogenesis.

The p53 gene encodes a transcription factor that regulates gene expression in

response to various stresses such as DNA damage and oncogene activation. The p53

tumour suppressor protein contains an N-terminal activation domain, a central DNA

binding domain, and a C-terminal regulatory domain. The N-terminal domain of p53 can

bind various transcriptional activators such as histone acetylates p300 and CBP (Riley et

Page 12: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

3

al., 2008). The C-terminus contains the regulatory domain that includes the

oligomerization domain as p53 binds to DNA as a homotetramer in the form of a dimer

of dimers (Davison et al., 2001). The p53 consensus sequence consists of at least two

repeats of the consensus 5’- PuPuPuC(A/T)(T/A)PyPyPy - 3’ with 0-13 base pairs of

non-specific sequence in between (el-Deiry et al., 1992); the consensus binding sequence

of p53 supports the model of p53 binding to DNA as tetramer. p53 induces the

expression of genes involved in cell cycle arrest and apoptosis in response to stimuli

(Levine, 1997). The central DNA binding domain of p53 is frequently mutated in

cancers, and suggests that the ability of p53 to transactivate gene expression is critical for

tumour suppression (Vousden and Lu, 2002).

p53-dependent cell cycle arrest

p53 promotes cell cycle arrest through the transcriptional activation of numerous

target genes including p21WAF1, 14-3-3σ, REPRIMO, BTG2, GADD45 (el-Deiry et al.,

1993; Hermeking et al., 1997; Ohki et al., 2000; Rouault et al., 1996; Wang et al., 1999).

p53 induces cell cycle arrest in G1 using p21 and BTG2; GADD45, REPRIMO, and 14-

3-3σ control the G2/M transition (Chan et al., 2000; el-Deiry et al., 1993; Guardavaccaro

et al., 2000; Harper et al., 1993; Wang et al., 1999).

A more recent discovery is that p53 can also modulate cell cycle arrest through

induction of microRNAs (miRNA). Activation of p53 causes expression of the miR34

family of miRNAs that are involved in downregulating genes that promote cell cycle

entry such as CDK4 and MET. Ectopic expression of miR34 causes cell cycle arrest in

Page 13: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

4

both G1 and G2 phase, and a decrease in S-phase cells in both tumour-derived cell lines

and normal cell lines (He et al., 2007).

Transcriptional repression by p53 is also an important part of the growth arrest

pathway. p53 mediates transcriptional repression of c-myc by binding to the c-myc

promoter and recruiting histone deacetylases. Repression of c-myc by p53 increases G1

arrest (Ho et al., 2005). These results indicate that p53 is able to cause cell cycle arrest

by both upregulating genes that promote arrest, and downregulating genes that promote

entry into the cell cycle.

Loss of p53 in cells causes inappropriate entry into S phase after treatment with

metabolic inhibitors that normally arrest cells in G1; these cells have a higher incidence

of genomic instability and gene amplification (Livingstone et al., 1992; Yin et al., 1992).

Cell cycle arrest is thought to aid in p53 mediated tumour suppression since it allows the

cell time to repair lesions in response to DNA damage.

Cell cycle arrest is also involved in senescence, a permanent state of G1 cell cycle

arrest. In experiments, replicative senescence is studied by taking normal mature cells

lacking telomerase activity, usually human fibroblasts and culturing them in vitro to

undergo multiple cell divisions until the telomeres at the end of the chromosomes are

shortened. These shortened telomeres trigger the activation of p53, primarily through the

ATM/ATR pathway. It is thought that shortened telomeres mimic double strand breaks

(Herbig et al., 2004). The p53 target gene p21 is elevated in replicative senescence, but it

is not the only p53 target that causes replicative senescence. Further studies are being

performed to determine which mediators of p53-dependent cell cycle arrest are important

for replicative senescence (Artandi and Attardi, 2005; Vaziri et al., 1999). Cell cycle

Page 14: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

5

arrest is also involved in p53-dependent tumour suppression as rat embryonic fibroblasts

expressing oncogenic ras undergo p53-dependent oncogene induced senescence;

senescence is a physiologically relevant pathway of p53-dependent tumour suppression in

murine models of sarcoma and hepatocellular carcinoma (Serrano et al., 1997; Ventura et

al., 2007; Xue et al., 2007). Similar to replicative senescence, the p53-dependent cell

cycle target genes involved in oncogene induced senescence are unknown, and research

into the field is ongoing.

p53-dependent apoptosis

The apoptotic arm of p53 tumour suppression eliminates cells with irreparable

DNA damage or cells exhibiting unrestrained growth as a result of oncogene activation

(Vousden and Lu, 2002). Various pro-apoptotic genes are induced by p53 including

PUMA, Noxa, Bax, p53AIP1 and PIDD (Lin et al., 2000; Nakano and Vousden, 2001;

Oda et al., 2000a; Oda et al., 2000b; Yin et al., 1997). PUMA, Noxa, and Bax are located

at the mitochondria, and aid in permeabilization of the outer membrane to induce

cytochrome c release causing activation of caspase-9 resulting in apoptosis (Fridman and

Lowe, 2003). Deletion of Apaf-1 or caspase-9 in cells results in protection from p53-

dependent apoptosis; in fact, loss of either Apaf-1 or caspase-9 can substitute for loss of

p53 to prevent c-myc-induced apoptosis, suggesting that this is the main pathway for

p53-dependent apoptosis (Soengas et al., 1999)

More recently, it has been determined that p53-dependent apoptosis can also

occur in a transcription-independent manner. The p53 protein has been found to be

localized to the mitochondria, and can directly activate the pro-apoptotic protein Bax to

Page 15: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

6

induce cell death (Chipuk et al., 2004). p53 has also been found to bind to Bcl-XL

through its DNA binding domain, and mitochondrial p53 can result in decreased viability

(Mihara et al., 2003). The model for apoptosis induced by mitochondrial p53 states that

p53 translocates to the mitochondria upon DNA damage where it is sequestered by Bcl-

XL. Concurrently, nuclear p53 induces the expression of PUMA which then binds Bcl-

XL and releases p53 to bind Bax and induce apoptosis (Chipuk et al., 2005). A second

model of how p53 can directly induce apoptosis at the mitochondria claims that

mitochondrial p53 can bind and activate Bak by causing Bak oligomerization which

results in cytochrome c release and apoptosis (Leu et al., 2004). These models of how

p53 can directly induce apoptosis at the mitochondria need to be resolved. More

importantly, how the transcription-dependent arms and the transcription-independent

arms of p53 cooperate to cause cell death is a key question that remains unanswered.

p53-dependent apoptosis is also an important component of p53-dependent

tumour suppression. Mice that overexpress c-myc from the IgH promoter in p53+/- B-

cells develop B-cell lymphoma, and the remaining wild-type allele of p53 is mutated or

lost. However, the wild-type allele of p53 can be retained by overexpressing Bcl-2 or a

dominant negative caspase-9; these are both methods to inhibit mitochondrial apoptosis

downstream of p53 (Schmitt et al., 2002). Furthermore, a knock-in mouse that contains a

p53 construct that has deleted the proline rich domain has reduced capacity for inducing

p53-dependent cell cycle arrest but retains the ability to induce p53-dependent apoptosis

is still protected from spontaneous tumour formation (Toledo et al., 2006). The

previously described results suggest that the apoptotic arm of p53 is critical for mediating

tumour suppression.

Page 16: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

7

Choice of Response

The ability of p53 to promote both cell cycle arrest and apoptosis and how p53

decides between these two cellular responses is one of the key questions in the field. This

question has been extensively addressed by many studies that have outlined several key

determinants that influence this choice. These factors include the intensity of stress, the

efficiency of DNA repair, extracellular survival factors, level of p53 expression and

activating, oncogenic composition of the cell, intracellular death/survival pathways and

selective transactivation/repression of p53-target genes in different cell types (Bouvard et

al., 2000; Fei et al., 2002; Johnson et al., 1993; Lin and Benchimol, 1995; Lin et al., 2002;

Weber and Zambetti, 2003; Wu et al., 2005). A classic example uses fibroblasts as a

model; normal fibroblasts undergo p53-dependent cell cycle arrest in response to DNA

damage. However, fibroblasts that have been transformed by an activated oncogene,

such as adenovirus E1A protein, cell cycle proteins E2F-1 or myc, undergo p53-mediated

apoptosis. Cells ectopically expressing pro-survival proteins Bcl-2 or Bcl-XL are

protected from p53-dependent apoptosis, and constitutively active PI3’K and PKB/Akt

are able to delay p53-induced apoptosis (Chiou et al., 1994; Levine, 1997; Lin et al., 2002;

Nahle et al., 2002; Sabbatini and McCormick, 1999; Schott et al., 1995). The p53-target

gene Slug is p53-responsive and is expressed in hematopoetic cells. Slug binds to the

promoter of the pro-apoptotic p53 target gene PUMA to represses p53-dependent

transactivation. This results in a preference for cell cycle arrest over apoptosis (Wu et al.,

2005). The cellular outcome of the p53 response may also reflect differences in the

affinity of various promoters for p53, such that some are responsive only to high levels of

Page 17: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

8

p53 or to certain modified forms of p53 (Resnick-Silverman et al., 1998; Sionov and

Haupt, 1999). p53 promoter selectivity leading to cell cycle arrest or apoptosis can also

be regulated by protein-protein interactions between p53 and other proteins including

ASPP, JMY, WT1, BRCA1, Hzf, p63 and p73 (Das et al., 2007; Flores et al., 2002;

MacLachlan et al., 2002; Maheswaran et al., 1995; Samuels-Lev et al., 2001; Shikama et

al., 1999).

In addition to cell cycle arrest and apoptosis as mediators of p53-dependent

tumour suppression, the antioxidant function of p53 may also play a crucial role. The

tumour prone phenotype observed in p53 knock out mice is abrogated by maintaining the

mice on an N-acetylcysteine (NAC; antioxidant) diet; p53 knockout mice treated with

NAC do not develop the characteristic thymic lymphomas observed in p53-/- mice

(Sablina et al., 2005). The p53 tumour suppressor gene also induces the expression of

antioxidant genes glutathione peroxidase 1, mitochondrial superoxide dismutase 2 and

two sestrins (Sablina et al., 2005). These results suggest that tumour prevention by p53

involves the removal of reactive oxygen species; it remains to be determined whether the

induction of antioxidant genes by p53 contributes to tumour suppression in an oncogenic

environment, or whether the involvement of p53 in dealing with reactive oxygen species

is more prophylactic.

Mouse models of p53 tumour suppression

In animal models, mice that have deleted both copies of the p53 gene are

extremely tumour prone, and die within 6 months of birth as a direct result of developing

cancer. Knockout p53 mice can develop a wide spectrum of tumours, but most

Page 18: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

9

commonly develop thymic lymphomas and sarcomas (Donehower et al., 1992). The p53

tumour suppressor gene is rarely deleted in cancers and LFS syndrome; more commonly,

a point mutation in the p53 gene is found. As a result, new models for LFS were

developed in mice where the p53 protein with a common point mutation (273H) was

knocked in. These mice are similar to the p53 knockout mice in that they are tumour

prone. However, p53 273H has a gain of function property since tumours in the mice are

also metastatic in contrast to tumours from the p53 knockout mouse (Lang et al., 2004;

Olive et al., 2004). These observations indicate that loss of p53 function contributes to

the development of malignancy, and normal p53 function is critical for tumour

suppression.

Although loss of p53 activity is one of the most common changes in

tumourigenesis, it is unclear whether loss of p53 simply facilitates the genetic changes

such as increased genetic instability, loss of growth-arresting signals and inappropriate

cell survival that contribute to tumour development, or whether tumour growth is

dependent on keeping the p53 pathway turned off permanently (Kastan, 2007). Three

studies addressed these questions independently. The Evans laboratory generated a

knock-in mouse where p53 is fused to the estrogen receptor, and p53 activity can be

turned on by the addition of tamoxifen. In the Eµ-myc model of lymphomagenesis,

restoration of p53 activity through the addition of tamoxifen increases survival of the

mouse. In addition, the tumours eventually developed resistance to p53 suggesting that

the pathway must be inactivated to ensure tumour proliferation (Martins et al., 2006).

The Jacks laboratory addressed this question using a mouse model where a stop cassette

was placed upstream of the p53 gene, but could be removed by a tamoxifen-inducible

Page 19: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

10

Cre-recombinase. In the absence of Cre, the mice are functionally p53-deficient and

develop spontaneous or irradiation-induced lymphomas or sarcomas. Restoration of p53

resulted in apoptosis in the lymphomas and senescence in the sarcomas (Ventura et al.,

2007). The Lowe laboratory addressed how p53 affects tumorigenesis by using purified

embryonic liver cells transformed by oncogenic Hras and a short hairpin against p53 that

is repressed by the addition of doxycycline. When these oncogenic cells are injected into

immunodeficient mice, hepatocellular carcinomas quickly develop. However, when p53

expression is restored through doxycycline treatment, the tumour shows signs of

regression after four days of doxycycline treatment, and becomes undetectable after

twelve days of treatment. In this model, the hepatocellular carcinomas undergo Hras

oncogene-induced senescence (Xue et al., 2007). These results suggest that restoration of

wild-type p53 in tumours that are deficient for p53 could be therapeutically advantageous.

Gene therapy approaches that directly introduce wild-type p53 genes into tumours are

already in clinical trials (Roth, 2006). Additional in vivo experiments support the

hypothesis that p53 restoration in tumours would be therapeutically effective. In the

Evans mouse model where p53 activity is directly induced by the addition of tamoxifen,

mice were exposed to whole body irradiation to cause radiation induced lymphoma.

Restoration of p53 shortly after irradiation had no effect on tumour suppression, but did

cause an acute radiation response where the spleen, thymus, and small intestine

underwent massive apoptosis. However, restoration of p53 after the acute radiation

response had subsided abrogated tumour formation. These observations suggest that p53

tumour suppression occurs as a result of the activation of oncogenes (Christophorou et al.,

2006). These observations also support the therapeutic efficacy for restoring p53 in

Page 20: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

11

tumours; the therapeutic effectiveness may be maximized by further activating p53 with

DNA damaging agents after it is introduced into the oncogenic environment (Kastan,

2007).

Regulation of p53 function

p53 is primarily regulated at the post-translational level and is very complex;

after stress, the protein is stabilized and accumulates in the cell. Regulation of the protein

can occur by a multitude of post-translational modifications such as phosphorylation,

acetylation, ubiquitination, sumoylation, neddylation, methylation, ADP-ribosylation, and

glycosylation (Figure 1.1) (Kruse and Gu, 2009a; Lu, 2005; Toledo and Wahl, 2006). In

addition, p53 function can also be regulated by cofactors that bind to p53. For example,

binding of 14-3-3σ to the C-terminus of p53 can increase the stability and enhance p53

transcriptional activity, and binding of BRCA1 to p53 also increases p53 activity (Yang

et al., 2003; Zhang et al., 1998). The ASPP family of protein can bind to p53 and

preferentially stimulates p53 induced apoptosis by directing p53 transcriptional activation

towards pro-apoptotic promoters (Samuels-Lev et al., 2001). The p53 target gene Hzf

controls p53 transcriptional activity by binding to the p53 DNA binding domain and

redirects p53 to cell cycle arrest promoters (Das et al., 2007; Wu et al., 2005). Inhibitory

proteins can also bind p53, as both MdmX and nucleophosmin (NPM/B23) can bind to

p53 in the N-terminus to inhibit transcriptional activation (Danovi et al., 2004; Maiguel et

al., 2004). MdmX is a critical negative regulator of p53 since MdmX null mice are

embryonic lethal at day 9.5, but embryonic lethality is completely rescued by loss of p53.

The MdmX and p53 double null mice are completely viable, but are cancer prone similar

Page 21: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

Figure 1.1 Post-translational Modifications of p53.

p53 is heavily regulated by post-translational modifications including phosphorylation(P), ubiquitination (Ub), acetylation (Ac), methylation (Me), sumoylation (S), neddylation (N8), glycosylation (O-Glc) and ribosylation (ADP). This figure shows the major sites of these modifications. (adapted from Kruse and Gu, 2009)

Me

N8

Ac Ub

P ADP

O-Glc

S

Methylation

Sumoylation

Acetylation

Phosphorylation

Ubiquitination

ADP-ribosylation

Glycosylation

Neddylation

1 92 100 300 307 355 356 393

Transactivation/Proline Rich

DomainDNA binding domain

Tetramerizationdomain

C-Terminaldomain

P P P

Ac Ac

AcADP ADP ADP

S149 S150 S155 E258 D259 E271 K292

Ac Ac Ac

K120 K164

Ub Ub

UbUb

N8 N8

UbP P P P

K305 S315 K319 K320 K321 K351 K357 S376 S378 S392

P P P P P PP P P

S6 S9 S15 T18 S20 S33 S36 S37 S46 T81

P

P

T55

K370

K372

K373

K386

K382

K381

Me

N8Ac

Ub Me

N8Ac

Ub Me

Ac

Ub

Ac

Ub

N8Ac

Ub

Ac

Ub

S

O-Glc

12

Page 22: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

13

to the phenotype of the p53 knock out mice (Parant et al., 2001). Another example of an

inhibitor protein is Parc that binds to p53 and relocates nuclear p53 to the cytoplasm.

Ablation of Parc using RNAi relocalizes p53 to the nucleus and induces cells to undergo

apoptosis (Nikolaev et al., 2003). Protein cofactors play an important role in regulating

p53 stability and function; the exact mechanism of when these protein cofactors come

into play remains a mystery.

Regulation of p53 through post-translational modifications is vitally important,

and extremely complex. Modifications on p53 can have both inhibitory and activating

effects. Acetylation of p53 occurs usually on C-terminal lysines and has been shown to

be an activating modification. Acetylation of p53 promotes apoptosis and cell cycle

arrest, and a mutant p53 that has lost all eight acetylation sites has lost the ability to

induce apoptosis and growth arrest (Tang et al., 2008). p53 can be acetylated on multiple

lysines throughout the protein, but most are located in the C-terminal regulatory region.

Lysines 120, 164, 370, 372, 373, 381, 382, 386 can all be acetylated by several proteins

including Tip60/hMOF, p300/CBP and PCAF; these lysines can also be ubiquitinated,

and it is thought that acetylation may act to block ubiquitin mediated proteolysis of p53

(Li et al., 2002b; Nakamura et al., 2000; Tang et al., 2006; Tang et al., 2008).

Acetylation can be reversed by HDAC or hSIRT1, resulting in deactivation and

destabilization of the p53 protein (Ito et al., 2002; Vaziri et al., 2001). Methylation can

occur on multiple residues and can have both an inhibitory and activating effect.

Methylation of lysine 372 by Set9 can activate p53-dependent apoptosis (Chuikov et al.,

2004). p53 activity can also be inhibited by methylation of lysine 370 by SMYD2, which

causes p53 destabilization. This methylation specific inhibitory effect can be reversed

Page 23: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

14

through lysine demethylation by LSD1 (Huang et al., 2007; Toledo and Wahl, 2006).

Sumoylation competes with acetylation and ubiquitination at lysine 386 of p53; the

addition of SUMO-1 to lysine 386 of p53 induces transcriptional activation (Rodriguez et

al., 1999). p53 transcriptional activity can also be inhibited through Mdm2-mediated

neddylation (Xirodimas et al., 2004). Ubiquitination of p53 is complex, but can have

both activating and inhibitory functions. Monoubiquitination of cytoplasmic p53 targets

the protein to the mitochondria where it can be deubiquitinated and induce apoptosis

(Marchenko et al., 2007). However, the p53 protein has a very short half-life, and under

unstressed conditions, p53 stability is regulated primarily through ubiquitin-mediated

proteolysis (Brooks and Gu, 2003).

Multiple kinases target p53 for phosphorylation on several serines and threonines

in the N-terminal transactivation domain, central DNA binding domain and C-terminal

regulatory domain. Phosphorylation of p53 is thought to be an activating modification.

Phosphorylation of serine 37 after DNA damage promotes the transcriptional activity

since mutation of this residue to alanine reduces p53-dependent transcription; in addition,

phosphorylation of serine 37 can be reversed by protein phosphatase 2A (Dohoney et al.,

2004). Also, phosphorylation of p53 on serine 46 has been shown to induce expression

of the pro-apoptotic protein p53-AIP (Oda et al., 2000b).

To determine the role of phosphorylation on p53 activity, knock-in mice have

been created. Phosphorylation of serine 20 after DNA damage by Chk2 kinase is thought

to be an important activating modification (Caspari, 2000). A mouse that has mutated

murine serine 23 to alanine (human serine 20) is defective for apoptosis in B-cells and

develops B-cell lymphomas after a long latency period relative to p53 knockout mice

Page 24: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

15

(MacPherson et al., 2004). These results suggest that phosphorylation of a single residue

on p53 may not be sufficient to activate the protein; abrogation of a single

phosphorylation site so far has not been able to recapitulate loss of the p53 protein.

Multiple mutations to delete phosphorylation sites or sites of other post-translational

modifications are probably required to completely mimic loss of function of the p53

protein.

The multiple modifications on several residues of p53 has led researchers to

hypothesize a ‘p53 code’ similar to the histone code (Toledo and Wahl, 2006). Perhaps

these multiple modifications are cell type and stress specific. For example, cells treated

with cisplatin, a DNA damaging agent, and taxol, a microtubule inhibitor, show

differences in phosphorylation at serine 15 and serine 20 (Damia et al., 2001). The

HCT116 p53+/+ colon carcinoma cell line shows differences in phosphorylation and

acetylation in response to various stresses such as ultraviolet radiation, ionizing radiation,

adriamycin, taxol and nocodozole (Saito et al., 2003). This set of experiments performed

in a single cell type suggests that p53 becomes modified in a stress-specific manner.

Specific modifications may occur sequentially; threonine 18 phosphorylation by casein

kinase 1 delta appears to be dependent on phopshorylation of serine 15 (Dumaz et al.,

1999). Regulation of post-translational modifications and the functional consequences of

the post-translational modifications remains a complex problem. Several key questions

include which modifications may be sequential, the prior modifications and how they

contribute to sequential modification, and whether all modifications, or just a subset of

modifications occur on p53 at one time. As well, questions about how cell or tissue type

and stress type influences the modification of the p53 protein are also questions that have

Page 25: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

16

yet to be determined. Finally, the question about how these modifications affect p53

activity also needs to be addressed.

Phosphorylation of p53 at serine 15

Phosphorylation of serine 15 is one of the earliest and best characterized

modifications on p53 and is thought to be an activating modification. Serine 15 is

phosphorylated by multiple kinases including ATM, DNA-PK, ATR, and AMPK under

stress conditions such as DNA damage and glucose deprivation (Araki et al., 1999; Banin

et al., 1998; Canman et al., 1998; Toledo and Wahl, 2006). Mouse and stem cell models

for serine 18 (human serine 15) have been made where serine has been mutated to alanine

to prevent phosphorylation. In differentiated murine embryonic stem cells expressing a

knocked-in p53 S18A allele, induction of p53 by both ionizing radiation (IR) and

ultraviolet radiation (UV) were partially impaired, and impaired induction of p21 and cell

cycle arrest in response to IR was also observed (Chao et al., 2000a). Thymocytes in the

p53S18A mice show a reduction in the amount of apoptotic cells when compared to their

wild-type counterparts. In addition, target gene activation in response to IR and UV in

murine embryonic fibroblasts was reduced. The p53 S18A mice, however, were not

prone to spontaneous tumours. Whether these mice develop tumours when challenged

with DNA damaging agents remains undetermined (Chao et al., 2003). The observations

in the mouse model are supported by transfection experiments of human p53S15A into

H1299 cells that show reduced levels of apoptosis compared to the wild-type (Unger et

al., 1999b). These results suggest that phosphorylation of p53 on serine 15 contributes to

Page 26: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

17

activation of p53 activity, but that this activation step alone cannot sufficiently explain

the full effects of p53 tumour suppression and activity.

In addition, mice where both the murine serine 18 and serine 23 (human serine 15

and serine 20) sites were mutated exhibit little apoptosis in response to IR in the thymus

comparable to the levels observed in thymocytes from p53-/- mice, and significantly

decreased levels of apoptosis compared to the single S18A mutant thymocytes. p53-

induced neuronal apoptosis is also lost in the S18A/S23A double mutant compared with

wild-type cells. The S18A/S23A mice are also prone to spontaneous tumours, but tumour

formation is significantly slower than that observed in p53 -/- mice (Chao et al., 2006).

The combined effect of phosphorylation of serine 18 and serine 23 suggests that multiple

modifications most likely contribute to activation of p53.

Ubiquitination

Ubiquitin

Ubiquitin is a highly conserved 76 amino acid tag that was discovered in 1978

using rabbit reticulocyte lysates as a signal to mediate proteolysis (Hershko, 1996).

Ubiquitin acts as a tag for protein to signal degradation, subcellular localization,

membrane trafficking, DNA repair and chromatin dynamics (Murata et al., 2009).

Ubiquitin has seven lysines at position 6, 11, 27, 29, 33, 48 and 63. Ubiquitin forms

polymers by forming covalent linkages through any of these lysines (Xu et al., 2009).

Polyubiquitin linkages through lysine 48 and lysine 63 are the most well-characterized.

Polyubiquitinated substrates with a lysine 48 linkages tend to be rapidly degraded.

However, proteins that are polyubiquitinated with lysine 63 linkages are not degraded and

Page 27: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

18

tend to be involved in signal transduction pathways (Kirkpatrick et al., 2005; Newton et

al., 2008). The functional consequences of the other ubiquitin linkages remains unclear,

and present research is being conducted to determine the how those lysine linkages affect

the protein substrates.

When ubiquitin is attached to the substrate, it can be attached in multiple forms

(Figure 1.2). Monoubiquitination occurs when a single ubiquitin monomer is attached to

a single lysine on the substrate. A multiubiquitinated substrate has multiple ubiquitin

monomers attached through lysines on the substrate. Polybiquitination of a substrate

indicates that a polymer of ubiquitin is attached to the substrate at one or more lysine

residues (Mukhopadhyay and Riezman, 2007).

The ubiquitination mechanism

The ubiquitination reaction forms a bond between the C-terminus of ubiquitin

(G76) and the ε-amino group of a substrate lysine residue either on ubiquitin or the

substrate to be ubiquitinated. The ubiquitination reaction requires at least three enzymes:

an E1 ubiquitin activating enzyme, an E2 ubiquitin conjugating enzyme, an E3 ubiquitin

ligase; an E4 ubiquitin chain assembly factor may be required if the E3 ubiquitin ligase is

unable to polyubiquitinate the protein (Koegl et al., 1999; Pickart, 2001).

During the ubiquitination reaction, the E1 activating enzyme forms a thiol ester

with the carboxyl group of G76 to activate the C-terminus of ubiquitin for nucleophilic

attack. The E2 conjugating enzyme then transiently carries the activated ubiquitin

molecule as a thiol ester, and the E3 ubiquitin ligase mediates transfer of ubiquitin to the

substrate. All ubiquitination reactions follow the same mechanism regardless of the fate

Page 28: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

SubstrateUb

Monoubiquitination

Multiubiquitiantion

Polyubiquitination

Figure 1.2 Ubiquitin Linkage Forms.

Ubiquitin can be conjugated to a substrate in many forms. Monoubiquitinationinvolves a single ubiquitin monomer conjugated to the substrate (A). Multiubiquitination involves several ubiquitin monomers conjugated to the substrate (B). Polyubiquitination involves conjugation of a ubiquitin polymer to the substrate (C).

A

B

C

SubstrateUb

UbUbUb

SubstrateUb

Ub

Ub

Ub

Ub

19

Page 29: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

20

of the substrate (Pickart, 2001; Pickart, 2004). The E4 chain assembly factor is important

for catalyzing polyubiquitination. In some specific cases, the E3 enzyme can only

catalyze monoubiquitination of the substrate, and association of the E4 enzyme with the

E3 ligase will promote polyubiquitination (Figure 1.3) (Hoppe, 2005; Koegl et al., 1999).

The circumstances that define when an E4 enzyme is required for ubiquitination are

unclear, and further studies will determine when and how the E4 chain assembly factor

functions.

The E1 ubiquitin activating enzyme activates ubiquitin for all the downstream

conjugating enzymes; there are two human E1 enzymes. The activation occurs when

magnesium-ATP binds ubiquitin to form a ubiquitin adenylate intermediate that serves as

the donor of ubiquitin to a cysteine in the E1 active site. Each fully loaded E1 molecule

carries two molecules of activated ubiquitin: one molecule is conjugated to the E1 and the

other ubiquitin is as an adenylate. The thiol-linked ubiquitin is transferred to the next

conjugating cascade, the E2 (Pickart, 2001).

There are approximately fifty ubiquitin conjugating enzymes, E2s, in humans

(Pickart, 2004). E2 enzymes bind tightly to loaded E1 molecules, but weakly to free

ubiquitin and free E1; this ensures that the ubiquitination reaction is efficiently carried

out. The E2 is also responsible for determining the type of ubiquitin linkage (lysine 6,

lysine 11, lysine 27, lysine 29, lysine 33, lysine 48, lysine 63) that is formed (Chen and

Sun, 2009). The E1 enzyme active site is responsible for transferring ubiquitin from the

E1 to the E2. Each ubiquitin conjugating enzyme can bind to a subset of E3 ubiquitin

ligases. Once the E2 is bound to an E3, the ubiquitin ligase catalyzes transfer of the

ubiquitin from the E2 to either the E3 or the substrate (Pickart, 2001).

Page 30: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

E1

E3

E4

Poly-ubiquitination

Substrate

Degradation

Ub-activating enzyme

Ub-conjugating enzyme

Ub-ligase

Ub-chain-assembly

factor

E2

Ub

E2

E3

ATP

AMP + PPi

Ub

Ub

Ub

Ub

Ub

UbUb

UbUb

Figure 1.3 Ubiquitination Reaction Mechanism

ATP is required to activate ubiquitin that binds the E1 ubiquitin activating enzyme. The E1 transfers the E2 to the ubiquitin conjugating enzyme. The E2 binds to the E3 ubiquitin ligase that binds to the substrate to mediate polyubiquitinatin of the substrate. Occasionally, an E3 ubiquitin ligase may require an E4 ubiquitin chain assembly factor to help mediate polyubiquitination, the signal for degradation.

21

Page 31: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

22

The E3 ubiquitin ligase confers substrate specificity. In the human system there

are about 600 E3 ubiquitin ligases. The E3 binds to the substrate via the degron – the

ubiquitination signal, conferring substrate specificity, and the E3 also catalyzes the

ligation of one or more ubiquitins to the subtrate (Pickart, 2004). There are two classes

of ubiquitin ligases based on structure: homologous to E6-associated protein carboxy

terminus (HECT) domain containing ligases and really interesting new gene (RING)

domain containing ubiquitin ligases (Pickart, 2001).

The first HECT domain E3 ligase was discovered in cells infected with human

papilloma virus (HPV). The E6 protein promotes degradation of p53 through ubiquitin

mediated proteolysis. The E6 protein of HPV binds to a cellular protein called the E6-

associated protein (E6-AP) to form a complex that ubiquitinates and degrades p53. The

C-terminus of the E6-AP contains the HECT domain for which this class of enzyme is

named. HECT domain E3 ligases are modular: the N-terminus of the protein is

responsible for conferring substrate specificity through binding, and the C-terminus

contains the HECT domain that binds the E2 to mediate the ubiquitination reaction.

HECT domains are not interchangeable, and the HECT domain will covalently bind

ubiquitin before transferring it to the substrate (Pickart, 2001).

The RING finger ubiquitin ligases contain the RING domain that consist of a

series of histidine and cysteine residues with specific spacing that coordinates two zinc

ions in a cross-brace structure with the consensus sequence CX2CX(9-39)CX(1-3)HX(2-

3)C/HX2CX(4-48)CX2C. The zinc ions and ligands are catalytically inert, and the

spacing of the zinc ligands is conserved in the RING finger family which allows the

domain to function as a scaffold for protein-protein interaction (Zheng et al., 2000). The

Page 32: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

23

RING finger domain is responsible for binding the E2 conjugating enzyme, and the

ubiquitin is transferred directly from the E2 to the substrate; RING finger ubiquitin

ligases serve to bring the E2 and the substrate into proximity. This class of E3 ubiquitin

ligases can further be subdivided into two classes: single subunit and multi-subunit

ligases (Pickart, 2001).

The multi-subunit ligases compartmentalize the RING finger domain and the

substrate recognition domain. For example, the SCF (Skp1-Cul-F-box) E3 ligase

complex contains Cullin1 (Cul1) as a scaffold protein. The Cullin family of proteins

form scaffolds for the multi-subunit ubiquitin ligases. Cul1 interacts with Rbx1 which

contains a RING finger to bind the E2 conjugating enzyme. Cul1 also interacts with

Skp1 to mediate interaction with the F-box protein that confers substrate specificity. The

multi-subunit ubiquitin ligases have all the components of a single unit ubiquitin ligase

broken into several proteins (Pickart, 2001). Another example of a multi-subunit is the

cyclosome/anaphase promoting complex (APC) that promotes degradation of mitotic

cyclins, some anaphase inhibitors and spindle-associated proteins, which are all degraded

at the end of mitosis and also degrades β-catenin (Hershko and Ciechanover, 1998).

The single-subunit RING finger ubiquitin ligases also interact with the E2 through

the RING finger. The same protein mediates substrate specificity with another part of the

protein through other motifs. RING E3 ubiquitin ligases may also mediate

autoubiquitination resulting in self degradation (Pickart, 2001). Examples of single

subunit RING E3 ligases include cIAP1 that ubiquitinates caspase-3 and caspase-7 and

Cbl that targets receptor tyrosine kinases such as EGFR (Jackson et al., 2000).

Page 33: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

24

Ubiquitination and signalling

Although ubiquitin was initially discovered as a tag for proteolytic degradation,

current findings indicate that ubiquitin also plays an important role in non-proteolytic

signalling. In NF-κB signalling, non-proteolytic ubiquitination plays a critical role in

activating the pathway. To activate transcription of downstream targets of NF-κB, the

IκB complex must be phosphorylated by IKK and then degraded through ubiquitin-

mediated proteolysis. There are several mechanisms in which non-proteolytic ubiquitin

plays a role in activation of NF-κB. In one example, TRAF6 has ubiquitin ligase activity,

and catalyzes lysine 63-linked polyubiquitination of IRAK1 and itself. The lysine 63

polyubiquitin chains on TRAF6 bind and activate the TAK1 complex. IRAK1 and

TRAF6 that are both polyubiquitinated through lysine 63 linkages, associate with the IL1

receptor complex. The NEMO/IKK complex binds to the lysine 63-linked polyubiquitin

chains on IRAK1 through the NEMO ubiquitin binding domain. NEMO/IKK binding to

polyubiquitinated IRAK1 brings the IKK complex into proximity with the activated

TAK1 complex that phosphorylates IKK targeting it for degradation (Figure 1.4) (Chen

and Sun, 2009). Based on the upstream receptor and cell type, the substrates that are

ubiquitinated and the ligases that catalyze ubiquitination in the NF-κB pathway may vary

(RIP1, NEMO and MALT1 can also be lysine 63 polyubiquitinated based on the

upstream signalling pathway); however, it remains clear that ubiquitination is an integral

part to the regulation of this pathway (Chen and Sun, 2009).

Ubiquitination represents an important component of chromatin biology and gene

expression. Monoubiquitinated histone H2B resides at the promoter of highly expressed

genes, including the p21 promoter after p53 activation (Minsky et al., 2008). In addition,

Page 34: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

IKKβ IKK

TRAF6

IRAK1MyD88

Figure 1.4 Ubiquitination in the NF-κB pathway.

Stimulation of the IL-1 receptor (IL-1R) leads to recuitment of adaptor proteins MyD88,IRAK1 and TRAF6 to the receptor. TRAF6 funcions as an E3 ubiquitinligase that catalyzes the synthesis of K63 polyubiquitin chains with the help of Ubc13/Uev1A E2 complex. TAB2 subunit of the TAK1 kinase complex binds the K63 polyubiquitin chains; this results in activation of TAK1 kinase the phosphorylates IKKβ to activate the kinase. IKKβ is recuited to the receptor complex by binding NEMO; NEMO binds the polyubiquitin chains on TRAF6 or IRAK1. IKKβ activation phosphorylates the inhibitor IκB leading to its degradation and NF-κB activation. (adapted from Chen and Sun, 2009)

Ubc13

Uev1A

Ub

Ub

Ub

Ub

Ub

Ub Ub Ub Ub

NEMO

Ub

TA

B2

TA

K1

PP

p65

p50

IκB p65

p50

IκB

P

P

Degradation

p65

p50

NF-κB target genes

IL-1R25

Page 35: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

26

at sites of double strand breaks, chromosomes are ubiquitinated by RNF-8. RNF168 aids

in RNF-8 mediated lysine 63 ubiquitination of H2A and H2AX. Lysine 63 polyubiquitin

chains at sites of double strand breaks are important for retaining 53BP1 and BRCA1,

factors that are required for double strand break repair (Doil et al., 2009). In addition,

ubiquitination also plays an important role in DNA damage signalling.

Ubiquitination on p53 is also important for non-proteolytic regulation. E4F1 is an

E3 ubiquitin ligase that ubiquitinates lysine 320 of p53. E4F1 catalyzes lysine 48-linked

polyubiquitination on p53 that leads to increased binding to chromatin and activation of

p53-dependent cell cycle arrest (Le Cam et al., 2006).

Ubiquitination and protein degradation

The 26S proteasome is responsible for degrading ubiquitinated substrates. The

26S proteasome is a 2.5 MDa complex that is made up of two components: a 20S core

particle and the 19S regulator particle (Hanna and Finley, 2007). The 20S core particle

is a barrel-shaped complex that consists of seven proteins. The peptidolytic activity of

the proteasome is found in the center of the core particle barrel (Groll et al., 1997). There

are three distinct peptidolytic activities in the core particle and each are represented twice;

the enzymes found in the core particle are a tryptic activity, a chymotryptic activity and a

post-acidic activity (Kisselev et al., 2006).

The 19S is the regulatory subunit and is a 1MDa complex. It can be further

subdivided into two subcomplexes the base and the lid. The base aids in substrate

unfolding and is located proximal to the core particle; the unfolding of the protein is

required since the barrel of the 20S proteasome core particle is too small to degrade

Page 36: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

27

folded proteins (Hanna and Finley, 2007). The base of the subunit contains ATPases that

have chaperone activity and are thought to be required for unfolding of the protein prior

to degradation (Braun et al., 1999). The lid of the regulatory subunit is involved in

substrate recognition. The regulatory complex contains proteins such as Dsk2 and Rad23

that are responsible for binding to lysine 48 linked ubiquitin and targeting them for

unfolding and subsequently proteolysis by the core particle (Lam et al., 2002; Rao and

Sastry, 2002).

Ubiquitin-mediated proteolysis regulates diverse cellular pathways including cell

cycle progression and NF-B activation. The mammalian G1 regulators cyclin E and

cyclin D1 are targeted for ubiquitination by phosphorylation at specific single sites, and

must be degraded to allow progression into S-phase. In the NF-κB pathway, the IκB

inhibitor is degraded by ubiquitin-mediated proteolysis and this is necessary for NF-B

activation (Hershko and Ciechanover, 1998).

Experiments with a lysine 48-linkage specific antibody indicates that lysine 48-

linked ubiquitin is associated with the 20S proteosome consistent with the hypothesis that

lysine 48-linked ubiquitin usually targets proteins for proteasomal degradation (Newton

et al., 2008). Experiments in yeast, show that lysine 48-linked ubiquitin is stabilized in

the presence of proteasomal inhibition which also supports the hypothesis that lysine 48-

linked polyubiquitination target proteins for degradation (Xu et al., 2009). As well, a

yeast strain that has expresses ubiquitin that has substituted arginine for lysine at position

48 is lethal (K48R), suggesting that lysine 48-linked ubiquitin-mediated proteolysis is

crucial for survival (Spence et al., 1995).

Page 37: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

28

Recent experiments have also implicated lysine 11 linked ubiquitin chains in

degradation. In yeast, inhibition of the proteasome results in a four-fold accumulation of

lysine 11 linked polyubiquitin chains; these polyubiquitin chains are implicated in

endoplasmic reticulum-associated degradation (ERAD) as deletion of Doa10, an E3

ubiquitin ligase involved in ERAD, also decreases lysine 11 polybiquitination. In

addition, a yeast strain that expresses ubiquitin with lysine 11 mutated to arginine has

increased sensitivity to endoplasmic reticulum stress-inducing reagents such as

dithiothreiotol and tunicamycin (inhibits the synthesis of N-linked glycoproteins in the

endoplasmic reticulum). These experiments suggest that lysine 11 linked ubiquitin plays

an important role in ERAD (Xu et al., 2009).

The previous observations suggest that although lysine 48 linked ubiquitin chains

are critical for survival and substrate degradation, other ubiquitin linkages are also

important for degradation. In the previously described example, it could suggest that

different linkages may be formed in response to different stresses for targeting to the

proteasome.

Deubiquitination

Deubiquitination is performed by deubiquitinating enzymes, ubiquitin specific

proteases that can reverse target-protein ubiquitination by editing or diassembling

polyubiquitin chains. Deubiquitinasese specifically cleave ubiquitin from ubiquitin-

conjugated protein substrates, ubiquitin precursors, ubiquitin adducts and polyubiquitin.

Deubiquitintases have been implicated in several diseases including cancer and

neurodegenerative disorders (Ventii and Wilkinson, 2008).

Page 38: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

29

There are five families of deubiquitinases: four are cysteine proteases and the fifth

is a family of metalloproteases. The four classes of cysteine proteases are subdivided

based on their ubiquitin protease domains: ubiqutin-specific proteases (USP), ubiquitin

C-terminal hydrolases (UCH), otubain proteases (OTU), and Machado-Joseph disease

proteases (MJD) (Ventii and Wilkinson, 2008).

Deubiquitinases are involved in several pathways. USP1 is a deubiquitinase

implicated in Fanconi-anemia related cancer; USP1 deubiquitinates monoubiquitinated

FANCD2 after cells exit S-phase or re-enter the cell cycle after DNA damage (Nijman et

al., 2005). In addition, the deubiquitinase USP11 interacts with BRCA2 and promotes

deubiquitination of BRCA2; downregulation of USP11 through RNAi increases cellular

sensitivity to DNA damaging agent mitomycin C in a BRCA-2 dependent manner

(Schoenfeld et al., 2004).

Deubiquitination plays an important role in regulating the NF-κB pathway.

CYLD mediates negative regulation of the pathway by specifically deubiquitinating

lysine 63 polyubiquitinated proteins TRAF2, TRAF6 and NEMO (Kovalenko et al., 2003;

Trompouki et al., 2003). A special case of a deubiquitinating enzyme is A20, a ubiquitin

editing enzyme that downregulates NF-κB signalling. A20 functions as a ubiquitin

editing enzyme on RIP1, an important mediator of TNFR1 induced NF-κB activation.

A20 removes ubiquitin from RIP1 that is polyubiquitinated through K63 linkages. In

addition, A20 has the ability to catalyze K48 linkages on RIP1 to target it for proteolysis

(Wertz et al., 2004).

At the proteasome, ubiquitin is recycled and only the substrate is degraded. The

26S proteasome is associated with deubiquitinating enzymes to facilitate ubiquitin

Page 39: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

30

recycling (Yao and Cohen, 2002). The deubiquitinases Rpn11/S13 and Uch37 are stably

associated with the proteasome and USP14/Ubp6 is transiently associated with the

proteasome. RNAi experiments suggest that Rpn11 and either Uch37 or Ubp6 are

required for proper substrate processing (Koulich et al., 2008). Interestingly, deletion of

Ubp6 in yeast results in increased degradation of ubiquitinated cyclin B relative to the

wild-type yeast strain. Ubp6 is proposed to function through progressive

deubiquitination in which deubiquitination occurs sequentially from the N-terminus to the

C-terminus of the ubiquitin chain one monomer at a time. In the absence of Ubp6, Rpn11

catalyzes a more efficient deubiquitination reaction through en block chain release

(cleavage of the whole ubiquitin chain at the C-terminus in one reaction) (Hanna et al.,

2006).

Deubiquitination enzymes play an important and complex role in many pathways.

The function and regulation of these enzymes remains for the most part to be determined,

but remain vital to the understanding of the function of ubiquitination in the cell.

Ubiquitination and regulation of p53

The p53 protein has a short half-life under unstressed conditions due to regulation

through ubiquitin mediated proteolysis. There are several ubiquitin ligases that regulate

p53 including Mdm2, Pirh2, Cop1, ARF-BP1, TOPORS, Synoviolin, Trim24 CARPs

(Allton et al., 2009; Chen et al., 2005a; Dornan et al., 2004b; Haupt et al., 1997; Honda et

al., 1997; Leng et al., 2003; Rajendra et al., 2004; Yamasaki et al., 2007; Yang et al.,

2007). The physiological role of these ubiquitin ligases in the cell, and in regulating p53

function is unclear at the moment, and further studies need to be performed.

Page 40: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

31

ARF-BP1 is HECT E3 ubiquitin ligase. ARF-BP1 is inhibited by the tumour

suppressor ARF. ARF-BP1 ubiquitinates p53 and targets it for proteasomal degradation.

Knockdown of ARF-BP1 by RNAi stabilizes p53, increases p21 levels, and increases

p53-dependent apoptosis (Chen et al., 2005a). ARF-BP1 is also known as Mule, and has

also been found to ubiquitinate and degrade the pro-survival protein Mcl-1 (Zhong et al.,

2005). The physiological function of ARF-BP1/Mule is unclear at the present time since

p53 and Mcl-1 have diametrically opposing functions. Further research needs to be

completed to determine the role of ARF-BP1/Mule in regulating p53 and Mcl-1; perhaps

co-factors help redirect the ubiquitin ligase function of ARF-BP1/Mule resulting in a

specific regulation for each substrate.

Cop1 is a RING finger E3 ubiquitin ligase that targets p53 for degradation. Cop1

siRNA enhances p53-dependent G1 arrest and sensitizes cells to ionizing radiation

(Dornan et al., 2004b). Cop1 is also phosphorylated by ATM on serine 387 after DNA

damage, and this phosphorylation site dissociates the p53-Cop1 interaction (Dornan et al.,

2006). Cop1 has also been found to be overexpressed in breast and ovarian

adenocarcinomas (Dornan et al., 2004a).

Mdm2 is the first ubiquitin ligase found to antagonize p53 (Momand et al., 1992;

Oliner et al., 1992). It is also the best characterized. Mdm2 knock out mice are

embryonic lethal prior to implantation, and these mice die by massive apoptosis;

embryonic lethality is fully rescued by deletion of p53 and partially rescued by deletion

of the pro-apoptotic gene bax (Chavez-Reyes et al., 2003; Jones et al., 1995; Montes de

Oca Luna et al., 1995). Mdm2 gene amplification has also been observed in over one

third of human sarcomas that retained wild-type p53, and patients overexpressing Mdm2

Page 41: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

32

in sarcomas have a worse prognosis (Oliner et al., 1992; Onel and Cordon-Cardo, 2004).

Mdm2 regulation of p53 has been targeted for chemotherapy. The nutlin group of

inhibitors abrogates the Mdm2/p53 interaction. Treating tumour xenografts in mice with

nutlins causes cell cycle arrest and apoptosis suggesting that nutlins could eventually be

used to treat tumours that use Mdm2 to inactivate p53 (Vassilev et al., 2004).

Mdm2 regulates p53 stability through ubiquitination and this ability to regulate

p53 function is dependent on the RING finger (Haupt et al., 1997; Honda et al., 1997;

Honda and Yasuda, 2000; Kubbutat et al., 1997). Mdm2 binds to p53 in both of their N-

terminal hydrophobic domains, the interaction is mediated by the hydrophobic residues

phenylalanine 19, tryptophan 23 and leucine 26 on p53 that insert into the hydrophobic

cleft on Mdm2 (Kussie et al., 1996). Binding of p53 to Mdm2 promotes ubiquitinatation

of p53 at six lysine residues in the C-terminus (370, 372, 373, 381, 382, 386) (Kubbutat

et al., 1998; Nakamura et al., 2000; Rodriguez et al., 2000). To catalyze

polyubiquitination of p53, Mdm2 requires an E4 chain elongation factor. To date, there

are two chain elongation factors that have been identified for Mdm2-mediated p53

polyubiquitination the first is histone acetylase p300, and the second is the transcription

factor Yin Yang 1 (Gronroos et al., 2004; Grossman et al., 2003; Sui et al., 2004). Yin

Yang 1 can form a ternary complex with p53 and Mdm2, and ablation of Yin Yang 1 by

siRNA results in increased levels of p53 and p21 and an inhibition of cellular

proliferation (Sui et al., 2004).

Mdm2 dissociates from p53 after DNA damage, and this presumably contributes

to stabilization of the p53 protein (Shieh et al., 1997). There are several mechanisms that

could account for this dissociation including Chk2-mediated phosphorylation of p53 on

Page 42: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

33

serine 20 that is proposed to disrupt the binding of Mdm2 to p53 (Chehab et al., 1999;

Unger et al., 1999a).

Regulation of Mdm2 occurs at the transcriptional and post-translational level. At

the transcriptional level, the Mdm2 gene has two promoters: the P1 promoter and the P2

promoter. The P1 promoter is required for maintaining basal levels of Mdm2. The P2

promoter is located in intron 1 and contains a p53 binding site and it is where p53 induces

Mdm2 expression (Iwakuma and Lozano, 2003; Wu et al., 1993). Mdm2 can also be

regulated transcriptionally by a naturally occurring polymorphism in the P1 Mdm2

promoter. SNP 309 contains either thymidine or guanosine bases in the DNA, the

guanosine polymorphism creates a higher affinity binding site for the transcriptional

activator Sp1 and results in increased Mdm2 mRNA and protein at the basal level.

Individuals with the guanosine polymorphism exhibit accelerated sporadic and hereditary

tumourigenesis (Bond et al., 2004).

In addition, Mdm2 also promotes auto-ubiquitination resulting in auto-inhibition

through proteasomal degradation (Honda and Yasuda, 2000). Mdm2 auto-ubiquitination

is antagonized by HAUSP that deubiquitinates Mdm2 and stabilizes it. Since HAUSP

stabilizes Mdm2, p53 becomes destabilized (Cummins et al., 2004; Li et al., 2004).

HAUSP has also been shown to deubiquitinate p53 and MdmX (Li et al., 2002a;

Meulmeester et al., 2005). In addition, p53 and Mdm2 compete for the same binding

pocket on HAUSP, and Mdm2 seems to have a stronger interaction with HAUSP than

p53 (Sheng et al., 2006). How HAUSP functions to regulate p53 levels is unclear at the

moment, perhaps there could be additional cofactors that modulate HAUSP activity either

promoting or inhibiting deubiquitination of a particular substrate.

Page 43: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

34

There are also several proteins that can bind to Mdm2 to modulate its activity.

Upon oncogenic stress, ARF binds to Mdm2, and relocates it to the nucleolus so that it is

unable to degrade p53 (de Stanchina et al., 1998; Honda and Yasuda, 1999; Palmero et al.,

1998; Pomerantz et al., 1998). Additional proteins such as PML, and nucleolar protein

NPM, L23, and L11 also bind to Mdm2 and sequester it to prevent Mdm2-mediated

degradation of p53 (Bernardi et al., 2004; Dai et al., 2004; Jin et al., 2004; Kurki et al.,

2004; Zhang et al., 2003).

Regulation of Mdm2 also occurs through post-translational modifications. ATM-

mediated phosphorylation of Mdm2 on serine 395 resulted in a decreased ability of

Mdm2 to degrade p53; Wip1 phosphatase dephosphorylates serine 395 on Mdm2 to

increase the degradation of p53 by Mdm2 (Khosravi et al., 1999; Lu et al., 2007; Maya et

al., 2001). Mdm2 is also phosphorylated by Akt/PKB on serines 166 and 188, and this

modification stabilizes Mdm2. Knockout of Akt/PKB in murine embryonic fibroblasts

increases p53 and p21 levels (Feng et al., 2004). Mdm2 is also phosphorylated by c-Abl

after DNA damage on tyrosines 276 and 294; these phosphorylations inhibit Mdm2

degradation of p53 by promoting association with the inhibitor ARF (Dias et al., 2006).

Regulation of p53 stability is complex, and further experiments are required to

determine not only how p53 is regulated, but also how different stress conditions affect

the proteins that regulate p53. Using Mdm2 as an example, several factors such as

protein cofactors, and post-translational modifications influence Mdm2 activity, and as a

result affect p53 activity. As well, there is increasing complexity for the regulation of

p53 as several ubiquitin ligases also target p53 for degradation, this high degree of

functional redundancy must be addressed through additional experiments and animal

Page 44: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

35

models to determine which are the critical ubiquitin ligases, and whether each of these

ligases may regulate p53 under different conditions or in different cell types.

Pirh2

Pirh2 was initially identified as a protein that interacts with the androgen receptor,

and is also known as ARNIP (androgen receptor N-terminal interacting protein) that

functions as a ubiquitin ligase (Beitel et al., 2002). The Benchimol lab identified Pirh2 in

DP16.1 p53ts cells. The DP16 cells are murine erythroleukemia cells transformed by

Friend cell virus, and lack endogenous p53 expression; the DP16.1 p53ts cells are stably

transfected with a mutant p53 allele that encodes valine instead of alanine at codon 135.

The p53 A135V mutant is temperature sensitive (ts) and results in a protein that adopts

the mutant conformation and is inactive at 37ºC and adopts the wild-type conformation

and is active at 32ºC (Johnson et al., 1993; Michalovitz et al., 1990). A differential

display experiment was performed in the DP16.1 p53ts cells to identify novel genes that

are regulated by p53 by comparing DP16.1 p53ts cells grown at 32ºC, with active p53, to

those grown at 37ºC, with mutant p53. One of the targets identified was Pirh2, a gene

that encodes a 261 amino acid, 30 kDa protein with a cysteine-rich RING motif (Leng et

al., 2003).

The Pirh2 gene is expressed in a wide variety of tissues including testis, kidney,

lung, and brain with highest expression observed in both heart and liver. Pirh2 is induced

by DNA damage in a p53-dependent manner in both human and murine cells, though p53

induction appears to be cell-type and stress specific (Feng et al., 2007). Chromatin

immunoprecipiation, luciferase assays and EMSA (electrophoretic mobility shift assay)

Page 45: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

36

experiments indicate that p53 binds to intron 3 of Pirh2 to induce its expression.

Antisense experiments also indicate that knockdown of Pirh2 causes an increase in p53

protein, and results in a modest increase in G1 arrest (Leng et al., 2003). Further

experiments have also suggested that Pirh2 can only ubiquitinate the tetrameric form of

p53, and that Pirh2 can also undergo auto-ubiquitination (Sheng et al., 2008). Pirh2

regulation of active p53 suggests that it is a less potent ubiquitin ligase for p53 when

compared to Mdm2.

Pirh2 has also been shown to be overexpressed in both murine and human lung

cancers. Using semi-quantitative analysis of immunoblots, Pirh2 protein expression was

elevated by at least two fold in 84% of tumour samples examined (Duan et al., 2004).

Pirh2 has also been shown to be overexpressed in 89% of prostate cancers, and Pirh2

overexpression was correlated with more aggressive tumours as well (Logan et al., 2006).

Additional targets for Pirh2 have been identified. These targets include HDAC1

(hisone deacetylase 1), the G1 cyclin-dependent kinase inhibitor p27/Kip1, SRβ (signal

recognition particle receptor beta subunit) and ε-COP subunit of the coatomer complex

(Abe et al., 2008; Hattori et al., 2007; Logan et al., 2006; Maruyama et al., 2008). Pirh2

has also been shown to interact with the histone acetyl transferase Tip60, and hNTKL-

BP1 (human N-terminal kinase-like binding protein 1) (Logan et al., 2004; Zhang et al.,

2005). Whether these proteins are substrates, or how they affect Pirh2 activity is unclear

at the present.

Pirh2 stability is enhanced by interaction with the transcription factor PLAG2

(pleomorphic adenoma gene like 2) and MV P (measles virus phosphor protein);

Page 46: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

37

interaction with either PLAG2 or MV P stabilizes Pirh2 by inhibiting Pirh2 auto-

ubiquitination (Chen et al., 2005b; Zheng et al., 2007).

Pirh2 can also interact with calmodulin, and is phosphorylated by CaMKII

(calmodulin dependent kinase II) on threonine 154 and serine 155. CaMKII

phosphorylation of Pirh2 decreases Pirh2 stability, and decreases Pirh2 mediated

ubiquitination and degradation of p53 (Duan et al., 2007).

Further studies need to determine the key substrates that are targeted for Pirh2-

mediated degradation, the cofactors that mediate degradation and the physiological role

of Pirh2 in regulating p53 and in the cell.

Thesis Hypothesis

It is known that Pirh2 targets p53 for ubiquitin mediated proteolysis, however,

several other ubiquitin ligases also target p53 for degradation. The reason for such a high

degree of functional redundancy is unclear, and furthermore, the physiological role of

Pirh2 in regulating p53 is also undefined. In addition to regulating p53, it is also well-

known that Pirh2 regulates multiple targets, and it is likely that not all substrates of Pirh2

have been identified, and any protein cofactors that aid in Pirh2-dependent ubiquitin

mediated proteolysis also have yet to be identified.

The purpose of this study is two fold. The first is to identify additional substrates

for Pirh2 and any additional protein cofactors that aid in ubiquitination. The second aim

of this study is to address Pirh2 regulation of p53 function.

Page 47: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

38

CHAPTER 2

Identification of Pirh2 Interacting

Proteins

Thank you to Dr. Jack Greenblatt and Dr. Cheryl Arrowsmith who provided the His-Flag

vector used for purification, Stephen Chung for cloning the His-Flag vector, Weili Ma for

generating Pirh2-/- E1A/Ras His-Flag and Pirh2 His-Flag clones and the York Mass

Spectrometry facility performed all Mass Spectrometry analysis.

Page 48: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

39

ABSTRACT

Pirh2 was identified as a p53 target gene in a Friend-virus transformed mouse

erythroleukemia cell line (Leng et al., 2003). Pirh2 has also been characterized as having

E3 ubiquitin ligase activity, and can target p53, HDAC1, p27/Kip1, SRβ and ε-COP for

ubiquitin mediated proteolysis. Pirh2 can also bind to the androgen receptor, hNTKL-

BP1, calmodulin, and Tip 60 (Abe et al., 2008; Beitel et al., 2002; Duan et al., 2007;

Hattori et al., 2007; Leng et al., 2003; Logan et al., 2004; Maruyama et al., 2008; Zhang

et al., 2005). Here, I described two approaches to identify additional targets for Pirh2

ubiquitination, and cofactors involved in Pirh2-mediated ubiquitination using tandem

affinity purification and a GST-pull down assay.

INTRODUCTION

Pirh2 is a newly identified gene that is regulated by p53 and can also regulate p53

through ubiquitin mediated proteolysis. Pirh2 has several targets including HDAC1

(hisone deacetylase 1), the G1 cyclin-dependent kinase inhibitor p27/Kip1, SRβ (signal

recognition particle receptor beta subunit) and ε-COP subunit of the coatomer complex

(Abe et al., 2008; Hattori et al., 2007; Logan et al., 2006; Maruyama et al., 2008).

Ubiquitin ligases are known to have several targets for ubiquitination, and require several

cofactors including ubiquitin conjugating enzymes (Deshaies and Joazeiro, 2009). To

identify these interactors, we used two methods: the first is a tandem affinity purification

method and the second a GST-pull down assay.

Tandem affinity purification (TAP) was initially developed in yeast. It involves

fusion of a protein to a protein A and calmodulin binding peptide tag separated by a

Page 49: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

40

tobacco etch virus (TEV) protease site. The dual tags allow for sequential purification of

a protein and any associated proteins; the dual tag also provides less background

contaminating proteins and false positives than purification performed with a single tag

(Rigaut et al., 1999). A TAP tagged protein is first purified by binding to IgG beads that

interact with the protein A. The complex is then released by cleavage with TEV protease,

and passed over calmodulin beads that interact with the calmodulin binding peptide. The

complex can then be eluted using EGTA that chelates the calcium ions required to

mediate the calmodulin/calmodulin binding peptide interaction (Puig et al., 2001).

One advantage of the dual tag approach is that it can identify complexes where as

the yeast two hybrid system was designed to identify binary interactions. In comparison,

TAP tagging also purifies proteins under more physiological conditions than the yeast

two hybrid system and can map complexes under different stress conditions that may be

dependent on post-translational modification. In contrast, the yeast two hybrid system

relies on overexpression of proteins and does not account for post-translational

modifications. When TAP tagging was developed in yeast, the tagged gene was knocked

into the genome to replace the normal gene – this strategy is advantageous since it

eliminates competition for interacting proteins from the untagged protein, and the tagged

protein is also regulated by the same promoter as the normal protein so expression levels

would be similar (Puig et al., 2001). In mammalian cells, the gene cannot be easily

knocked into somatic cells, but to express similar levels of the tagged protein to the

endogenous protein, stable transfectants are constructed and clones that express the

protein at similar levels to the endogenous protein are selected (Gingras et al., 2005).

Page 50: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

41

Several variations of dual purification tags have emerged. In addition to the

classical TAP-tag, we have used two different variations in an attempt to identify proteins

that interact with Pirh2. The first is the SPA (Sequential Peptide Affinity) tag that

contains a triple flag tag and calmodulin binding peptide separated by the TEV protease

site. The second tag is a triple flag tag and six times histidine separated by the TEV

protease site (Zeghouf et al., 2004). In addition, we also performed a GST-pull down

experiment to identify interacting proteins under normal conditions and conditions of

DNA damage.

EXPERIMENTAL PROCEDURES

Cell Culture. H1299 cells and Pirh2 -/- immortalized kidney fibroblasts were cultured in

-MEM with 10% fetal calf serum (FCS). Pirh2 -/- MEFs were transformed by retroviral

infection with pBABE-E1A/Ras and cultured in Dulbecco’s modified Eagles medium

(DMEM) with 10% FCS. Pirh2 -/- immortalized kidney fibroblasts were prepared by

harvesting the kidney from the mouse. The kidney was separated into cells by passing

through a 70 µm strainer (BD Biosciences). Cells were incubated overnight in -MEM

with 10% FCS. The following morning, cellular debris and media were removed, and

replaced with fresh -MEM with 10% FCS. Adhered cells were washed gently with PBS,

and grown in -MEM with 10% FCS for one week. Cells were then immortalized

through the 3T3 protocol.

Plasmids. Human Pirh2 cDNA was PCR amplified from pcDNA3-hPirh2 (Leng et al.,

2003) and cloned into pCMV-TAP to generate an N-terminal TAP-tagged Pirh2 construct.

Page 51: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

42

PCR amplification of hPirh2 was also used to create a 3’ SPA tagged hPirh2 construct

and a 3’ His-Flag tagged hPirh2 construct. PCR amplification of hPirh2-SPA and

hPirh2-His-Flag was used to clone into pBabe neo and pBabe puro respectively.

Generation of Clones Expressing pCMV TAP hPirh2. H1299 cells were plated (4 x

105) in a 10 cm dish the day before transfection. Calcium phosphate was used to transfect

10 µg of pCMV-TAP, 10 µg pCMV-TAP-hPirh2, 10 µg of pcDNA3 SPA or 10 µg

pcDNA3 hPirh2-SPA. H1299 cells were selected with 400 µg/mL neomycin (Invitrogen)

and then transferred to 6-well plates and then screened for clone expression.

Generation of Clones Expressing pBabe neo hPirh2-SPA. 293 cells were plated at a

density of 3 x 105 per 6 cm plate and FuGENE (Roche) was used to transfect 1 µg pCL-

Eco and 1 µg of pBabe neo SPA or pBabe neo hPirh2 SPA. After transfection, media

was removed and replaced with 5 mL of DMEM 10% FCS. After 48 hours, media was

collected and filtered (0.45 µm, Millipore) and used for infection of Pirh2-/- E1A/Ras

MEFs. Pirh2-/- E1A/Ras MEFs were plated at a density of 1 x 105 cells per 10 cm plate.

Cells were selected in 200 µg/mL neomycin for one week. Clones were transferred to 6-

well plates and then screened for clone expression by western blot analysis.

Generation of Clones Expressing pBabe puro hPirh2-HF. 293 cells were plated at a

density of 3 x 105 per 6 cm plate and FuGENE (Roche) was used to transfect 1 µg pCL-

Eco and 1 µg of pBabe puro HF or pBabe puro hPirh2-HF. After transfection, media was

removed and replaced with 5 mL of DMEM 10% FCS. After 48 hours, media was

Page 52: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

43

collected and filtered (0.45 µm, Millipore) and used for infection of Pirh2-/- E1A/Ras

MEFs. Pirh2-/- E1A/Ras MEFs were plated at a density of 1 x 105 cells per 10 cm plate.

Puromycin (1 µg/mL, Invivogen) was used to select the cells for two days. Clones were

transferred to 6-well plates and then screened for clone expression by western blot

analysis.

Ubiquitination Assay. H1299 cells, TAP or TAP-hPirh2 clone 11 were plated at a

density of 5 x 105 cells per 10 cm plate the day before transfection. Cells were

transfected using calcium phosphate with 10 µg pcDNA3 or pcDNA3 hPirh2 and 1 µg

HA-ubiquitin vector and harvested 40 hours after transfection for western blot analysis.

Coimmunoprecipitation. H1299 cells, TAP or TAP-hPirh2 clone 5 were plated at a

density of 5 x 105 cells per 10cm plate the day before transfection. Cells were transfected

using calcium phosphate with 1µg pcDNA3 or pcDNA3 p53 and harvested 40 hours after

transfection in IP lysis buffer (50 mM Tris-Cl pH 8.0, 5 mM EDTA, 150 mM sodium

chloride, 0.5% NP-40). 1 mg of protein was loaded on rabbit IgG-agarose beads (GE

Healthcare) and rotated at 4ºC for 2 hours. Beads were washed three times with IP wash

buffer (50 mM Tris-Cl pH 8.0, 5 mM EDTA, 150 mM sodium chloride, 0.1% NP-40).

Beads were boiled in 1x Laemmli SDS buffer and separated by SDS-PAGE for western

analysis.

Page 53: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

44

Western blots. Antibodies against hPirh2 (Leng et al., 2003), calmodulin binding

peptide (CBP, Calbiochem), histidine (Qiagen), Flag and β-actin (Sigma) and non-

immune rabbit IgG (Cedarlane) were used for western blots.

TAP Purification. H1299 CMV TAP or H1299 TAP-hPirh2 cells were plated in 20

plates each at a density of 1 x 106 per 15 cm plate. Cells were grown to approximately

90% confluency, and lysed 40 mM HEPES pH 7.5, 120 mM sodium chloride, 1 mM

EDTA, 0.5% NP-40, EDTA-free protease inhibitor tablet (Roche). Lysates were spun at

16,000g. An equal volume and protein concentration of TAP and TAP-hPirh2 lysates

were loaded onto rabbit IgG-agarose beads (200 µL, GE Healthcare) and rotated for 4

hours at 4ºC. IgG beads were washed three times with wash buffer (10 mM Tris pH 8.0,

100 mM sodium chloride, 0.5 mM EDTA, 0.1% NP40) at 4ºC, and once with TEV

cleavage buffer (10 mM Tris pH 8.0, 150 mM sodium chloride, 0.5 mM EDTA, 0.1%

NP40, 1 mM DTT). IgG beads were rotated in 300 µL TEV cleavage buffer with 100 µg

TEV protease at 4ºC overnight. Beads were washed with 200 L TEV cleavage buffer

twice, and the fractions were combined. The eluate was supplemented with 2 L of 2 M

calcium chloride and 400 L calmodulin binding buffer (10 mM Tris pH 8.0, 150 mM

sodium chloride, 1 mM imidazole, 1 mM magnesium acetate, 0.2 mM calcium chloride,

10 mM β-mercaptoethanol, 0.1% NP-40). Calmodulin beads (100 µL, GE Healthcare)

were added to the mixture and rotated at 4ºC for 4 hours. Beads were washed three

times with calmodulin binding buffer and washed once with calmodulin binding buffer

without calcium chloride. To elute, the beads were boiled in 1x Laemmli SDS buffer,

Page 54: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

45

and the samples were separated using SDS-PAGE. Silver staining was performed using a

method compatible for mass spectrometry (Shevchenko et al., 1996).

SPA Purification. Pirh2-/- E1A/Ras SPA MEFs or Pirh2-/- E1A/Ras hPirh2-SPA MEFs

were plated into 20 plates each at a density of 3 x 106 per 15 cm plate. Cells were grown

to approximately 90% confluency, and lysed in 40 mM HEPES pH 7.5, 120 mM sodium

chloride, 1 mM EDTA, 0.5% NP-40, EDTA-free protease inhibitor tablet (Roche). The

lysate was cleared by spinning at 16,000 g. An equal concentration and volume of SPA

and hPirh2-SPA lysate was loaded onto 200 µL packed M2-agarose beads (Sigma) and

incubated for 4 hours with rotation at 4ºC. M2 beads were washed three times with wash

buffer (10 mM Tris pH 8.0, 100 mM sodium chloride, 0.5 mM EDTA, 0.1% NP40) and

then washed once with TEV cleavage buffer (10 mM Tris pH 8.0, 150 mM sodium

chloride, 0.5 mM EDTA, 0.1% NP40, 1 mM DTT) at 4ºC. M2 beads were incubated in

300 µL TEV cleavage buffer with 100 µg TEV protease at 4ºC with rotation overnight.

M2 beads were then washed with 200 L TEV cleavage buffer twice and the fractions

were combined. The eluate was supplemented with 2 L of 2 M calcium chloride and

400 L of calmodulin binding buffer (10 mM Tris pH 8.0, 150 mM sodium chloride, 1

mM imidazole, 1 mM magnesium acetate, 0.2 mM calcium chloride, 10 mM β-

mercaptoethanol, 0.1% NP-40). Calmodulin beads (100 µL, GE Healthcare) were added

to the mixture and rotated at 4ºC for 4 hours. Beads were washed three times with

calmodulin binding buffer and washed once with calmodulin binding buffer without

calcium chloride. Beads were then washed once with elution buffer without EGTA (50

mM ammonium bicarbonate). The protein mixture was eluted by rotating at 4ºC with

Page 55: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

46

500 µL elution buffer (50 mM ammonium bicarbonate, 25 mM EGTA). A second

elution was performed, and the fractions were combined. Samples were lyophilized, and

proteins were identified using LC-MS/MS.

His-Flag Purification. Pirh2-/- E1A/Ras MEFs expressing either His-Flag or Pirh2-His-

Flag were plated each into 80 plates (15 cm) and grown to 90% confluency. Cells were

harvested and resuspended in 40 mL lysis buffer (10 mM Tris pH 8.0, 100 mM sodium

chloride, 5 mM EDTA pH 8.0, 10 mM sodium diphosphate, 10 mM sodium fluoride, 1

mM sodium orthovanadate 0.35% NP-40, 10% glycerol, EDTA-free protease inhibitor

tablet (Roche)). Cell lysates were cleared by centrifugation at 16,000 rpm. A equal

volume and concentration of His-Flag or Pirh2-His-Flag lysate was added to 1 mL of

M2-agarose beads (Sigma), and rotated for 4 hours at 4ºC. Beads were washed three

times with Flag wash buffer (10 mM Tris pH 8.0, 100 mM Sodium chloride, 5 mM

EDTA, pH 8.0, 0.1% NP-40, EDTA-free Protease inhibitor tablet (Roche)) at 4ºC. The

M2-beads were then resuspended in 3mL of Flag-peptide elution buffer (0.1 mg/mL Flag

peptide (Sigma), 20 mM Tris pH 8.0, 100 mM sodium chloride, 0.1 mM EDTA, 5 mM

Imidazole) and rotated overnight at 4ºC. The M2 beads were washed twice with 2 mL

Flag-peptide elution buffer. Eluates were pooled and incubated with 800 µL nickel-

agarose beads (Sigma), and rotated for 2 hours at 4ºC. Nickel-agarose beads were

washed three times with Ni-NTA wash buffer (20 mM Tris pH 8.0, 100 mM sodium

chloride, 0.1 mM EDTA, 5 mM Imidazole). Three elutions were performed with 2 mL

of Ni-NTA elution buffer (10 mM Tris pH 8.0, 100 mM ammonium bicarbonate, 0.1 mM

EDTA, 0.4 M imidazole). The eluted fractions were pooled, and dialyzed overnight at

Page 56: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

47

4ºC against 100 mM ammonium bicarbonate. Eluates were frozen at -80ºC, and

lyophilized for identification of the protein mixture using LC-MS/MS.

GST-pull down. Purification of GST-Pirh2 was performed as described (Sheng et al.,

2008). Pirh2-/- kidney fibroblasts were grown to 85% confluency and harvested or

treated with 200 ng/mL of adriamycin (Sigma) for 24 hours. Cells were resuspended in

GST-pull down lysis buffer (50 mM Tris pH 8.0, 5 mM EDTA, 5 mM EGTA, 100 mM

sodium chloride, 0.4% NP-40, 10% glycerol, 10 mM sodium diphosphate, 10 mM

sodium fluoride, 1 mM sodium orthovanadate, and EDTA-free protease inhibitor

(Roche)). 2 mg of Pirh2-/- immortalized kidney lysate either untreated or treated with

adriamycin was incubated with GST-glutathione beads or GST-Pirh2 beads overnight at

4ºC. Beads were washed three times with wash buffer (50 mM Tris pH 8.0, 5 mM EDTA,

5 mM EGTA, 100 mM sodium chloride, 0.1% NP-40, 10 mM sodium diphosphate, 10

mM sodium fluoride, 1 mM sodium orthovanadate, and EDTA-free protease inhibitor

tablet (Roche)). Protein was eluted by boiling in 1x Laemmli SDS buffer. Samples were

run on 4-12% gradient gel (Invitrogen), and stained with Gel Code Blue Stain Reagent

(Pierce). Bands were excised for identification by mass spectrometry (MALDI-MS/MS).

RESULTS

Generation and characterization of TAP-hPirh2 clones.

To identify proteins that interact with Pirh2, we created an N-terminal TAP tag

Pirh2 construct (Fig 2.1A). Stable transfectants were made in H1299 cells and individual

Page 57: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

Pirh2

Protein ACBP

TEV Cleavage

___ 24

___ 35

___ 48

___ 54

Actin

TAP Tag

hPirh2

TAP-hPirh2

H1299

pcDNA3

hPirh

2

TAP hPirh

2 CL2

TAP hPirh

2 CL4

TAP hPirh

2 CL7

TAP hPirh

2 CL9

TAP hPirh

2 CL1

1

pCM

VTAP

A

B

WB: hPirh2

Figure 2.1 Generation of TAP-hPirh2 clones in H1299 cells.

(A) A diagram of TAP-Pirh2 .

(B) Western blot of Pirh2 and TAP-Pirh2 expression in H1299 stable clones (CL) and after transfection in H1299 cells. The top panel shows blotting with Pirh2 antibodies and the bottom panel shows blotting with rabbit IgG.

48

H1299

pcDNA3

hPirh

2

TAP hPirh

2 CL2

TAP hPirh

2 CL4

TAP hPirh

2 CL7

TAP hPirh

2 CL9

TAP hPirh

2 CL1

1

Actin

TAP Tag

TAP-hPirh2

___ 24

___ 35

___ 48

___ 54

WB: non-immunerabbit IgG

pCM

VTAP

Page 58: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

49

colonies were examined to identify clones that express TAP-Pirh2 at levels comparable to

endogenous Pirh2. Overexpression of TAP-Pirh2 is not desired since this could lead to

non-physiological interactions with the fusion protein. The results of the Pirh2 western

blot are shown in Fig 2.1B. As a result of the protein A tag, TAP-Pirh2 can be detected

either with Pirh2 antibodies or with rabbit IgG (Fig 2.1B). It is difficult to compare TAP-

Pirh2 levels with endogenous Pirh2 levels because the rabbit Pirh2 antibodies will bind to

the protein A tag as well as to the Pirh2 region so the best one can do is to identify clones

that express TAP-Pirh2 but do not overexpress TAP-Pirh2. Five clones were isolated:

two that express TAP-Pirh2 at relatively low levels (clones 2,4), two at more moderate

levels (clones 7, 9) and a single clone that expresses at high levels (clone 11) (Fig. 2.1B).

The TAP tag comprises about half the molecular mass of TAP-hPirh2. We were

concerned that the tag may interfere with protein function, so we performed two assays to

determine function. The first is a ubiquitination assay; H1299 cells stably transfected

with TAP or TAP-hPirh2 were co-transfected with empty vector and HA-ubiquitin (Fig

2.2A). After performing a western blot for HA, and observing the characteristic smear in

the TAP-hPirh2 indicative of ubiquitination activity, we determined that the TAP-hPirh2

fusion protein construct retains the ability to promote ubiquitination of proteins. We used

HA-ubiquitin cotransfected with either empty vector or Pirh2 as negative and positive

controls, respectively. We also tested to determine if TAP-hPirh2 retained the ability to

bind p53. We transfected either empty vector or p53 into parental H1299 cells, TAP or

TAP-hPirh2 cells, and performed immunoprecipitation assays with rabbit IgG and

blotted for p53 (Fig 2.2B). p53 only coimmunoprecipitated with TAP-hPirh2. This

suggests that TAP-hPirh2 retains the ability to bind p53. Based on the ubiquitination and

Page 59: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

Vecto

r + H

A-Ub

TAP: V +

HA-Ub

TAP-h

Pirh2:

V +

HA-Ub

Actin

170 __

130 __

100 __

72 __

55 __

hPirh

2 +

HA-Ub

WB: HA

IP: Rabbit IgGWB: p53

Transfected withempty pcDNA3

Transfected with pcDNA3 p53

H1299

TAP

TAP-hPirh

2

p53

A

B

Figure 2.2 Functional Assays for TAP-hPirh2 Activity.

(A) Parental H1299 cells were transiently transfected with empty vector (pcDNA3) or pcDNA3 hPirh2 and HA-ubiquitin. TAP and TAP hPirh2 CL11 were transiently transfected with empty vector and HA ubiquitin. Western blots were performed for HA ubiquitin and actin as a loading control.

(B) H1299, TAP or TAP hPirh2 CL9 cells were transiently transfected with either empty vector or pcDNA3 p53. Lysates were immunoprecipitated with rabbit IgG and western blotted for p53 (B).

50

Page 60: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

51

coimmunoprecipitation assays, we conclude that TAP-hPirh2 retains activity and is a

suitable reagent to identify Pirh2 interacting proteins.

Purification of TAP and TAP-hPirh2.

Purification was optimized using TAP and the high expressing TAP-hPirh2 clone

(CL11). Cells were lysed and fractions were collected at each stage in the purification

process: Cell lysate, lysate unbound to IgG, elution after TEV protease, unbound to

calmodulin beads, and bound to calmodulin beads. Calmodulin binding peptide antibody

was used to track TAP-hPirh2 at each step in the purification (Fig 2.3A). We were able

to successfully purify TAP-hPirh2 from high expressing cells, however, we were unable

to detect purification of TAP-hPirh2 from the lower expressing clones (data not shown).

TAP purification was repeated on a preparative scale using CL11, and the fractions were

separated on SDS-PAGE and silver stained to visualize protein bands. Bands were then

excised and sent for identification by mass spectrometry (Fig 2.3B). Many of the bands

were unidentifiable; those that could be identified (Hsc70, alpha-s-casein, tubulin) appear

to be contaminants or non-specific interacting proteins.

Generation of clones and purification of Pirh2-SPA.

Based on the results of the TAP purification experiment, we decided to create a

Pirh2-SPA (Fig 2.4A). The SPA tag is a triple flag tag, TEV protease site and calmodulin

binding peptide construct. The SPA construct is a smaller tag, and is a C-terminal tag; in

contrast, the protein A section of the TAP tag was about 20 kDa and the construct was an

N-terminal tag. We felt that the TAP-hPirh2 construct may not be folding properly, and

Page 61: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

WB: CBP

55 __

40 __

33 __

24 __

72 __

17 __

TAP TAP-hPirh2

CBP–hPirh2

TAP-hPirh2

Lysate

Unbou

nd Ig

G

TEV Elua

te

Unbou

nd

Calm.

Bound

Calm

Lysate

Unbou

nd Ig

G

TEV Elua

te

Unbou

nd

CalmBou

nd C

alm

TAP

CBP

A

Figure 2.3 Purification of TAP and TAP-hPirh2

(A) TAP and TAP-hPirh2 CL11 cells were lysed in NP-40 lysis buffer. After centrifugation, TAP and TAP-hPirh2 were purified by binding to IgG, cleavage with TEV protease, and binding to calmodulin beads. Fractions in each step were separated by SDS-PAGE and transferred to PVDF, and blotted with antibody against CBP.

(B) Eluates from TAP Purification were separated by SDS-PAGE and silver stained. Bands were excised and identified by mass spectrometry.

TAP TAP-hPirh2

55 __

40 __

33 __

24 __

72 __

CBP-Pirh2

Hsc70

Tubulin

Alpha-S-casein

B

52

Page 62: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

B

40__

33__

24__

17__

PARENT

SPAhPirh2-SPA

H1299

PARENTSPA

hPirh2-SPAPirh2-/- MEFs

SPA

hPirh2-SPA

WB: Flag

C

D

Actin

55 __

40 __

33 __

SPA Pirh2-SPA

Pirh2-CBP

EWB:hPirh2

Pirh2-SPA55__

40__

33__

24__

17__

11__

Pirh2

WB: hPirh2

H1299

hPirh

2

11 20 24 25 1 2 3 4 9 1914SPA hPirh2-SPA

Pirh2-SPA

SPA

55__

40__

33__

24__

17__

11__

Actin

WB: Flag

11 20 24 25 1 2 3 4 9 1914SPA hPirh2-SPA

H1299

hPirh

2

Figure 2.4 Generation and Purification of hPirh2-SPA Clones

(A) Diagram of hPirh2-SPA fusion protein.

(B) Western blots were performed on lysates prepared from the population stably transfected H1299 cells with SPA and hPirh2 SPA vectors, and Pirh2-/-E1A/ras MEFs infected with pBabe neo SPA or hPirh2-SPA.

(C, D) H1299 cells stably transfected were cloned and western blots were performed as indicated.

(E) Purification of CBP-hPirh2 from Pirh2-/-E1A/ras transformed cells (E).

Pirh2

FlagCBP

TEV Cleavage

A 53

Page 63: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

54

therefore it was associated with heat shock proteins; we thought that by reducing the size

and the placement of the tag could help the protein fold properly. Pirh2-SPA clones were

stably transfected into H1299 cells or retrovirally infected into Pirh2-/- E1A/Ras MEFs

(Fig. 2.4B). Expression of Pirh2-SPA was quickly shut down in the Pirh2-/- E1A/Ras

MEFs; as a result, stable Pirh2-SPA expressing clones could not be generated (data not

shown). Pirh2-SPA and SPA clones were generated in H1299 cells, and analyzed by

blotting with Flag (Fig 2.4C) and Pirh2 (Fig 2.4D) antibodies. The Pirh2-SPA fusion

protein was expressed stably, but at significantly lower levels than endogenous Pirh2 (Fig.

2.4C,D). Since there is no endogenous Pirh2 to compete with the tagged construct in the

Pirh2-/- MEFs, we used the Pirh2-SPA-expressing Pirh2-/- MEFs to purify the protein.

We fractionated the eluate using SDS-PAGE, and expression of the tagged construct was

greatly reduced, but not completely lost as we were able to purify Pirh2-CBP (Fig. 2.4E).

The final eluate was lyophilized and sent for analysis by mass spectrometry resulting in

the identification of calpain small subunit 1 and S-100 calcium binding protein beta

subunit. Since purification using the calmodulin binding peptide is a calcium dependent

reaction, calcium associated proteins are often purified as contaminants. We did not

purify heat shock proteins or chaperone proteins suggesting that changing the tag, cell

type, expression level, or a combination of these factors may help to identify potential

interactors.

Generation of clones and purification of Pirh2-HF.

Based on the results of the SPA experiment, and that the construct lost expression

very quickly in the Pirh2 -/- E1A/Ras MEF population, we decided to use the His-Flag

Page 64: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

His

CL1 CL2 CL9 CL10 CL11 CL14

Actin

His-Flag Pirh2-His-FlagB

His-Flag Pirh2 – His-Flag

Pirh2 – His - Flag

C

Pirh2

FlagHis

TEV CleavageA

Figure 2.5 Generation and Purification of Pirh2-His-Flag Clones.

(A) Diagram of Pirh2-His-Flag fusion protein.

(B) Pirh2-/- E1A/Ras MEFs were infected with retrovirus made from pBabepuro His-Flag or Pirh2-His-Flag and selected in puromycin. Clones (CL) were generated and lysates from the clones were used for western blot analysis with the indicated antibodies.

(C) A pool of His-Flag and Pirh2-His-Flag clones were used for purification. Successful purification was confirmed using western blot analysis.

His

55

Page 65: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

56

expression construct. The new construct uses a six times histidine tag and three time flag

peptide for sequential purification (Figure 2.5A). We suspected that the calmodulin

binding peptide present in Pirh2-SPA may have been toxic to the Pirh2-/- E1A/Ras MEFs

(resulting in rapid loss of expression of Pirh2-SPA), and that replacing this peptide with

the histidine tag might alleviate the toxicity. Using retroviral infection of Pirh2-/-

E1A/Ras MEFs, we generated clones of the empty vector (His-Flag) or Pirh2-His-Flag

and picked clones of variable expression (Figure 2.5B). We then pooled a number of the

Pirh2-His-Flag or His-Flag clones, and performed the dual purification protocol. A pool

of clones 9, 10, 11 and 14 to for Pirh2-His-Flag and clones 1 and 2 for His-Flag was used

for the experiment (Fig 2.5B). We chose to make of pool of the clones for the experiment

to reduce clonal artefacts that may arise from purification from a single clone. Also there

is variability in expression levels in each clone; a pool of the Pirh2-His-Flag clones

equalizes expression levels among the four clones. A Western blot performed on the

eluates from the purification protocol indicates that Pirh2-His-Flag was successfully

purified (Figure 2.5C). Mass spectrometry was performed on the eluates. Only

contaminating proteins (keratin, trypsin) were identified.

Identification of Pirh2 interactors using GST-pull down.

Since we were unable to identify specific Pirh2 interacting proteins using the dual

purification techniques, we additionally used a GST-hPirh2 construct to identify

interacting proteins from immortalized Pirh2-/- kidney fibroblasts. Immortalized kidney

fibroblast cells were used since immortalized cells undergo less morphological and

genetic changes than transformed fibroblasts, and we wanted to identify targets under

Page 66: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

57

physiological conditions. Pirh2 is also expressed in the kidney (Leng et al., 2003).

Identification of targets in the kidney is important in determining the role of Pirh2 in this

tissue. GST-hPirh2 and GST were bound to glutathione beads. Lysates from untreated

fibroblasts and fibroblasts treated with adriamycin were bound to GST-hPirh2 or GST to

determine if Pirh2 binds proteins differentially under conditions of stress (Fig 2.6).

Interacting proteins were separated by SDS-PAGE, bands were excised and identified

using mass spectrometry. Based on the results of the mass spectrometry analysis, we

identified PKC as a candidate interacting protein.

DISCUSSION

In an attempt to identify proteins that interact with Pirh2 we used TAP tagging,

and variations on the TAP tag (SPA tag, and His-Flag tag). In all cases, we purified

tagged Pirh2, but had difficulty identifying interacting proteins. We then tried to identify

interacting proteins by using GST-hPirh2 in a GST pull down experiment. We were able

to identify a single candidate interactor PKC that needs to be confirmed.

The difficulties encountered in the dual pull down and GST pull down

experiments may be explained by several reasons. The most likely explanation is that

substrate – ubiquitin ligase interactions and cofactor (i.e. ubiquitin conjugating enzyme) –

ubiquitin ligase interactions are thought to be weak and transient, and, hence, may not

survive the washes involved in the dual purification strategies or the GST-pull down

approach that were used (Deshaies and Joazeiro, 2009). If dual purification strategies are

used to identify substrates and cofactors for Pirh2, crosslinking the proteins would

Page 67: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

Figure 2.6 GST-pull Down to Identify Novel Pirh2 Interacting Proteins.

Immortalized Pirh2-/- kidney fibroblasts were either untreated or treated for 24 hours with 200 ng/mL adrimycin. Cells were lysed in non-denaturing conditions, and an equal mass and volume of protein was incubated with either GST or GST-hPirh2 bound to glutathione beads. Bound proteins were eluted by boiling SDS, run on an SDS-page gel and stained with coomassie. Bands appearing only in the GST-hPirh2 lanes were excised and identified by mass spectrometry.

GSTGST-hPirh

2

GSTGST-hPirh

2Untreated Adriamycin

GST***

*

Pirh2

55 __

40 __

33 __

24 __

72 __

17 __

130 __

100 __

170 __

GST-hPirh2

Pirh2

Semaphorin 4D

PKC

* Bands sent for mass spectrometry but not identified

58

Page 68: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

59

strengthen the interaction and facilitate the identification of interactors (Gingras et al.,

2005). Based on the results of the GST-pull down and mass spectrometry, PKC is a

candidate interacting protein. This interaction will need to be confirmed by endogenous

coimmunoprecipitation and confocal microscopy. If the interaction is confirmed, the

nature of the interaction between Pirh2 and PKC will also need to be determined.

There are several possibilities the most obvious being that Pirh2 may ubiquitinate PKC

or that PKC may phosphorylate Pirh2. Future experiments will need to address these

issues should the interaction be confirmed by endogenous coimmunoprecipitation.

PKC is a serine/threonine kinase, and activation of PKC is a critical pro-

apoptotic signal (Reyland, 2007). When cells are exposed to apoptotic stress such as

DNA damaging agents, oxidative stress and death receptors, PKC becomes

phosphorylated on multiple tyrosine residues and translocates into the nucleus.

Phosphotyrosine PKC is then cleaved by caspase-3. Caspase-3 cleavage causes the

release of an activated pro-apoptotic C-terminal fragment (Reyland, 2007). PKC and

the active cleavage fragment must be retained in the nucleus to cause apoptosis (DeVries-

Seimon et al., 2007). In a mouse model, PKC -/- mice exhibit a reduction in IR, UV

and etoposide induced apoptosis in the parotid gland (Humphries et al., 2006). Activated

PKC is targeted for ubiquitin-mediated proteolysis (Lu et al., 1998).

The results of the GST-pull down show little if any difference between

adriamycin treated and untreated cells (Fig. 2.6). There could be several possibilities.

We harvested cells from adriamycin treatment after 24 hours, and perhaps this time point

was too late to catch changes in the interaction profile. Additional pull-downs should be

performed with cells treated with adriamycin at various time points; a time course of pull

Page 69: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

60

downs would be ideal to track the differences, if any. Also, varying the stress may

produce different interacting proteins; adriamycin causes double strand breaks, UV

radiation, serum starvation, and heat shock could be other options to determine if

interactors vary with the stress response. Again, a time course could also be useful to

determine when these interactions occur.

Clearly, additional approaches to identifying proteins that interact with Pirh2 need

to be performed. The approaches that are to be undertaken must address the transient

nature of the substrate/cofactor and ubiquitin ligase interaction to be able to successfully

purify the interactors.

Page 70: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

CHAPTER 3

Pirh2 targets phosphoserine 15 p53

for ubiquitination

Thank you to Dr. Razq Hakem and the Hakem lab that provided the WT and Pirh2-/-

mice, immunohistochemistry for p53 on irradiated spleen (Anne Hakem), and TUNEL on

irradiated spleen and thymus (Benedicte Lemmers).

61

Page 71: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

62

ABSTRACT

The p53 protein is activated and stabilized by post-translational modifications in

response to stress and promotes cell cycle arrest and apoptosis. Pirh2 is a p53-regulated

gene which encodes an E3 RING finger ubiquitin ligase that binds to p53 and negatively

regulates p53 by targeting it for ubiquitin-mediated proteolysis. Pirh2 is functionally

similar to Mdm2 a well-characterized E3 RING finger ubiquitin ligase; they both target

p53 for degradation, and are transcriptional targets for p53 activity. The stability of the

p53 protein is primarily regulated through ubiquitin mediated proteolysis, and there are

multiple ubiquitin ligases targeting p53. Here we address the question of functional

redundancy and show that Pirh2 can target serine 15 phosphorylated p53 which is

reported to not be regulated by Mdm2.

Phosphoserine 15 p53 levels are significantly higher in irradiated splenocytes and

thymocytes from Pirh2-/- mice compared with Pirh2+/+ mice and this has a functional

consequence on the kinetics of bax induction and apoptosis. In addition, cells stably

expressing ectopic Pirh2 have decreased levels of phosphoserine 15 p53 and p21 after

DNA damage.

INTRODUCTION

The p53 tumour suppressor is a sequence-specific transcription factor that

promotes cell cycle arrest or apoptosis in response to various forms of cellular stress

(Levine, 1997). Cell cycle arrest normally occurs through transcriptional activation of

target genes such as the p21WAF1 cyclin-dependent kinase inhibitor; induction of p21 is a

key step in causing G1 cell cycle arrest (el-Deiry et al., 1993; Harper et al., 1993). Cells

Page 72: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

63

from p21 knockout mice are deficient in undergoing p53-dependent cell cycle arrest in

response to DNA damage (Brugarolas et al., 1995; Deng et al., 1995). p53-dependent

apoptosis is partially regulated by transcriptional activation of target genes including bax

(Yin et al., 1997).

The p53 protein is primarily regulated at the post-translational level. Under

normal conditions, the p53 protein is extremely unstable, and rapidly targeted for

ubiquitin-mediated proteolysis by the E3 ubiquitin ligase Mdm2 (Michael and Oren,

2003). After DNA damage, both Mdm2 and p53 undergo several post-translational

modifications including acetylation and phosphorylation. In the presence of double

strand breaks, activated ATM kinase can phosphorylate both p53 on serine 15 and Mdm2

on serine 395. These modifications are proposed to aid in the dissociation of the Mdm2-

p53 complex and contribute to the stabilization and activation of p53 as part of the stress

response (Banin et al., 1998; Maya et al., 2001; Shieh et al., 1997). Mice that express a

p53 serine 15 to alanine mutant are partially deficient in p53-dependent apoptosis and cell

cycle arrest in response to DNA damage suggesting that phosphorylation of p53 on serine

15 reflects p53 protein activation (Chao et al., 2000a; Chao et al., 2000b).

Several ubiquitin ligases have been identified that target p53 for ubiquitin

mediated proteolysis, and the reason for such a high degree of functional redundancy is

not known (Toledo and Wahl, 2006). Among the E3 ubiquitin ligases that target p53

have been identified is Pirh2 (p53-induced protein with a ring H2 domain) (Leng et al.,

2003). Pirh2 was identified as a p53 target gene in a differential display experiment

performed using the p53-null Friend-virus transformed mouse erythroleukemia cell line

(DP16.1/p53ts) that stably expresses a gene that encodes a temperature sensitive (ts)

Page 73: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

64

mutant p53 allele. Pirh2 gene expression is p53-dependent, however there appears to be

tissue or stress specific components to p53-induction of Pirh2. Previous experiments

have shown that Pirh2 can ubiquitinate p53 and target it for degradation; antisense

oligonucleotide-mediated repression of Pirh2 in normal fibroblast cells results in an

increase in p53 protein level, and a modest increase in cell cycle arrest (Leng et al., 2003).

In this study, we used splenocytes and thymocytes from Pirh2 null mice, and

noted that basal levels of p53 in these tissues remain unchanged compared to the wild-

type counterparts. After irradiation, both induction of total p53 levels and phosphoserine

15 p53 levels were higher in the Pirh2 null splenocytes and thymocytes compared to

wild-type splenocytes and thymocytes. This increase in total p53 and phosphoserine 15

p53 affects the kinetics of bax and apoptotic induction – bax and apoptosis are induced

earlier relative to the wild-type controls. In cells that overexpress Pirh2, levels of

phosphoserine 15 p53 and the p53 target gene p21 are reduced after DNA damage

compared to the vector controls. We also show that Pirh2 can coimmunoprecipitate

phosphoserine 15 p53 and Pirh2 null splenocytes show decreased levels of ubiquitinated

phosphoserine 15 p53. These results suggest that Pirh2 may directly regulate

phosphoserine 15 p53, and begins to address the question of functional redundancy

among the ubiquitin ligases that target p53 for degradation.

EXPERIMENTAL PROCEDURES

Cell Culture. MCF-7 cells and H1299 cells were cultured in -MEM supplemented with

10% fetal calf serum (FCS). MCF-7 clones were previously created and published (Leng

et al., 2003). 293 cells were cultured in Dulbecco’s MEM in 10% FCS. Splenocytes and

Page 74: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

65

thymocytes were cultured in RPMI supplemented with 10% FCS and 568 µM β-

mercaptomethanol.

Splenocyte and Thymocyte Culture Preparation. To prepare splenocyte and

thymocyte culture, spleen and thymus were harvested from age matched wild-type and

Pirh2-/- mice. The organs were homogenized in 70 µm cell strainers (BD Biosciences).

Cells were resuspended in red blood cell lysis buffer (Sigma) and washed once with

phosphate buffered saline (PBS). Splenocytes and thymocytes were cultured in RPMI,

10% FCS and 568 µM β-mercaptomethanol, and irradiated with 6 Gy of γ-IR from a

cesium 137 source. Wild-type and Pirh2-/- splenocytes and thymocytes were harvested at

time points after irradiation, and lysed in a 1x Laemmli SDS sample buffer for western

blot analysis.

Preparation of MCF-7 p53 shRNA Cells. MCF-7 cells were plated (1 x 105) in a 10 cm

dish one day before transfection and were then transfected with 10 µg of pcDNA3 Eco

plasmid using calcium phosphate. 293 cells (3 x 106) were plated in a 10 cm dish one day

before transfection with calcium phosphate and 5 µg pCl-Eco and 5 µg pSuper.puro

p53shRNA plasmid. The medium was changed one day after transfection, and the viral

supernatant was harvested 24 hours later and used to infect MCF-7 cells expressing the

ecotropic receptor in the presence of 8 µg/mL hexadimethrine bromide (Sigma) for 8

hours during the day at 37ºC. MCF-7 cells were then infected a second time with viral

supernatant and 8 µg/mL hexadimethrine bromide (Sigma) for 16 hours overnight at 37ºC.

Page 75: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

66

Infected MCF-7 cells were washed with PBS and incubated for 24 hours in -MEM and

10% FCS. MCF-7 cells were then selected with 2 µg/mL puromycin (Invivogen).

Western Blotting. Antibodies for p53 (DO-1, FL393), Mdm2 (SMP14) and p21 (C-19)

were obtained from Santa Cruz. The phosphoserine 15 p53 antibody was obtained from

Cell Signaling, and β-actin antibody from Sigma. Antibodies for Pirh2 were described

previously (Leng et al., 2003) or were obtained from Bethyl laboratories. Quantitation of

the phosphoserine 15 p53 and p53 signal were performed on the LiCOR Odyssey

fluorescence imager.

Immunohistochemistry. Whole body irradiation (6 Gy) was performed on matched

wild-type and Pirh2-/- mice. Tissues were fixed in buffered formalin, processed for

paraffin-embedded sectioning at 5 µm, and stained with H&E (Fisher). IHC was

performed using anti-p53 (FL393, Santa Cruz). TUNEL staining was performed on

histological sections using an in situ cell death detection kit (Boehringer Mannheim).

Coimmunoprecipitation. Rabbit IgG (Cedarlane), phosphoserine 15 p53 (Cell

Signaling) and human Pirh2 antibodies were irreversibly crosslinked to protein A beads

using DSS (Pierce) as per Seize X immmunoprecipitation kit instructions (Pierce). MCF-

7 cells were treated for 16 hours with 200 ng/mL adriamycin (Sigma), and 4 hours with

50 µM MG132 (Sigma). Cells were lysed in 600 µL IP lysis buffer (50mM HEPES pH

8.0, 5 mM EDTA, 100 mM sodium chloride, 0.35% NP-40, 10 mM sodium diphosphate,

10 mM sodium fluoride, 1 mM sodium orthovanadate, 50 µM MG132, and EDTA-free

Page 76: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

67

protease inhibitor tablet (Roche)). Cell lysates were rotated for 30 minutes at 4ºC with

0.625 mM DSP (Pierce), and 150 µL of 1 M Tris pH 8.0 was added and rotated at 4ºC for

30 minutes. Protein lysate (2 mg) was mixed with the crosslinked antibody beads and

rotated at 4ºC for 3 hours. Beads were washed three times with wash buffer (50 mM Tris

pH 8.0, 5 mM EDTA, 100 mM sodium chloride, 0.1% NP-40, 10 mM sodium

diphosphate, 10 mM sodium fluoride, 1 mM sodium orthovanadate). Elution was

performed 1x Laemmli SDS sample buffer with 0.1M DTT and heated at 60ºC for 5

minutes.

Ubiquitination Assay. Splenocyte cultures were prepared as previously described.

Wild-type and Pirh2-/- splenocytes were treated with 6 Gy γ-IR and 50 µM MG132 for 4

hours. Cells were lysed in 1x Laemmli SDS sample buffer with 10 mM sodium

diphosphate, 10 mM sodium fluoride, 1 mM sodium orthovanadate. Lysates were used

for western analysis.

Flow Cytometry. MCF-7 cells and clones were plated in a (1 x 106) 10 cm dish one day

before drug treatment. Cells were treated with 200 ng/mL adriamycin (Sigma) for 14

hours; for the last half hour of adriamycin treatment, cells were also treated with 10 µM

of BrdU (Sigma). Cells were fixed in 70% ethanol overnight at -20ºC. Cells were

counted and 1x106 cells were used per sample. Cells were resuspended in denaturing

solution (2 M hydrochloric acid and 0.5% Triton-X) for half an hour at room temperature.

Cells were then resuspended in 0.1 M sodium borate pH 8.5 for two minutes at room

temperature. Cells were washed in 2% bovine serum albumin (BSA) in PBS. Cells were

Page 77: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

68

resuspended in anti-BrdU-FITC antibody (BD Biosciences), RNase A, 0.5% Tween-20

and 1% BSA in PBS and incubated overnight at 4ºC. Cells were washed with 2% BSA in

PBS and resuspended in 10µg/mL propidium iodide (Sigma) and 0.05% Triton-X in PBS

for half an hour at room temperature. Cells were washed with 2% BSA in PBS and

resuspended in PBS for analysis by flow cytometry on a BD FACS Calibur.

RESULTS

Pirh2-/- Tissues Show Elevated Levels of p53 After DNA Damage. Pirh2-/- mice

were provided by Dr. Razq Hakem. Spleen and thymus were harvested from age

matched wild-type and Pirh2-/- mice, and irradiated with 6 Gy IR.

Immunohistochemistry on the spleen indicates that p53 levels are elevated 1 hour post-

irradiation in the Pirh2-/- spleen relative to Pirh2+/+ spleen (Fig. 3.1A). Western blot

analysis of non irradiated spleen (Fig 3.1B) and thymus (Fig 3.1C) revealed no difference

in p53 protein levels between the wild-type and Pirh2-/- strains. After irradiation,

however, p53 levels appeared to increase earlier in the Pirh2-/- spleen and thymus; at

later times after irradiation, the levels of p53 were similar in both strains indicating that

Pirh2 regulation of p53 may affect the kinetics of induction.

Pirh2-/- Tissues Show Elevated Levels of Phosphoserine 15 p53 After DNA Damage.

After DNA damage, p53 is phosphorylated by ATM on serine 15 (Banin et al., 1998).

Pirh2 has also been shown to interact with the DNA binding domain and the

oligomerization domain of p53, which are not affected by serine 15 phosphorylation

(Leng et al., 2003; Sheng et al., 2008). Irradiated splenocytes (Fig 3.1B) and thymocytes

Page 78: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

WT Pirh2-/-

Total p53

mPirh2

β-actin

0 0.5 1 2 4 6 0 0.5 1 2 4 6

WT Pirh2-/-Time(hr)

A

B

C

Figure 3.1 Elevated p53 levels in irradiated Pirh2-/- tissues

(A) Age matched WT and Pirh2-/- mice were irradiated with 6 Gy of IR and sacrificed at one hour. Spleens were harvested and stained for p53 levels by immunohistochemistry.

Splenocytes (B) and thymocytes (C) were harvested fro age matched WT and Pirh2-/- mice. Cells were irradiated with 6 Gy IR, harvested and lysed at indicated time points for western blotting

Total p53

mPirh2

β-actin

0 0.5 1 2 4 6 0 0.5 1 2 4 6

WT Pirh2-/-Time(hr)

PhosphoSer 15 p53

69

Page 79: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

70

(Fig 3.2A) from Pirh2-/- and wild-type mice were used to determine levels of

phosphoserine 15 p53. Levels of phosphoserine 15 p53 are increased in the Pirh2

knockout mice relative to the wild-type counterparts. Since levels of p53 are also

elevated in the Pirh2-/- mice, quantitation of phosphoserine p53 and total p53 levels were

performed using irradiated splenocytes in order to normalize for the increase in total p53.

The data are summarized in Fig. 3.2B, and are displayed as a ratio of phosphoserine 15

p53 levels to total p53 levels. The ratio of phosphoserine 15 p53 to total p53 is higher in

the Pirh2-/- splenocytes suggesting that the observed increase in total p53 does not

account for the increase in phosphoserine 15 p53. These data suggest that Pirh2 regulates

the level of the transcriptionally active form of p53 that is phosphorylated on serine 15.

Apoptotic Rate is Increased in Pirh2-/- Mice. To determine if the increased levels of

p53 protein, and phosphoserine 15 p53 has functional significance, we measured levels of

the p53 target gene Bax in splenocytes (Fig. 3.3A) and thymocytes (Fig 3.3B) after

irradiation. In accordance with the observed increase in total and phosphoserine 15 p53

levels, Pirh2-/- splenocytes and thymocytes also have increased levels of Bax protein.

Moreover, Bax induction occurs earlier compared with wild-type controls. Levels of

apoptosis were visualized using the TUNEL assay (Fig. 3.3C). Apoptosis in irradiated

spleen and thymus from Pirh2-/- mice occurs earlier than in wild-type cells. At later time

points, the number of apoptotic (TUNEL-positive) cells is similar. Hence the kinetics of

apoptosis induction is increased in Pirh2-/- thymocytes and splenocytes compared with

wild-type cells.

Page 80: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

0

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.8

0.9

1

0 1 2 4 6

Hours

Rat

io o

f S

15p

53/p

53to

t

Pirh2-/-

WT

PhosphoSer 15p53

Total p53

0 1 2 4 6 0 1 2 4 6

WTPirh2-/-Time (hrs)

A

PhosphoSer 15p53

β-actin

0 1 2 4 6 0 1 2 4 6

WT Pirh2-/-Time (hr)

Total p53

B

Figure 3.2 Elevated levels of phosphoserine 15 p53 in Pirh2-/- mice

Thymocytes were harvested from age matched WT and Pirh2-/- mice (A). Cells were treated with 6 Gy IR and harvested at various time points. Westerns were performed using indicaed antibodies and developed using film. Quantitation of splenocytes was performed using the LiCOR Odyssey (B).

71

Page 81: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

Thymus Spleenwt -/- wt -/-

0 hr

4 hr

8 hr

12 hr

β-actin

Bax

0 0.5 1 2 4 6 0 0.5 1 2 4 6

WT Pirh2-/-

mPirh2

Time(hr)

β-actin

Bax

0 0.5 1 2 4 6 0 0.5 1 2 4 6

WT Pirh2-/-

mPirh2

Time(hr)

A

B

C

Figure 3.3 Elevated levels of apoptosis in Pirh2-/- irradiated mice

Western blot analysis of Bax levels in irradiated splenocytes (A) and thymocytes (B) from age matched WT and Pirh2-/- mice. Cells were irradiated with 6 Gy IR and harvested at various time points.

(C) TUNEL staining of spleen and thymus harvested from age matched WT and Pirh2-/- mice irradiated with 6 Gy of IR.

72

Page 82: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

73

Pirh2 Can Regulate p53 Mutants for Serine 15. The observation that the accumulation

of phosphoserine 15 p53 occurs earlier after irradiation in Pirh2-/- cells prompted us to

test whether Pirh2 can regulate p53 serine 15 mutants. Pirh2, Mdm2 or empty vector

(pcDNA3) were cotransfected with wild-type p53 or p53S15A or p53S15D into p53-null

H1299 cells (Fig. 3.4). We used the wild-type p53 as a control, the p53S15A remains

unmodified as the protein cannot be phosphorylated and the p53S15D protein mimics

constitutive phosphorylation at serine 15 due to the acidic aspartic acid residue. Our

hypothesis is that Pirh2 can regulate all three constructs, where as Mdm2 would be

predicted to regulate wild-type p53, and the p53S15A protein, but not the p53S15D

protein. Transfected cells were collected 40 hours after transfection, and cell extracts

were prepared and analyzed by western blotting. As expected, Mdm2 and Pirh2 were

able to degrade wild-type p53 and p53S15A and resulted in decreased levels of p53

protein. Pirh2 is also capable of degrading p53S15D, and surprisingly, Mdm2 can

degrade this form of p53 as well. Results for Mdm2 degradation of p53S15D are

reported in the literature, and suggest that serine 15 phosphorylation p53 is not sufficient

to abrogate the interaction between p53 and Mdm2, and Mdm2 is still able to degrade the

p53 S15D mutant (Ashcroft et al., 1999; Dumaz and Meek, 1999). The ability of Pirh2 to

degrade the p53S15D mutant is consistent with the previous results that suggest that

Pirh2 regulates phosphoserine 15 p53.

Cells Overexpressing Pirh2 Decrease Levels of Phosphoserine 15 p53 and p21, But

Do Not Affect Cell Cycle Arrest. To complement the knock out experiments, MCF-7

cells that overexpress Pirh2 were used (Leng et al., 2003). If loss of Pirh2 results in an

Page 83: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

p53

Mdm2

hPirh2

β-actin

pcDNA3(

neo)

Mdm

2

hPirh

2pcD

NA3(ne

o)

Mdm

2

hPirh

2pcD

NA3(ne

o)

Mdm

2

hPirh

2

p53 wt p53 S15A p53 S15D

Figure 3.4 Mdm2 and Pirh2 can regulate p53 serine 15 phosphomutants

H1299 cells were cotransfected using calcium phosphate with either empty vector (pcDNA3), Mdm2 or hPirh2 and a p53 construct (wt, S15A, S15D). Lysates were made from the transfected cells at 40 hours after transfection, and western blots were performed for the indicated antibodies.

74

Page 84: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

75

increase in phosphoserine 15 p53 and the p53 target gene bax, in cells that overexpress

Pirh2, we should see an opposing effect: there should be a decrease in phosphoserine 15

p53 and the p53 target gene p21. MCF-7 cells were treated with adriamycin for 16 hours.

Clones overexpressing Pirh2 have decreased levels of phosphoserine 15 p53 and

consequently, p53 target gene, p21 (Fig 3.5) compared to vector controls and a control

that overexpresses Mdm2. However, we do not see a difference in the induced p53 levels

among the two vector controls, the Mdm2 overexpressing clone, and the two clones that

overexpress p53. Also, all clones were able to induce the p53 target gene Mdm2 equally

well suggesting that the effect of reduced phosphoserine 15 p53 protein level in the Pirh2

overexpressing clone may affect specific p53 target genes. To determine if there is a

functional consequence of decreased p21 induction in the Pirh2 overexpressing clones,

cell cycle analysis was performed using bromodeoxyuridine and propidium iodide flow

cytometry analysis. MCF-7 cells that stably express p53shRNA were used as a negative

control for cell cycle arrest. These cells show a reduced levels p53 and p21 in the

absence of stress by western blot and after treatment with adriamycin for 24 hours, the

MCF-7 p53shRNA the cells show no induction of p53 and p21 (Figure 3.6A). Results

are summarized in Fig. 3.6B by plotting the percentage of cells in S-phase. MCF-7 cells

that stably express p53shRNA fail to arrest in G1 in response to adriamycin treatment.

After 14 hours of adriamycin treatment, there is no significant decrease in cell cycle

arrest in response to Pirh2 overexpresssion. We have also examined various time points

using 200 ng/mL adriamycin (12 hours, 16 hours, 20 hours, and 24 hours) and different

concentrations of adriamycin (50 ng/mL, 100 ng/mL, 150 ng/mL adriamycin) and noted

no significant decrease in cell cycle arrest (data not shown). Although, the levels of

Page 85: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

p21

Pirh2

β-actin

Total p53

PhosphoSer 15 p53

Mdm2

pcDNA3.1

pcDNA3.

2

Mdm2

Pirh2-1

Pirh2-2

Control 200ng/mL Adriamycin

pcDNA3.1

pcDNA3.

2

Mdm2

Pirh2-1

Pirh2-2

Figure 3.5 Decreased levels of phosphoserine 15 p53 and p21 in Pirh2 overexpressing cells.

MCF-7 clones stably transfected with vector, Mdm2, or Pirh2 were created. Cells were either untreated or treated with 200 ng/mL adriamycin for 16 hours and harvested. Western blots were performed with the indicated antibodies.

76

Page 86: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

Figure 3.6 Overexpression of Pirh2 does not decrease S-phase after DNA damage

(A) To generate p53shRNA expressing cells, MCF-7 cells were transiently transfected with the ecotropic receptor, and infected with ecotropic virus that expresses the p53shRNA in the pSuper.retro puro backbone. MCF-7 cells were selected using 2ug/mL puromycin for 24 hours. Validation of the p53shRNA was performed by treating cells with 200µg/mL of adriamycin for 24 hours. Western blots were performed as indicated

(B) The stably transfected MCF-7 cells were either untreated or treated with 200 ng/mL of adriamycin for 14 hours. Cells were pulsed with 10 µM BrdU for half an hour and then fixed and stained with anti-BrdU-FITC antibody and PI. Cell cycle analysis was performed using flow cytometry.

77

0

10

20

30

40

50

60

p53s

hRNA C

ontro

l

pcDNA3.

1 Con

trol

pcDNA3.

2 Con

trol

Mdm

2 Con

trol

Pirh2-

1 Con

trol

Pirh2-

2 Con

trol

p53s

hRNA A

dr

pcDNA3.

1 Adr

pcDNA3.

2 Adr

Mdm

2 Adr

Pirh2-

1 Adr

Pirh2-

2 Adr

Ave

rage

S-p

hase

(%

)

A

B

p21

β-actin

p53

Adr - + - +

MCF-7Parental

MCF-7 p53shRNA

Page 87: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

78

phosphoserine 15 p53 and p21 are reduced in MCF-7 cells that overexpress Pirh2, this

does not appear to have an effect on cell cycle progression.

Pirh2 Coimmunoprecipitates with Phosphoserine 15 and Promotes Phosphoserine

15 p53 Ubiquitination. To determine if the regulation of phosphoserine 15 p53 by Pirh2

is direct, we performed coimmunoprecipitation and ubiquitination experiments.

Previous experiments have shown that unmodified p53 can interact with Pirh2, and Pirh2

can target p53 for polyubiquitination in in vitro experiments (Leng et al., 2003). An

endogenous coimmunoprecipitation experiment using MCF-7 cells treated with

adriamycin and the proteasomal inhibitor MG132 was performed. The results presented

in Fig 3.7A show that p53 phosphoserine 15 coimmunoprecipitates with Pirh2. In

addition, Pirh2 coimmunoprecipitates with phosphoserine 15 p53. These results clearly

show that Pirh2 can interact physically with p53 phosphorylated on serine 15.

Ubiquitination of phosphoserine 15 p53 was also performed in vivo using splenocytes

from wild-type and Pirh2-/- mice. Splenocytes were treated with IR and MG132 and

harvested for analysis by western blotting (Fig. 3.7B). Splenocytes from wild-type mice

show an increase in high molecular weight laddering of phosphoserine 15 p53 that

usually suggests polyubiquitination. These experiments indicate that phosphoserine 15

p53 interacts with Pirh2, and suggest that Pirh2 can promote the ubiquitination of

phosphoserine 15 p53.

Page 88: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

WB: PhosphoSer 15 p53

IP: R

abbi

t IgG

IP: P

irh2

IP: S

er15

p53

Inpu

t 1%

WB: Pirh2

A

B

Figure 3.7 Pirh2 interacts with and ubiquitinates phosphoserine 15 p53

(A) MCF-7 were treated for 16 hours with 200 ng/mL of adriamycin and for 4 hours with 50 µM MG132. Cells were harvested in 0.35% NP-40 lysis buffer and an equal amount and volume of protein was bound to either rabbit IgG, Pirh2 or phosphoserine 15 p53 antibody. Western blots were performed for the indicated antibodies .

(B) Splenocytes were harvested from age matched WT and Pirh2-/- mice that were irradiated with 6 Gy IR and treated for 4 hours with 50 µM MG132. Lysates were prepared and western blotting was performed with indicated antibodies.

PhosphoSer 15 p53

mPirh2

β-actin

WTPirh2-/-

Phosphoserine

15 p53

PhosphoSer 15 p53 -

Ubiquitin

170 __

100 __

130 __

72 __

55 __

79

Page 89: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

80

DISCUSSION

In this study, splenocytes and thymocytes from Pirh2-/- mice were used to

investigate the regulation of p53 by Pirh2. Several key observations arise from these

studies. First, the levels of p53 in unstressed splenocytes and thymocytes remain

unchanged in Pirh2-/- cells compared with wild-type cells. This observation would

suggest that Pirh2 does not have a critical role in regulating basal levels of p53 in

unstressed conditions. The second set of experiments tests whether Pirh2 can regulate

p53 after DNA damage. We determined that levels of p53 are modestly elevated and the

levels of phosphoserine 15 p53 are elevated in Pirh2-/- splenocytes and thymocytes after

DNA damage. The increased levels of total and phosphoserine 15 p53 has a functional

consequence as the rate of apoptosis was increased in Pirh2-/- cells as was the rate of

induction of p53-dependent pro-apoptotic target bax. These results suggest that Pirh2

may play a role in regulating p53 after DNA damage.

In the absence of Pirh2, the rate of p53-dependent apoptosis is increased along

with induction of bax. This observation fits with the model that Pirh2 regulates p53 after

DNA damage. However, in MCF-7 cells overexpressing Pirh2, p53-dependent cell cycle

arrest is not attenuated though levels of the cell cycle inhibitor p21 are reduced. This

discrepancy could be explained by several factors. In the MCF-7 cells, there may be a

low threshold of p21 activity required to produce cell cycle arrest or other

downregulation of other Pirh2 substrates may promote cell cycle progression. Also, we

measured a difference in the kinetics of apoptosis; if Pirh2 affects the kinetics of p53-

induction it may be difficult to measure the kinetics of p53-induced cell cycle arrest. It is

also possible that Pirh2 may regulate cell cycle arrest in specific cell types or under

Page 90: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

81

specific cell conditions. In the MCF-7 clones that overexpress Pirh2, phosphoserine 15

p53 protein levels and induction of p21 protein was reduced in response to adriamycin,

but induction Mdm2 protein was unaffected by the decreased phosphoserine 15 p53

levels (Fig 3.5). Further experiments with different cell types and different cell stresses

and additional p53 target genes should be performed to determine whether

overexpression of Pirh2 can affect the kinetics of p53-dependent apoptosis or p53-

dependent cell cycle arrest. We can also harvest fibroblasts from Pirh2+/+ and Pirh2-/-

mice to test various DNA damaging agents for p53-dependent target gene expression, and

to measure cell cycle arrest. In Pirh2-/- fibroblasts we would expect to see an increase in

p53-dependent cell cycle arrest target genes, and an increase in p53-dependent cell cycle

arrest.

Pirh2 was identified as a p53-regulated gene that targets p53 for ubiquitin-

mediated proteolysis. Leng et al. (2003) reported that Pirh2 directly binds to p53 and can

catalyze polyubiquitination of p53; moreover knock down of Pirh2 by antisense modestly

increased p53 dependent G1 cell cycle arrest (Leng et al., 2003). Transfection

experiments also show that Pirh2 only targets the tetrameric form of p53 while Mdm2 is

proposed to target all forms of p53 (Sheng et al., 2008). Our data shows that Pirh2 can

coimmunoprecipitate with phosphoserine 15 p53, and Pirh2 also promotes ubiquitination

of phosphoserine 15 p53, an activating modification on p53 after DNA damage (Fig 3.7)

(Chao et al., 2000a). Based on these observations, we propose that Pirh2 may regulate

the active form of p53. In contrast, the role of Mdm2 is to regulate the basal levels p53.

This is consistent with observations from the literature. Mdm2 knockout mice are

embryonic lethal, and this lethality can be rescued by deletion of p53 (Jones et al., 1995;

Page 91: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

82

Montes de Oca Luna et al., 1995). Also, a mouse model was made where a hypomorphic

Mdm2 allele was conditionally knocked into the thymus; the thymus in the hypomorphic

Mdm2 mouse was much smaller than the thymus in the Mdm2+/+ mouse under

unstressed conditions. In double mutant p53-/- Mdm2 hypomorphic mice, the thymus is

larger, and is comparable in size to the Mdm2+/+ thymus. These results suggest that

unrestrained p53 activity causes the atrophy of the thymus in the Mdm2 hypomorphic

mice, and that Mdm2 keeps p53 activity in check in unstressed conditions in the thymus

(Mendrysa et al., 2003; O'Leary et al., 2004). The Pirh2 knockout mice are viable and

develop normally, and in unstressed conditions show no difference in basal p53 level

when compared to the wild-type mice in the spleen and thymus. This would suggest that

Mdm2 is the main regulator of p53 under unstressed conditions. Its main role is to keep

p53 levels in check, as unrestrained p53 activity is detrimental to the organism – it would

cause massive apoptosis and cell cycle arrest. There is also significant evidence that

Mdm2 is dissociated from p53 after DNA damage, and as a result could not induce the

degradation of p53 (Maya et al., 2001; Shieh et al., 1997). For example, a time course of

coimmunoprecipitation of p53 and Mdm2 shows that the complex dissociates after DNA

damage (Shieh et al., 1997). In addition, ATM has been shown to phosphorylate Mdm2

on serine 395, and an aspartic acid phosphomimic on serine 395 on Mdm2 shows reduced

ability to degrade p53 in transfection experiments (Maya et al., 2001). The results of Fig

3.4 suggest that phosphorylation of serine 15 p53 is insufficient to prevent degradation of

p53 by Mdm2. Fig 3.5 shows that overexpression of Mdm2 does not prevent activation

of p53 and implies that Mdm2 is dissociated from p53 after DNA damage as levels of

p53, phosphoserine 15 p53 and p21 in Mdm2 overexpressing cells are similar to those

Page 92: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

83

from the vector control. We suggest that under conditions of DNA damage where Mdm2

does not degrade p53, Pirh2 degradation of active p53 can be observed. The role of Pirh2

in regulating p53 may be to fine-tune the DNA damage response.

Page 93: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

CHAPTER 4

Summary and Future Directions

84

Page 94: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

85

I. Summary of Findings: Identification of Pirh2 Interactors

The ubiquitin ligase Pirh2 has several known targets for degradation including

p53, HDAC1, p27/Kip1, SRβ and ε-COP (Abe et al., 2008; Hattori et al., 2007; Leng et

al., 2003; Logan et al., 2006; Maruyama et al., 2008). We used several dual purification

strategies, the TAP tag, a flag-calmodulin binding peptide tag, and a histidine-flag tag, to

identify proteins that interact with Pirh2 and planned to characterize the interactors as

cofactors or substrates. We also used a GST-pull down approach to identify interactors

under both normal and stress conditions. We had very little success with the two

approaches, with the exception of potentially identifying PKC, and have determined that

additional approaches to identify substrates and cofactors must be investigated.

Future Directions: Identification of Pirh2 Interactors

Identification of Pirh2 co-factors and substrates.

Additional approaches to identify substrates and cofactors for Pirh2 are required.

These strategies need to account for the weak and transient nature of the interaction

between Pirh2 and either substrate or cofactor. Pull downs, or coimmunoprecipitation

experiments may not identify specific interacting proteins since they could be lost in the

numerous washes. There are several alternative approaches to identify substrates for

Pirh2. The first is to fluorescently label cells from Pirh2+/+ and Pirh2-/- mice with

different fluorochromes. One would mix equal amounts of the protein lysates from both

Pirh2+/+ and Pirh2-/- mice, and separate the proteins using two dimensional gel

electrophoresis. The spots from the protein mixture could be visualized on the Typhoon

or an appropriate fluorescence reader and one would look for proteins that are highly

Page 95: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

86

expressed in Pirh2-/- cells. These proteins would be candidate substrates, and one would

need to confirm that they are direct substrates of Pirh2.

A second potential approach relies on advances in the mass spectrometry. This

approach would involve labelling of whole cell lysate with one of two tags cICAT

(cleavable Isotope-Coded Affinity Tags) or ITRAQ (Isobaric Tags for Relative and

Absolute Quantification). In these two labelling techniques, cICAT involves labelling of

cysteine residues with either a heavy or light isotope and a biotin tag. The biotin tag

causes problems in the mass spectrometry analysis and the cICAT tag involves acid

cleavage of the biotin tag. ITRAQ labelling techniques involves labelling of the N-

terminus and amino side chains with a set of amine reactive isobaric tags (Wu et al.,

2006). The two labelled samples are then mixed, trypsinized and analyzed by

multidimensional LC-MS/MS (the lysates would be separated by cation exchange and

C18 reverse-phase column chromatography) (Wu et al., 2006). To identify Pirh2

substrates, one would isolate cells from Pirh2+/+ and Pirh2-/- mice, and label the lysate.

For example, the Pirh2+/+ lysate would be labelled with the heavy isotope, and Pirh2-/-

lysate would be labelled with the light isotope. An equal mass of protein from labelled

Pirh2+/+ and Pirh2-/- splenocyte lysates would be mixed, trypsinized, and analyzed by

multi-dimensional LC-MS/MS. The proteins whose peptides have higher peaks from the

mass spectrometry spectrum from the light isotope sample would be considered candidate

substrates.

A third potential approach to identify substrates would be to perform an in vitro

ubiquitination assay using Pirh2 as the ligase and whole cell lysate as the substrate. One

would perform the ubiquitination assay using histidine tagged 7KR ubiquitin mutant.

Page 96: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

87

The ubiquitinated substrates can be purified using the histidine tag, and the substrates can

be eluted and then identified by mass spectrometry. The histidine tagged 7KR ubiquitin

is used because the histidine tag allows for purification of substrates under denaturing

conditions that will inactivate proteases and deubiquitinases, and the 7KR ubiquitin

reduces the amount of ubiquitin co-purified with the substrates. In the presence of excess

ubiquitin, substrates may not be identified using mass spectrometry; ubiquitin peptides

will be dominant, and will mask signals from other peptides (Yi Sheng, unpublished data,

personal communication).

A fourth possibility for identifying substrates uses protein arrays. In this approach,

proteins are spotted on an array and these serve as substrates in an in vitro ubiquitination

assay with Pirh2 as the E3 ubiquitin ligase and biotinylated ubiquitin. The protein array

is then washed with SDS to thoroughly remove any non-covalently bound ubiquitin. The

biotinylated ubiquitin is then incubated with a strepavidin conjugated to a fluorochrome.

Potential substrates for the ubiquitin ligase can be identified using a fluorescence imaging

detector (Andrews PS, personal communication, Keystone Conference 2009: The Many

Faces of Ubiquitin).

Each of these approaches has advantages and disadvantages. The main advantage

of the two dimensional gel electrophoresis and cICAT/ITRAQ labelling experiments are

that they rely on physiological conditions; however, the results may be caused by an

indirect mechanism. For example, if Pirh2 were to degrade a transcriptional activator, in

the absence of Pirh2 all the targets of that transcriptional activator would be

overexpressed and appear to be Pirh2 targets; hence their overexpression would be an

indirect result of the loss of Pirh2. Candidate substrates would have to be validated using

Page 97: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

88

in vitro studies and coimmunoprecipitation and colocalization studies. The third and

fourth approaches rely on in vitro ubiquitination assays that may give false positives. The

key difference in the third and fourth approach is how the substrates are identified. The

third approach uses mass spectrometry to identify the substrate; the sensitivity of mass

spectrometry is heavily dependent on the number of peptides present in the mixture, and

the presence of ubiquitin peptides may reduce the sensitivity of being able to identify the

protein by mass spectrometry analysis (Steen and Mann, 2004). This limitation of mass

spectrometry analysis is also a disadvantage in the cICAT and ITRAQ approach to

identifying substrates – the labelled peptides that show a difference between Pirh2+/+

cells and Pirh2-/- cells must be in high abundance to be successfully detected using mass

spectrometry. The protein chip approach is limited by the number of proteins on the chip,

and any post-translational modifications that may influence binding to Pirh2 since protein

chips contain unmodified proteins. For example, in the two dimensional gel

electrophoresis, cICAT/ITRAQ and the in vitro 7KR ubiquitination assay approaches to

substrate identification, one can treat the cells with drugs, serum starve or use many other

stresses that could influence the substrate specificity of Pirh2.

Identification of cofactors is more difficult. There are approximately fifty E2

ubiquitin conjugating enzymes in the human genome; the E2s often determines the

pattern of ubiquitin conjugates that are formed, and this may govern the biological

outcome of ubiquitination (Deshaies and Joazeiro, 2009). Each E3 ubiquitin ligase only

uses a subset of these E2s and identifying these E2 enzymes has been a challenge. In pull

down/mass spectrometry experiments, E2s are usually not identified due to the weak

interaction between the ubiquitin conjugating enzyme and the ligase (Deshaies and

Page 98: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

89

Joazeiro, 2009). The current method of identifying E2 –E3 partners is through screening

purified E2s to find ones that works best with the particular E3 ubiquitin ligase in an in

vitro experiment. This approach can be problematic for several reasons as there are only

a limited number of E2s that can be expressed, purified and stored as recombinant protein.

In addition, E2 and E3 enzymes that require post-translational modification to become

active would be excluded from these in vitro approaches (Deshaies and Joazeiro, 2009).

A physiological approach would be best to identify the proper E2-E3 pairs. Deshaies and

Joazeiro (2009) suggest an approach using in vivo crosslinking, and potentially

immunoprecipitation of the ubiquitin ligase and identification of the associated proteins

using mass spectrometry. This would identify proteins under physiological conditions,

and also account for post-translational modifications that may be required for activity or

binding of the E2-E3 partners.

The first E4 enzyme, UFD2, was discovered in yeast as a factor that binds to

ubiquitin and promotes chain elongation (Koegl et al., 1999). E4 enzymes were thought

to contain a U-box, such as UFD2 (Hoppe, 2005). However, not all E4 chain assembly

factors contain a U-box, and not all U-box proteins act as E4 ubiquitin chain assembly

factors; U-box proteins have been implicated as E3 ubiquitin ligases (Hatakeyama and

Nakayama, 2003). For example, p300 and Yin Yang 1 are a histone acetyl transferase

and a transcription factor, respectively; these proteins are E4 enzymes for p53 and do not

contain a U-box (Gronroos et al., 2004; Grossman et al., 2003; Sui et al., 2004).

Identification of E4 enzymes then becomes problematic since like substrates and E2

enzymes, they are weakly bound to the ubiquitin ligase. In addition, unlike E2 enzymes

that have already been identified and have a characteristic globular domain, a

Page 99: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

90

characteristic domain for E4 enzymes has not yet been determined (Pickart and Eddins,

2004). A similar method used to identify of E2 enzymes could be used to identify E4

enzymes – a yeast two hybrid approach and in vivo crosslinking-immunoprecipitation.

However, characterizing the interaction could be problematic as substrates would also be

identified in these experiments. Candidate E4 enzymes would not be substrates, and

potential ubiquitination activity would have to be tested in a ubiquitination assay.

Characterization of the interactors.

Interacting proteins can broadly be classified into two categories: substrates and

cofactors. To characterize substrates one would compare protein levels in the Pirh2+/+

cells and tissues with those isolated from the Pirh2-/- background using western blot

analysis. One would expect higher protein levels for the candidate substrate in the Pirh2

-/- background. In addition, one would perform in vitro ubiquitination assays using

purified proteins, or transfecting into cell lines. Once the candidate substrate was verified,

one would map the interaction domains on Pirh2 and on the substrate by GST-pull down

experiments. One would also want to determine what type of ubiquitin linkages Pirh2

catalyzes on the substrate. One could take two approaches. The first is an in vitro

approach using ubiquitin constructs where a single lysine residue has been mutated to

arginine to prevent chain elongation at that residue; one would cotransfect Pirh2, the

substrate, and the ubiquitin mutant into cells. One would then immunoprecipitate the

substrate and perform a western blot for ubiquitin. One would compare each of the seven

lysine to arginine ubiquitin mutants to the wild-type ubiquitin to determine if a single

residue is preferred to create polyubiquitin chains. One could also treat Pirh2+/+ and

Page 100: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

91

Pirh2-/- cells with proteasomal inhibitor and ubiquitin aldehyde (to inhibit

deubiquitinases), immunoprecipitate the substrate and compare the differences in

linkages found in the Pirh2+/+ cells relative to the Pirh2-/- cells. Pirh2 may not

necessarily target the substrate for ubiquitin-mediated proteolysis, but may change the

subcellular localization, or activity of the protein based on the ubiquitin linkage (Chen

and Sun, 2009). These experiments could determine whether Pirh2-mediated

ubiquitination is solely present to target substrates for degradation, or may have a more

complex role in affecting protein function in the absence of proteolysis.

Cofactors are likely to be identified in crosslinking/immunoprecipitation

experiments or yeast two hybrid experiments. They are also likely not to be found in the

substrate identification experiments. If ubiquitin conjugating enzymes are identified, the

important question then becomes identifying which substrates the E2 helps ubiquitinate.

Knockdown of the E2 in question using RNAi will help determine this as the Pirh2

substrates in question will increase. If one compares Pirh2-/- cells with control RNAi

with Pirh2+/+ cells with E2 RNAi and Pirh2-/- cells with RNAi, one can determine the

E2-Pirh2 substrates. Pirh2 substrates will be increased in all three experiments; since

each E2 will cooperate with several other ubiquitin ligases, targets of those ubiquitin

ligases will also be increased when treated with RNAi against the E2. However, the non-

specific targets will not be increased in the Pirh2-/- cells without RNAi against the E2.

For other cofactors, such as E4 enzymes, one can perform an in vitro ubiquitination assay

using GST-hPirh2 on total cell lysate. One can then titrate levels of GST-hPirh2 so that it

weakly catalyzes ubiquitination; by western blot analysis for ubiquitin, there should be a

faint smear near background levels. If a cofactor promotes Pirh2 activity, addition of this

Page 101: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

92

cofactor to the GST-hPirh2 ubiquitination reaction should increase the efficiency of the

reaction; i.e. using low levels of GST-hPirh2 with the cofactor should catalyze

polyubiquitination more efficiently. By western blot analysis polyubiquitination of the

cell lysate should be increased. One would also want to map the interaction domains on

the cofactor and Pirh2. Also, one would use RNAi against the cofactor to determine if it

preferentially increases polyubiquitination on specific Pirh2 substrates. RNAi against the

cofactor should increase the protein level of specific Pirh2 substrates, or may generally

increase the protein level of all substrates. Determining the cofactors that regulate Pirh2

expression may help determine which cell types and under which stress conditions Pirh2

activity may be important for the regulation of cellular processes.

Identification of the Pirh2 degron

The degron is the signal for a particular ubiquitin ligase to initiate degradation of

its substrate, and it is also where the ubiquitin ligase binds to the substrate (Deshaies and

Joazeiro, 2009). If the binding site for several Pirh2 substrates were known, one would

compare the substrates to look for sequence similarity in those binding regions. One

would then synthesize ten amino acid long peptides corresponding to the sequence

similarities between the Pirh2 binding sites. For example, if one found a fifteen peptide

long amino acid sequence that is similar among the substrates, one would synthesize five

peptides; the first peptide would correspond to amino acids one to ten, the second would

correspond to two to eleven, etc. One would spot these peptides onto a membrane, and

then perform a far western blot using Pirh2. One would determine which peptides Pirh2

binds to, and this would help us to identify the degron. To test the proposed degron, one

Page 102: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

93

would mutate the degron in a substrate, and determine if the mutant degron abolishes

Pirh2-mediated ubiquitination and proteolysis and binding. Once one has confirmed the

degron, one can then use this sequence to potentially predict Pirh2 substrates rather than

experimentally identify them.

II. Summary of Findings: Pirh2 Regulation of Phosphoserine 15 p53

The Benchimol lab has previously published that Pirh2 participates in a negative

feedback loop with p53; under stress conditions, p53 induces Pirh2 expression, and Pirh2

targets p53 for ubiquitin mediated proteolysis. To further characterize Pirh2 regulation of

p53, we used the Pirh2-/- mouse generated by the Hakem lab. Splenocytes and

thymocytes have similar levels of basal p53 in both Pirh2+/+ and Pirh2-/- mice

suggesting that Pirh2 is not a major regulator of p53 in unstressed conditions (Fig. 3.1

B,C). After IR, splenocytes and thymocytes from Pirh2-/- mice show an increase in the

level of p53 and an increase in the level of phosphoserine 15 p53. The relative amount of

phosphoserine 15 p53 is higher in Pirh2-/- cells compared with Pirh2+/+ after

normalization to total p53 suggesting that Pirh2 modulates the level of phosphoserine 15

p53 (Fig. 3.2B). The relatively higher level of phosphoserine 15 p53 in Pirh2-/-

splenocytes and thymocytes has functional consequences since the induction of the pro-

apoptotic p53-target gene bax and induction of apoptosis occurs faster in the Pirh2-/- cells

(Fig. 3.3). To determine if high levels of Pirh2 affect p53 induction, we took cells that

stably express ectopic Pirh2 and measured the levels of p53 and p21 after adriamycin

treatment. We determined that ectopic expression of Pirh2 reduces induction of

Page 103: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

94

phosphoserine 15 p53 and p21 after DNA damage (Fig. 3.5). Hence, the activation of

p53 is facilitated by Pirh2-depletion and impeded by Pirh2 overexpression.

We also noted that Pirh2 coimmunoprecipitates with phosphoserine 15 p53 and

that in the absence of Pirh2, there appears to be reduced polyubiquitination of

phosphoserine 15 p53 (Fig 3.7). These results suggest that Pirh2 directly regulates

phosphoserine p53. We propose a model where under unstressed conditions Mdm2 and

Pirh2 can both degrade p53, but Mdm2 is the main regulator of p53. After DNA damage,

Mdm2 and p53 both are phosphorylated and this aids in the dissociation of the

Mdm2/p53 complex; Mdm2 no longer regulates p53 after DNA damage and p53 levels

increase. Pirh2 then becomes the main regulator of p53, and modulates the p53 response

(Figure 4.1).

There are many ubiquitin ligases that target p53 for ubiquitin mediated proteolysis:

Mdm2, Pirh2, Cop1, Synoviolin, CARPs, TOPORs, Trim24 and ARF-BP1 (Allton et al.,

2009; Chen et al., 2005a; Dornan et al., 2004b; Honda et al., 1997; Leng et al., 2003;

Rajendra et al., 2004; Yamasaki et al., 2007; Yang et al., 2007). Based on several

findings in the literature, it is generally accepted that Mdm2 is a critical regulator of p53

activity in unstressed cells (Brooks and Gu, 2006; Momand et al., 2000). The role of the

other ubiquitin ligases remains to be determined, but there are some differences among

the ubiquitin ligases. For example, under conditions of stress both Mdm2 and Cop1 can

be phosphorylated by ATM, and are no longer able to target p53 for ubiquitination

(Dornan et al., 2006; Maya et al., 2001). In contrast, both Pirh2 and CARPs can target

p53 for ubiquitination after DNA damage. CARP1/2 targets phosphoserine 20 p53 for

degradation, a modified form of p53 that only appears after DNA damage as a result of

Page 104: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

A

B

Figure 4.1 Model for Pirh2 regulation of p53.

Under normal conditions, Mdm2 is the main regulator of p53. Pirh2 can regulate p53, but is a minor contributor (A). Under conditions of DNA damage, both p53 and Mdm2 are phosphorylated; these phosphorylation sites do not affect Pirh2, and it becomes a regulator of p53 (B).

95Mdm2

Mdm2

Pirh2

Pirh2

Ub

p53

p53

Ub

Ub

Ub

Ub

Ub

Ub

Ub

Ub

Ub

Ub

Ub

Ub

Ub

Ub

Ub

P

P

Page 105: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

96

phosphorylation by the double strand break activated kinase Chk2 (Kruse and Gu, 2009a;

Yang et al., 2007). Based on the preliminary observations outlined above, it appears that

each ubiquitin ligase may target a subset of p53, or may target p53 in a subset of

conditions or cell types. These factors have yet to be determined, but pose an interesting

challenge for the field.

Future Directions

Dissecting the Functional Redundancy of p53 Ubiquitin Ligases

The issue of functional redundancy among the many p53 ubiquitin ligases is a

complex problem. To being resolving the question, one would look at the tissue and

developmental expression of each ubiquitin ligase to see whether there are differences in

the pattern of expression. This could suggest that there may be tissue or developmental

specific regulation of p53. It may also be possible that each ubiquitin ligase regulates a

different post-translationally modified forms of p53, different oligomeric forms, different

isoforms of p53 or mutant p53. Different post-translational modifications on p53 could

be a response to the exposure to various stresses that would cause differing post-

translational modifications. The p53 protein has been found to have several isoforms, the

Δ133p53 and the Δ40p53 isoforms each contain an N-terminal deletion that eliminates

the Mdm2 binding domain (Bourdon et al., 2005). Presumably these isoforms are not

regulated by Mdm2 and may be targeted by one of the other ubiquitin ligases. Cofactors

could influence the binding of the ubiquitin ligases to p53, the stability of the ubiquitin

ligase or the ability of the ubiquitin ligase to promote ubiquitination; expression of the

cofactors can be influenced by cell type and stress response.

Page 106: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

97

Post-translational modifications on the E3 ubiquitin ligases can also influence

binding to p53, stability and may also influence the activity of the ubiquitin ligase. For

example phosphorylation of Mdm2 and Cop1 by ATM reduces their ability to target p53

for ubiquitination as well as phosphorylation of Pirh2 by calmodulin-dependent kinase II

decreases p53 ubiquitination (Dornan et al., 2006; Duan et al., 2007; Maya et al., 2001).

Post-translational modifications can be activating as neddylation of the cullin in the SCF

complex is required for ubiquitination activity (Saha and Deshaies, 2008). Post-

translational modifications may be stress-specific, and may be identified using

bioinformatics to predict phosphorylation sites or potentially by mass spectrometry.

There could also be an organism specific effect to regulating p53. Mdm2-/- mice

are embryonic lethal, and die by massive apoptosis preimplantation; the embryonic

lethality is fully rescued by deletion of p53 (Jones et al., 1995; Montes de Oca Luna et al.,

1995). In addition, amplification of Mdm2 has been found in tumours as a mechanism of

inactivating p53, and anti-cancer therapies are being developed to inhibit the Mdm2-p53

interaction (Onel and Cordon-Cardo, 2004; Vassilev et al., 2004). This finding strongly

supports that Mdm2 is a key regulator of p53 of in mice and humans. However, p53

orthologs are found in Drosophila Melanogaster and Caenorhabditis Elegans, but these

organisms do not have an Mdm2 ortholog (Brooks and Gu, 2006). An ortholog of one of

the other ubiquitin ligases may be important to regulate p53 in these organisms. Trim24

appears to be an important regulator of Drosophila p53 (Allton et al., 2009). Orthologs

of Pirh2 exist in Schizosaccharomyces pombe and Drosophila Melanogaster (Leng et al.,

2003).

Page 107: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

98

Non-Proteolytic Ubiquitination of p53

Two ubiquitin ligases have been identified that target p53 for non-proteolytic

ubiquitination. E4F1 catalyzes lysine 48 linked polyubiquitination of p53 on lysine 320.

This polyubiquitination occurs after DNA damage and promotes the cell cycle arrest

pathway (Le Cam et al., 2006). The ubiquitin ligase MSL2 catalyzes polyubiquitination

of p53 on lysines 351 and 357. MSL2 mediated polyubiquitination promotes cytoplasmic

translocation of p53 (Kruse and Gu, 2009b).

Lysine 48 linked polyubiquitination traditionally targets the substrate for

proteolysis (Xu et al., 2009). E4F1 mediated ubiquitination of p53 targets cell cycle

arrest promoters instead of promoting polyubiquitination (Le Cam et al., 2006). This

finding raises several questions: how does the E4F1 lysine 48 polyubiquitin chain evade

degradation, how does the polyubiquitin chain redirect p53 to cell cycle arrest promoters,

and whether E4F1 activity is required to induce cell cycle arrest.

MSL2 catalyzes polyubiquitination on lysines 351 and 357. The linkage that is

used to create these polyubiquitin chains is unknown and should to be determined (Kruse

and Gu, 2009b). In addition, how polyubiquitination of these lysines causes

relocalization of p53 to the cytoplasm should be determined. How MSL2 affects p53

biological activity also needs to be determined. MSL2 activity is not affected by DNA

damage. Key questions include whether high levels of MSL2 reduces the level of p53

transcriptional targets, or the amount of p53 bound to promoters. Also, p53 is usually a

nuclear protein and translocation to the mitochondria causes apoptosis (Kruse and Gu,

2009b; Mihara et al., 2003). How p53 translocates from the nucleus to the mitochondria

is still a question that needs to be resolved. Perhaps high levels of MSL2 may promote

Page 108: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

99

transcription independent, p53-dependent apoptosis after DNA damage by relocalizing

p53 to the cytoplasm where it can then reach the mitochondria. Confocal microscopy

could determine whether high levels of MSL2 promotes p53 translocation to the

mitochondria. In addition, apoptotic assays could help determine whether the ectopic

expression of MSL2 increases p53-dependent apoptosis.

Pirh2-Mediated Ubiquitination of p53

We have determined that Pirh2 can catalyze ubiquitination of phosphoserine 15

p53. It is possible that Pirh2 can ubiquitinate other post-translationally modified forms of

p53 such as phosphoserine 6, 9, 20, 33, 36, 37, 46 and/or phosphothreonine 18, 55, 81.

Several modifications on p53 occur throughout the protein; one would focus on

modifications that occur in the N-terminus because Pirh2 binds to p53 in the DNA

binding domain and the C-terminal oligomerization domain, and modifications in these

regions may help to dissociate the Pirh2-p53 complex (Leng et al., 2003; Sheng et al.,

2008). In addition, we have shown that phosphorylation of serine 15 on p53 does not

affect Pirh2-mediated degradation. The set of experiments performed to characterize

Pirh2-mediated ubiquitination of phosphoserine 15 p53 can be used to determine whether

Pirh2 can target other p53 phosphoforms. One would expect an increase in

phosphorylated p53 in Pirh2-/- cells, and a decrease in phosphorylated p53 in Pirh2

overexpressing cells. One would also perform two dimensional gel electrophoresis on

irradiated Pirh2+/+ and Pirh2-/- samples, and use western blot for total p53. If Pirh2

regulates phosphorylated p53, one would expect to see an accumulation of the acidic

(phosphorylated) forms of p53 in the Pirh2-/- irradiated cells. The two dimensional gel

Page 109: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

100

electrophoresis results could also determine whether Pirh2 regulates forms of p53 that

contain multiple phosphorylation sites. Should Pirh2 regulate other forms of activated

p53, it would support the hypothesis that Pirh2 may modulate p53 activation.

In addition, one can look at whether post-translational modifications in the DNA

binding domain and the tetramerization domain can inhibit ubiquitination or binding of

Pirh2 to p53. One would also determine whether post-translational modification of the

C-terminal lysines by acetylation, neddylation or methylation can inhibit ubiquitination or

binding of p53 by Pirh2. Understanding the conditions that determine when Pirh2

regulates p53 will contribute to our understanding of p53 ubiquitination and may begin to

resolve the issue of functional redundancy.

One could also determine whether Pirh2 regulates p53 under different stress

conditions. For example, one could use hypoxic conditions, ER stress, UV irradiation,

and nutritional stress to determine whether Pirh2 affects p53 levels. This can be done by

looking at total p53, post-translationally modified p53, and two dimensional gel

electrophoresis.

There are seven possible lysines on ubiquitin that can form polyubiquitin chains

(Xu et al., 2009). One would determine which lysines on ubiquitin that Pirh2 uses to

catalyzes polyubiquitin chains on p53. One would do this using seven mutant ubiquitin

construct where six lysines are mutated to arginine. One would peform an in vivo

ubiquitination assay using p53 as a substrate, Pirh2 as the ubiquitin ligase, and the mutant

ubiquitin. Laddering of p53 on a Western blot for p53 will only be evident if Pirh2 can

catalyze polyubiquitination using the available lysine on ubiquitin. The polyubiquitin

chain that is catalyzed on p53 could suggest that Pirh2 may target p53 under different

Page 110: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

101

conditions. For example, polyubiquitination through lysine 63 could suggest a role in

signalling or subcellular localization, or polyubiquitination through lysine 29 could

suggest a role in ERAD (Xu et al., 2009).

Knock down of several ubiquitin ligases by RNAi increases p53 levels and cell

cycle arrest and apoptosis (Chen et al., 2005a; Dornan et al., 2004b). It is possible that

the ubiquitin ligases may work synergistically to target p53 for ubiquitin-mediated

proteolysis. For example, Mdm2 binds to p53 in the N-terminal domain, and this binding

site does not overlap with the Pirh2 binding site in the oligomerization domain or the

DNA binding domain; it is possible that both ubiquitin ligases can simultaneously bind to

p53. Whether ubiquitin ligases act synergistically can be tested by cotrasfecting siRNAs

to two ubiquitin ligases and determining whether p53-dependent cell cycle arrest or

apoptosis are increased beyond the sum of the single transfections. The dual binding of

these ubiquitin ligases can also be confirmed by coimmunoprecipitation. Possible

synergism between the ubiquitin ligases may suggest that one of the ubiquitin ligases may

function as an E4 or cofactor for the ubiquitination reaction. A cofactor role for one of

the ubiquitin ligases could suggest a cooperative relationship between the ligases in

regulating p53.

III. Pirh2 and Cancer

Pirh2 has been found to be overexpressed in lung cancer, and the overexpression

of Pirh2 does not correlate with p53 mutation status (Duan et al., 2004). In contrast,

Mdm2 has been found to be overexpressed in osteosarcomas, and is a mechanism to

inactivate wild-type p53 by targeting it for degradation (Onel and Cordon-Cardo, 2004).

Page 111: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

102

This suggests that p53 may not be the key target for Pirh2-mediate ubiquitination in lung

cancer. In addition, Pirh2 overexpression has also been found in prostate cancer and head

and neck cancers. In the head and neck cancers, overexpression of Pirh2 correlates with

low expression of p27, a G1 cell cycle inhibitor. RNAi against Pirh2 in a head and neck

squamous cell carcinoma cell line increases the expression of p27 and increases the cell

doubling time. High expression of Pirh2 and low expression of p27 in the head and neck

cancers correlate with low survival rates compared with low expression of Pirh2 and high

expression of p27 respectively. This study classified patients into four groups: low Pirh2

and high 27, low Pirh2 and low p27, high Pirh2 and low p27, high Pirh2 and high p27.

The patients with low Pirh2 and high p27 have the best prognosis followed by high Pirh2

and high p27, low Pirh2 and low p27, and high Pirh2 and low p27 have the worst

prognosis (Shimada et al., 2009). Since patients with high Pirh2 and low p27 have a

worse prognosis than patients with low Pirh2 and low p27, these results suggest that

Pirh2 is not only regulating p27 in head and neck squamous cell carcinomas, but has

other targets that contribute to oncogenesis.

In prostate cancer, Pirh2 has been found to be overexpressed and high levels of

Pirh2 expression correlate with the presence of bone metastases. Pirh2 targets HDAC1 in

prostate cancers, HDAC1 is an androgen receptor corepressor. Expression of Pirh2 leads

to reduced levels of HDAC1 resulting in increased transcription of the androgen receptor

target gene prostate specific antigen (PSA) (Logan et al., 2006). In contrast, observations

made when Pirh2 was initially characterized as an ARNIP (androgen receptor N-terminal

interacting protein) suggest that Pirh2 does not affect androgen receptor transactivation.

Cotransfection of Pirh2 and the androgen receptor did not affect androgen receptor

Page 112: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

103

transactivation in luciferase assays (Beitel et al., 2002). Beitel et al. (2002) used monkey

cells and mouse cells (Cos-1, CV-1 and NSC-34) while Logan et al (2006) performed

their cotransfection experiments in human 293T cells. While Pirh2 is a highly conserved

protein, perhaps the region of Pirh2 that binds HDAC1 may not be conserved across the

species or the binding site for Pirh2 on HDAC1 is not conserved across the species

leading to the differences in results between the two groups (Beitel et al., 2002).

Overexpression of Pirh2 leads to increased transcription by the androgen receptor and

increased levels of PSA mRNA (Logan et al., 2006). However, when Pirh2 is transfected

into the androgen receptor prostate cancer cell line LNCaP, PSA mRNA levels are

increased while PSA protein levels are decreased since ε-COP is required for PSA

secretion; increased levels of Pirh2 leads to degradation of ε-COP (Maruyama et al.,

2008). PSA is a serine protease that is responsible for liquefying seminal fluid. High

levels of PSA are used as a marker of prostate cancer in the PSA test. High levels of PSA

have not been shown to affect oncogenesis; PSA is an androgen receptor responsive gene,

and high androgen receptor activity is linked to the early stages of prostate cancer

development (Kaarbo et al., 2007; Lilja et al., 2008). If overexpression of Pirh2 is an

early event in prostate cancer, this could hinder the effectiveness of the PSA test for

predicting prostate cancer. As well, these cancers are statistically more aggressive, and

early detection of these cancers could help survival (Logan et al., 2006). Pirh2 also

regulates HDAC1, and derepression of HDAC1 target genes may also contribute to the

progression of cancer. Experiments need to determine whether inhibiting Pirh2

degradation of HDAC1 can potentially be developed as a therapeutic option for prostate

cancer.

Page 113: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

104

Recent observations have suggested that Pirh2 could act as an oncogene or a

tumour suppressor gene. For example, Pirh2 is overexpressed in head and neck

squamous cell carcinomas and Pirh2 degradation of the p27 G1 cell cycle inhibitor

appears to contribute to tumorigenesis (Hattori et al., 2007; Shimada et al., 2009). Pirh2

degradation of ε-COP decreases PSA protein levels, and may prevent early detection of

prostate cancer (Maruyama et al., 2008). In these examples, Pirh2 would appear to act as

an oncogene. In contrast, Pirh2 degrades HDAC1; in breast cancer, overexpression of

HDAC1 in the breast cancer cell line MCF-7 causes increased cell proliferation and

colony formation (Kawai et al., 2003; Logan et al., 2006). These observations suggest

that Pirh2 has the potential to also mediate tumour suppression. Determining the

differences between cell types and what determines which substrates Pirh2 targets would

be valuable in understanding how Pirh2 functions as a ubiquitin ligase, and how

deregulation of Pirh2 contributes to cancer. Targeting Pirh2 activity in cancer may prove

to be a useful therapeutic target or potentially a prognostic marker.

Page 114: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

105

References

Abe, K., Hattori, T., Isobe, T., Kitagawa, K., Oda, T., Uchida, C., and Kitagawa, M. (2008). Pirh2 interacts with and ubiquitylates signal recognition particle receptor beta subunit. Biomed Res 29, 53-60. Allton, K., Jain, A. K., Herz, H. M., Tsai, W. W., Jung, S. Y., Qin, J., Bergmann, A., Johnson, R. L., and Barton, M. C. (2009). Trim24 targets endogenous p53 for degradation. Proc Natl Acad Sci U S A 106, 11612-11616. Araki, R., Fukumura, R., Fujimori, A., Taya, Y., Shiloh, Y., Kurimasa, A., Burma, S., Li, G. C., Chen, D. J., Sato, K., et al. (1999). Enhanced phosphorylation of p53 serine 18 following DNA damage in DNA-dependent protein kinase catalytic subunit-deficient cells. Cancer Res 59, 3543-3546. Artandi, S. E., and Attardi, L. D. (2005). Pathways connecting telomeres and p53 in senescence, apoptosis, and cancer. Biochem Biophys Res Commun 331, 881-890. Ashcroft, M., Kubbutat, M. H., and Vousden, K. H. (1999). Regulation of p53 function and stability by phosphorylation. Mol Cell Biol 19, 1751-1758. Banin, S., Moyal, L., Shieh, S., Taya, Y., Anderson, C. W., Chessa, L., Smorodinsky, N. I., Prives, C., Reiss, Y., Shiloh, Y., and Ziv, Y. (1998). Enhanced phosphorylation of p53 by ATM in response to DNA damage. Science 281, 1674-1677. Beitel, L. K., Elhaji, Y. A., Lumbroso, R., Wing, S. S., Panet-Raymond, V., Gottlieb, B., Pinsky, L., and Trifiro, M. A. (2002). Cloning and characterization of an androgen receptor N-terminal-interacting protein with ubiquitin-protein ligase activity. J Mol Endocrinol 29, 41-60. Ben David, Y., Prideaux, V. R., Chow, V., Benchimol, S., and Bernstein, A. (1988). Inactivation of the p53 oncogene by internal deletion or retroviral integration in erythroleukemic cell lines induced by Friend leukemia virus. Oncogene 3, 179-185. Bernardi, R., Scaglioni, P. P., Bergmann, S., Horn, H. F., Vousden, K. H., and Pandolfi, P. P. (2004). PML regulates p53 stability by sequestering Mdm2 to the nucleolus. Nat Cell Biol 6, 665-672. Bond, G. L., Hu, W., Bond, E. E., Robins, H., Lutzker, S. G., Arva, N. C., Bargonetti, J., Bartel, F., Taubert, H., Wuerl, P., et al. (2004). A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell 119, 591-602. Bourdon, J. C., Fernandes, K., Murray-Zmijewski, F., Liu, G., Diot, A., Xirodimas, D. P., Saville, M. K., and Lane, D. P. (2005). p53 isoforms can regulate p53 transcriptional activity. Genes Dev 19, 2122-2137.

Page 115: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

106

Bouvard, V., Zaitchouk, T., Vacher, M., Duthu, A., Canivet, M., Choisy-Rossi, C., Nieruchalski, M., and May, E. (2000). Tissue and cell-specific expression of the p53-target genes: bax, fas, mdm2 and waf1/p21, before and following ionising irradiation in mice. Oncogene 19, 649-660. Braun, B. C., Glickman, M., Kraft, R., Dahlmann, B., Kloetzel, P. M., Finley, D., and Schmidt, M. (1999). The base of the proteasome regulatory particle exhibits chaperone-like activity. Nat Cell Biol 1, 221-226. Brooks, C. L., and Gu, W. (2003). Ubiquitination, phosphorylation and acetylation: the molecular basis for p53 regulation. Curr Opin Cell Biol 15, 164-171. Brooks, C. L., and Gu, W. (2006). p53 ubiquitination: Mdm2 and beyond. Mol Cell 21, 307-315. Brugarolas, J., Chandrasekaran, C., Gordon, J. I., Beach, D., Jacks, T., and Hannon, G. J. (1995). Radiation-induced cell cycle arrest compromised by p21 deficiency. Nature 377, 552-557. Canman, C. E., Lim, D. S., Cimprich, K. A., Taya, Y., Tamai, K., Sakaguchi, K., Appella, E., Kastan, M. B., and Siliciano, J. D. (1998). Activation of the ATM kinase by ionizing radiation and phosphorylation of p53. Science 281, 1677-1679. Caspari, T. (2000). How to activate p53. Curr Biol 10, R315-317. Chan, T. A., Hwang, P. M., Hermeking, H., Kinzler, K. W., and Vogelstein, B. (2000). Cooperative effects of genes controlling the G(2)/M checkpoint. Genes Dev 14, 1584-1588. Chao, C., Hergenhahn, M., Kaeser, M. D., Wu, Z., Saito, S., Iggo, R., Hollstein, M., Appella, E., and Xu, Y. (2003). Cell type- and promoter-specific roles of Ser18 phosphorylation in regulating p53 responses. J Biol Chem 278, 41028-41033. Chao, C., Herr, D., Chun, J., and Xu, Y. (2006). Ser18 and 23 phosphorylation is required for p53-dependent apoptosis and tumor suppression. EMBO J 25, 2615-2622. Chao, C., Saito, S., Anderson, C. W., Appella, E., and Xu, Y. (2000a). Phosphorylation of murine p53 at ser-18 regulates the p53 responses to DNA damage. Proc Natl Acad Sci U S A 97, 11936-11941. Chao, C., Saito, S., Kang, J., Anderson, C. W., Appella, E., and Xu, Y. (2000b). p53 transcriptional activity is essential for p53-dependent apoptosis following DNA damage. EMBO J 19, 4967-4975.

Page 116: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

107

Chavez-Reyes, A., Parant, J. M., Amelse, L. L., de Oca Luna, R. M., Korsmeyer, S. J., and Lozano, G. (2003). Switching mechanisms of cell death in mdm2- and mdm4-null mice by deletion of p53 downstream targets. Cancer Res 63, 8664-8669. Chehab, N. H., Malikzay, A., Stavridi, E. S., and Halazonetis, T. D. (1999). Phosphorylation of Ser-20 mediates stabilization of human p53 in response to DNA damage. Proc Natl Acad Sci U S A 96, 13777-13782. Chen, D., Kon, N., Li, M., Zhang, W., Qin, J., and Gu, W. (2005a). ARF-BP1/Mule is a critical mediator of the ARF tumor suppressor. Cell 121, 1071-1083. Chen, M., Cortay, J. C., Logan, I. R., Sapountzi, V., Robson, C. N., and Gerlier, D. (2005b). Inhibition of ubiquitination and stabilization of human ubiquitin E3 ligase PIRH2 by measles virus phosphoprotein. J Virol 79, 11824-11836. Chen, Z. J., and Sun, L. J. (2009). Nonproteolytic functions of ubiquitin in cell signaling. Mol Cell 33, 275-286. Chiou, S. K., Rao, L., and White, E. (1994). Bcl-2 blocks p53-dependent apoptosis. Mol Cell Biol 14, 2556-2563. Chipuk, J. E., Bouchier-Hayes, L., Kuwana, T., Newmeyer, D. D., and Green, D. R. (2005). PUMA couples the nuclear and cytoplasmic proapoptotic function of p53. Science 309, 1732-1735. Chipuk, J. E., Kuwana, T., Bouchier-Hayes, L., Droin, N. M., Newmeyer, D. D., Schuler, M., and Green, D. R. (2004). Direct activation of Bax by p53 mediates mitochondrial membrane permeabilization and apoptosis. Science 303, 1010-1014. Christophorou, M. A., Ringshausen, I., Finch, A. J., Swigart, L. B., and Evan, G. I. (2006). The pathological response to DNA damage does not contribute to p53-mediated tumour suppression. Nature 443, 214-217. Chuikov, S., Kurash, J. K., Wilson, J. R., Xiao, B., Justin, N., Ivanov, G. S., McKinney, K., Tempst, P., Prives, C., Gamblin, S. J., et al. (2004). Regulation of p53 activity through lysine methylation. Nature 432, 353-360. Cummins, J. M., Rago, C., Kohli, M., Kinzler, K. W., Lengauer, C., and Vogelstein, B. (2004). Tumour suppression: disruption of HAUSP gene stabilizes p53. Nature 428, 1 p following 486. 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. Mol Cell Biol 24, 7654-7668.

Page 117: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

108

Damia, G., Filiberti, L., Vikhanskaya, F., Carrassa, L., Taya, Y., D'Incalci, M., and Broggini, M. (2001). Cisplatinum and taxol induce different patterns of p53 phosphorylation. Neoplasia 3, 10-16. Danovi, D., Meulmeester, E., Pasini, D., Migliorini, D., Capra, M., Frenk, R., de Graaf, P., Francoz, S., Gasparini, P., Gobbi, A., et al. (2004). Amplification of Mdmx (or Mdm4) directly contributes to tumor formation by inhibiting p53 tumor suppressor activity. Mol Cell Biol 24, 5835-5843. Das, S., Raj, L., Zhao, B., Kimura, Y., Bernstein, A., Aaronson, S. A., and Lee, S. W. (2007). Hzf Determines cell survival upon genotoxic stress by modulating p53 transactivation. Cell 130, 624-637. Davison, T. S., Nie, X., Ma, W., Lin, Y., Kay, C., Benchimol, S., and Arrowsmith, C. H. (2001). Structure and functionality of a designed p53 dimer. J Mol Biol 307, 605-617. de Stanchina, E., McCurrach, M. E., Zindy, F., Shieh, S. Y., Ferbeyre, G., Samuelson, A. V., Prives, C., Roussel, M. F., Sherr, C. J., and Lowe, S. W. (1998). E1A signaling to p53 involves the p19(ARF) tumor suppressor. Genes Dev 12, 2434-2442. Deng, C., Zhang, P., Harper, J. W., Elledge, S. J., and Leder, P. (1995). Mice lacking p21CIP1/WAF1 undergo normal development, but are defective in G1 checkpoint control. Cell 82, 675-684. Deshaies, R. J., and Joazeiro, C. A. (2009). RING domain E3 ubiquitin ligases. Annu Rev Biochem 78, 399-434. Dias, S. S., Milne, D. M., and Meek, D. W. (2006). c-Abl phosphorylates Hdm2 at tyrosine 276 in response to DNA damage and regulates interaction with ARF. Oncogene 25, 6666-6671. Dohoney, K. M., Guillerm, C., Whiteford, C., Elbi, C., Lambert, P. F., Hager, G. L., and Brady, J. N. (2004). Phosphorylation of p53 at serine 37 is important for transcriptional activity and regulation in response to DNA damage. Oncogene 23, 49-57. Doil, C., Mailand, N., Bekker-Jensen, S., Menard, P., Larsen, D. H., Pepperkok, R., Ellenberg, J., Panier, S., Durocher, D., Bartek, J., et al. (2009). RNF168 binds and amplifies ubiquitin conjugates on damaged chromosomes to allow accumulation of repair proteins. Cell 136, 435-446. Donehower, L. A., Harvey, M., Slagle, B. L., McArthur, M. J., Montgomery, C. A., Jr., Butel, J. S., and Bradley, A. (1992). Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours. Nature 356, 215-221.

Page 118: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

109

Dornan, D., Bheddah, S., Newton, K., Ince, W., Frantz, G. D., Dowd, P., Koeppen, H., Dixit, V. M., and French, D. M. (2004a). COP1, the negative regulator of p53, is overexpressed in breast and ovarian adenocarcinomas. Cancer Res 64, 7226-7230. Dornan, D., Shimizu, H., Mah, A., Dudhela, T., Eby, M., O'Rourke, K., Seshagiri, S., and Dixit, V. M. (2006). ATM engages autodegradation of the E3 ubiquitin ligase COP1 after DNA damage. Science 313, 1122-1126. Dornan, D., Wertz, I., Shimizu, H., Arnott, D., Frantz, G. D., Dowd, P., O'Rourke, K., Koeppen, H., and Dixit, V. M. (2004b). The ubiquitin ligase COP1 is a critical negative regulator of p53. Nature 429, 86-92. Duan, S., Yao, Z., Hou, D., Wu, Z., Zhu, W. G., and Wu, M. (2007). Phosphorylation of Pirh2 by calmodulin-dependent kinase II impairs its ability to ubiquitinate p53. EMBO J 26, 3062-3074. Duan, W., Gao, L., Druhan, L. J., Zhu, W. G., Morrison, C., Otterson, G. A., and Villalona-Calero, M. A. (2004). Expression of Pirh2, a newly identified ubiquitin protein ligase, in lung cancer. J Natl Cancer Inst 96, 1718-1721. Dumaz, N., and Meek, D. W. (1999). Serine15 phosphorylation stimulates p53 transactivation but does not directly influence interaction with HDM2. EMBO J 18, 7002-7010. Dumaz, N., Milne, D. M., and Meek, D. W. (1999). Protein kinase CK1 is a p53-threonine 18 kinase which requires prior phosphorylation of serine 15. FEBS Lett 463, 312-316. el-Deiry, W. S., Kern, S. E., Pietenpol, J. A., Kinzler, K. W., and Vogelstein, B. (1992). Definition of a consensus binding site for p53. Nat Genet 1, 45-49. el-Deiry, W. S., Tokino, T., Velculescu, V. E., Levy, D. B., Parsons, R., Trent, J. M., Lin, D., Mercer, W. E., Kinzler, K. W., and Vogelstein, B. (1993). WAF1, a potential mediator of p53 tumor suppression. Cell 75, 817-825. Eliyahu, D., Raz, A., Gruss, P., Givol, D., and Oren, M. (1984). Participation of p53 cellular tumour antigen in transformation of normal embryonic cells. Nature 312, 646-649. Fei, P., Bernhard, E. J., and El-Deiry, W. S. (2002). Tissue-specific induction of p53 targets in vivo. Cancer Res 62, 7316-7327. 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. J Biol Chem 279, 35510-35517.

Page 119: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

110

Feng, Z., Hu, W., de Stanchina, E., Teresky, A. K., Jin, S., Lowe, S., and Levine, A. J. (2007). The regulation of AMPK beta1, TSC2, and PTEN expression by p53: stress, cell and tissue specificity, and the role of these gene products in modulating the IGF-1-AKT-mTOR pathways. Cancer Res 67, 3043-3053. Finlay, C. A., Hinds, P. W., and Levine, A. J. (1989). The p53 proto-oncogene can act as a suppressor of transformation. Cell 57, 1083-1093. Flores, E. R., Tsai, K. Y., Crowley, D., Sengupta, S., Yang, A., McKeon, F., and Jacks, T. (2002). p63 and p73 are required for p53-dependent apoptosis in response to DNA damage. Nature 416, 560-564. Fridman, J. S., and Lowe, S. W. (2003). Control of apoptosis by p53. Oncogene 22, 9030-9040. Gingras, A. C., Aebersold, R., and Raught, B. (2005). Advances in protein complex analysis using mass spectrometry. J Physiol 563, 11-21. Groll, M., Ditzel, L., Lowe, J., Stock, D., Bochtler, M., Bartunik, H. D., and Huber, R. (1997). Structure of 20S proteasome from yeast at 2.4 A resolution. Nature 386, 463-471. Gronroos, E., Terentiev, A. A., Punga, T., and Ericsson, J. (2004). YY1 inhibits the activation of the p53 tumor suppressor in response to genotoxic stress. Proc Natl Acad Sci U S A 101, 12165-12170. Grossman, S. R., Deato, M. E., Brignone, C., Chan, H. M., Kung, A. L., Tagami, H., Nakatani, Y., and Livingston, D. M. (2003). Polyubiquitination of p53 by a ubiquitin ligase activity of p300. Science 300, 342-344. Guardavaccaro, D., Corrente, G., Covone, F., Micheli, L., D'Agnano, I., Starace, G., Caruso, M., and Tirone, F. (2000). Arrest of G(1)-S progression by the p53-inducible gene PC3 is Rb dependent and relies on the inhibition of cyclin D1 transcription. Mol Cell Biol 20, 1797-1815. Hanna, J., and Finley, D. (2007). A proteasome for all occasions. FEBS Lett 581, 2854-2861. Hanna, J., Hathaway, N. A., Tone, Y., Crosas, B., Elsasser, S., Kirkpatrick, D. S., Leggett, D. S., Gygi, S. P., King, R. W., and Finley, D. (2006). Deubiquitinating enzyme Ubp6 functions noncatalytically to delay proteasomal degradation. Cell 127, 99-111. Harper, J. W., Adami, G. R., Wei, N., Keyomarsi, K., and Elledge, S. J. (1993). The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell 75, 805-816.

Page 120: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

111

Hatakeyama, S., and Nakayama, K. I. (2003). U-box proteins as a new family of ubiquitin ligases. Biochem Biophys Res Commun 302, 635-645. Hattori, T., Isobe, T., Abe, K., Kikuchi, H., Kitagawa, K., Oda, T., Uchida, C., and Kitagawa, M. (2007). Pirh2 promotes ubiquitin-dependent degradation of the cyclin-dependent kinase inhibitor p27Kip1. Cancer Res 67, 10789-10795. Haupt, Y., Maya, R., Kazaz, A., and Oren, M. (1997). Mdm2 promotes the rapid degradation of p53. Nature 387, 296-299. He, L., He, X., Lim, L. P., de Stanchina, E., Xuan, Z., Liang, Y., Xue, W., Zender, L., Magnus, J., Ridzon, D., et al. (2007). A microRNA component of the p53 tumour suppressor network. Nature 447, 1130-1134. Herbig, U., Jobling, W. A., Chen, B. P., Chen, D. J., and Sedivy, J. M. (2004). Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Mol Cell 14, 501-513. Hermeking, H., Lengauer, C., Polyak, K., He, T. C., Zhang, L., Thiagalingam, S., Kinzler, K. W., and Vogelstein, B. (1997). 14-3-3 sigma is a p53-regulated inhibitor of G2/M progression. Mol Cell 1, 3-11. Hershko, A. (1996). Lessons from the discovery of the ubiquitin system. Trends Biochem Sci 21, 445-449. Hershko, A., and Ciechanover, A. (1998). The ubiquitin system. Annu Rev Biochem 67, 425-479. Hinds, P., Finlay, C., and Levine, A. J. (1989). Mutation is required to activate the p53 gene for cooperation with the ras oncogene and transformation. J Virol 63, 739-746. Ho, J. S., Ma, W., Mao, D. Y., and Benchimol, S. (2005). p53-Dependent transcriptional repression of c-myc is required for G1 cell cycle arrest. Mol Cell Biol 25, 7423-7431. Honda, R., Tanaka, H., and Yasuda, H. (1997). Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett 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. EMBO J 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. Hoppe, T. (2005). Multiubiquitylation by E4 enzymes: 'one size' doesn't fit all. Trends Biochem Sci 30, 183-187.

Page 121: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

112

Huang, J., Sengupta, R., Espejo, A. B., Lee, M. G., Dorsey, J. A., Richter, M., Opravil, S., Shiekhattar, R., Bedford, M. T., Jenuwein, T., and Berger, S. L. (2007). p53 is regulated by the lysine demethylase LSD1. Nature 449, 105-108. 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. EMBO J 21, 6236-6245. Iwakuma, T., and Lozano, G. (2003). MDM2, an introduction. Mol Cancer Res 1, 993-1000. Jackson, P. K., Eldridge, A. G., Freed, E., Furstenthal, L., Hsu, J. Y., Kaiser, B. K., and Reimann, J. D. (2000). The lore of the RINGs: substrate recognition and catalysis by ubiquitin ligases. Trends Cell Biol 10, 429-439. Jenkins, J. R., Rudge, K., and Currie, G. A. (1984). Cellular immortalization by a cDNA clone encoding the transformation-associated phosphoprotein p53. Nature 312, 651-654. Jin, A., Itahana, K., O'Keefe, K., and Zhang, Y. (2004). Inhibition of HDM2 and activation of p53 by ribosomal protein L23. Mol Cell Biol 24, 7669-7680. Johnson, P., Chung, S., and Benchimol, S. (1993). Growth suppression of Friend virus-transformed erythroleukemia cells by p53 protein is accompanied by hemoglobin production and is sensitive to erythropoietin. Mol Cell Biol 13, 1456-1463. 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. Kaarbo, M., Klokk, T. I., and Saatcioglu, F. (2007). Androgen signaling and its interactions with other signaling pathways in prostate cancer. Bioessays 29, 1227-1238. Kastan, M. B. (2007). Wild-type p53: tumors can't stand it. Cell 128, 837-840. Kawai, H., Li, H., Avraham, S., Jiang, S., and Avraham, H. K. (2003). Overexpression of histone deacetylase HDAC1 modulates breast cancer progression by negative regulation of estrogen receptor alpha. Int J Cancer 107, 353-358. Khosravi, R., Maya, R., Gottlieb, T., Oren, M., Shiloh, Y., and Shkedy, D. (1999). Rapid ATM-dependent phosphorylation of MDM2 precedes p53 accumulation in response to DNA damage. Proc Natl Acad Sci U S A 96, 14973-14977. Kirkpatrick, D. S., Denison, C., and Gygi, S. P. (2005). Weighing in on ubiquitin: the expanding role of mass-spectrometry-based proteomics. Nat Cell Biol 7, 750-757.

Page 122: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

113

Kisselev, A. F., Callard, A., and Goldberg, A. L. (2006). Importance of the different proteolytic sites of the proteasome and the efficacy of inhibitors varies with the protein substrate. J Biol Chem 281, 8582-8590. Koegl, M., Hoppe, T., Schlenker, S., Ulrich, H. D., Mayer, T. U., and Jentsch, S. (1999). A novel ubiquitination factor, E4, is involved in multiubiquitin chain assembly. Cell 96, 635-644. Koulich, E., Li, X., and DeMartino, G. N. (2008). Relative structural and functional roles of multiple deubiquitylating proteins associated with mammalian 26S proteasome. Mol Biol Cell 19, 1072-1082. Kovalenko, A., Chable-Bessia, C., Cantarella, G., Israel, A., Wallach, D., and Courtois, G. (2003). The tumour suppressor CYLD negatively regulates NF-kappaB signalling by deubiquitination. Nature 424, 801-805. Kruse, J. P., and Gu, W. (2009a). Modes of p53 regulation. Cell 137, 609-622. Kruse, J. P., and Gu, W. (2009b). MSL2 promotes Mdm2-independent cytoplasmic localization of p53. J Biol Chem 284, 3250-3263. Kubbutat, M. H., Jones, S. N., and Vousden, K. H. (1997). Regulation of p53 stability by Mdm2. Nature 387, 299-303. Kubbutat, M. H., Ludwig, R. L., Ashcroft, M., and Vousden, K. H. (1998). Regulation of Mdm2-directed degradation by the C terminus of p53. Mol Cell Biol 18, 5690-5698. Kurki, S., Peltonen, K., Latonen, L., Kiviharju, T. M., Ojala, P. M., Meek, D., and Laiho, M. (2004). Nucleolar protein NPM interacts with HDM2 and protects tumor suppressor protein p53 from HDM2-mediated degradation. Cancer Cell 5, 465-475. 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. Lam, Y. A., Lawson, T. G., Velayutham, M., Zweier, J. L., and Pickart, C. M. (2002). A proteasomal ATPase subunit recognizes the polyubiquitin degradation signal. Nature 416, 763-767. Lane, D. P., and Crawford, L. V. (1979). T antigen is bound to a host protein in SV40-transformed cells. Nature 278, 261-263. Lang, G. A., Iwakuma, T., Suh, Y. A., Liu, G., Rao, V. A., Parant, J. M., Valentin-Vega, Y. A., Terzian, T., Caldwell, L. C., Strong, L. C., et al. (2004). Gain of function of a p53 hot spot mutation in a mouse model of Li-Fraumeni syndrome. Cell 119, 861-872.

Page 123: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

114

Le Cam, L., Linares, L. K., Paul, C., Julien, E., Lacroix, M., Hatchi, E., Triboulet, R., Bossis, G., Shmueli, A., Rodriguez, M. S., et al. (2006). E4F1 is an atypical ubiquitin ligase that modulates p53 effector functions independently of degradation. Cell 127, 775-788. Leng, R. P., Lin, Y., Ma, W., Wu, H., Lemmers, B., Chung, S., Parant, J. M., Lozano, G., Hakem, R., and Benchimol, S. (2003). Pirh2, a p53-induced ubiquitin-protein ligase, promotes p53 degradation. Cell 112, 779-791. Leu, J. I., Dumont, P., Hafey, M., Murphy, M. E., and George, D. L. (2004). Mitochondrial p53 activates Bak and causes disruption of a Bak-Mcl1 complex. Nat Cell Biol 6, 443-450. Levine, A. J. (1997). p53, the cellular gatekeeper for growth and division. Cell 88, 323-331. Li, M., Brooks, C. L., Kon, N., and Gu, W. (2004). A dynamic role of HAUSP in the p53-Mdm2 pathway. Mol Cell 13, 879-886. Li, M., Chen, D., Shiloh, A., Luo, J., Nikolaev, A. Y., Qin, J., and Gu, W. (2002a). Deubiquitination of p53 by HAUSP is an important pathway for p53 stabilization. Nature 416, 648-653. Li, M., Luo, J., Brooks, C. L., and Gu, W. (2002b). Acetylation of p53 inhibits its ubiquitination by Mdm2. J Biol Chem 277, 50607-50611. Lilja, H., Ulmert, D., and Vickers, A. J. (2008). Prostate-specific antigen and prostate cancer: prediction, detection and monitoring. Nat Rev Cancer 8, 268-278. Lin, Y., and Benchimol, S. (1995). Cytokines inhibit p53-mediated apoptosis but not p53-mediated G1 arrest. Mol Cell Biol 15, 6045-6054. Lin, Y., Brown, L., Hedley, D. W., Barber, D. L., and Benchimol, S. (2002). The death-promoting activity of p53 can be inhibited by distinct signaling pathways. Blood 100, 3990-4000. Lin, Y., Ma, W., and Benchimol, S. (2000). Pidd, a new death-domain-containing protein, is induced by p53 and promotes apoptosis. Nat Genet 26, 122-127. Linzer, D. I., and Levine, A. J. (1979). Characterization of a 54K dalton cellular SV40 tumor antigen present in SV40-transformed cells and uninfected embryonal carcinoma cells. Cell 17, 43-52. Livingstone, L. R., White, A., Sprouse, J., Livanos, E., Jacks, T., and Tlsty, T. D. (1992). Altered cell cycle arrest and gene amplification potential accompany loss of wild-type p53. Cell 70, 923-935.

Page 124: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

115

Logan, I. R., Gaughan, L., McCracken, S. R., Sapountzi, V., Leung, H. Y., and Robson, C. N. (2006). Human PIRH2 enhances androgen receptor signaling through inhibition of histone deacetylase 1 and is overexpressed in prostate cancer. Mol Cell Biol 26, 6502-6510. Logan, I. R., Sapountzi, V., Gaughan, L., Neal, D. E., and Robson, C. N. (2004). Control of human PIRH2 protein stability: involvement of TIP60 and the proteosome. J Biol Chem 279, 11696-11704. Lu, X. (2005). p53: a heavily dictated dictator of life and death. Curr Opin Genet Dev 15, 27-33. Lu, X., Ma, O., Nguyen, T. A., Jones, S. N., Oren, M., and Donehower, L. A. (2007). The Wip1 Phosphatase acts as a gatekeeper in the p53-Mdm2 autoregulatory loop. Cancer Cell 12, 342-354. MacLachlan, T. K., Takimoto, R., and El-Deiry, W. S. (2002). BRCA1 directs a selective p53-dependent transcriptional response towards growth arrest and DNA repair targets. Mol Cell Biol 22, 4280-4292. MacPherson, D., Kim, J., Kim, T., Rhee, B. K., Van Oostrom, C. T., DiTullio, R. A., Venere, M., Halazonetis, T. D., Bronson, R., De Vries, A., et al. (2004). Defective apoptosis and B-cell lymphomas in mice with p53 point mutation at Ser 23. EMBO J 23, 3689-3699. Maheswaran, S., Englert, C., Bennett, P., Heinrich, G., and Haber, D. A. (1995). The WT1 gene product stabilizes p53 and inhibits p53-mediated apoptosis. Genes Dev 9, 2143-2156. Maiguel, D. A., Jones, L., Chakravarty, D., Yang, C., and Carrier, F. (2004). Nucleophosmin sets a threshold for p53 response to UV radiation. Mol Cell Biol 24, 3703-3711. Malkin, D., Li, F. P., Strong, L. C., Fraumeni, J. F., Jr., Nelson, C. E., Kim, D. H., Kassel, J., Gryka, M. A., Bischoff, F. Z., Tainsky, M. A., and et al. (1990). Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science 250, 1233-1238. Marchenko, N. D., Wolff, S., Erster, S., Becker, K., and Moll, U. M. (2007). Monoubiquitylation promotes mitochondrial p53 translocation. EMBO J 26, 923-934. Martins, C. P., Brown-Swigart, L., and Evan, G. I. (2006). Modeling the therapeutic efficacy of p53 restoration in tumors. Cell 127, 1323-1334.

Page 125: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

116

Maruyama, S., Miyajima, N., Bohgaki, M., Tsukiyama, T., Shigemura, M., Nonomura, K., and Hatakeyama, S. (2008). Ubiquitylation of epsilon-COP by PIRH2 and regulation of the secretion of PSA. Mol Cell Biochem 307, 73-82. Maya, R., Balass, M., Kim, S. T., Shkedy, D., Leal, J. F., Shifman, O., Moas, M., Buschmann, T., Ronai, Z., Shiloh, Y., et al. (2001). ATM-dependent phosphorylation of Mdm2 on serine 395: role in p53 activation by DNA damage. Genes Dev 15, 1067-1077. Mendrysa, S. M., McElwee, M. K., Michalowski, J., O'Leary, K. A., Young, K. M., and Perry, M. E. (2003). mdm2 Is critical for inhibition of p53 during lymphopoiesis and the response to ionizing irradiation. Mol Cell Biol 23, 462-472. Meulmeester, E., Pereg, Y., Shiloh, Y., and Jochemsen, A. G. (2005). ATM-mediated phosphorylations inhibit Mdmx/Mdm2 stabilization by HAUSP in favor of p53 activation. Cell Cycle 4, 1166-1170. Michael, D., and Oren, M. (2003). The p53-Mdm2 module and the ubiquitin system. Semin Cancer Biol 13, 49-58. Michalovitz, D., Halevy, O., and Oren, M. (1990). Conditional inhibition of transformation and of cell proliferation by a temperature-sensitive mutant of p53. Cell 62, 671-680. 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. Mol Cell 11, 577-590. Minsky, N., Shema, E., Field, Y., Schuster, M., Segal, E., and Oren, M. (2008). Monoubiquitinated H2B is associated with the transcribed region of highly expressed genes in human cells. Nat Cell Biol 10, 483-488. Momand, J., Wu, H. H., and Dasgupta, G. (2000). MDM2--master regulator of the p53 tumor suppressor protein. Gene 242, 15-29. 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. Mowat, M., Cheng, A., Kimura, N., Bernstein, A., and Benchimol, S. (1985). Rearrangements of the cellular p53 gene in erythroleukaemic cells transformed by Friend virus. Nature 314, 633-636. Mukhopadhyay, D., and Riezman, H. (2007). Proteasome-independent functions of ubiquitin in endocytosis and signaling. Science 315, 201-205.

Page 126: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

117

Munroe, D. G., Peacock, J. W., and Benchimol, S. (1990). Inactivation of the cellular p53 gene is a common feature of Friend virus-induced erythroleukemia: relationship of inactivation to dominant transforming alleles. Mol Cell Biol 10, 3307-3313. Murata, S., Yashiroda, H., and Tanaka, K. (2009). Molecular mechanisms of proteasome assembly. Nat Rev Mol Cell Biol 10, 104-115. Nahle, Z., Polakoff, J., Davuluri, R. V., McCurrach, M. E., Jacobson, M. D., Narita, M., Zhang, M. Q., Lazebnik, Y., Bar-Sagi, D., and Lowe, S. W. (2002). Direct coupling of the cell cycle and cell death machinery by E2F. Nat Cell Biol 4, 859-864. Nakamura, S., Roth, J. A., and Mukhopadhyay, T. (2000). Multiple lysine mutations in the C-terminal domain of p53 interfere with MDM2-dependent protein degradation and ubiquitination. Mol Cell Biol 20, 9391-9398. Nakano, K., and Vousden, K. H. (2001). PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell 7, 683-694. Newton, K., Matsumoto, M. L., Wertz, I. E., Kirkpatrick, D. S., Lill, J. R., Tan, J., Dugger, D., Gordon, N., Sidhu, S. S., Fellouse, F. A., et al. (2008). Ubiquitin chain editing revealed by polyubiquitin linkage-specific antibodies. Cell 134, 668-678. Nijman, S. M., Huang, T. T., Dirac, A. M., Brummelkamp, T. R., Kerkhoven, R. M., D'Andrea, A. D., and Bernards, R. (2005). The deubiquitinating enzyme USP1 regulates the Fanconi anemia pathway. Mol Cell 17, 331-339. Nikolaev, A. Y., Li, M., Puskas, N., Qin, J., and Gu, W. (2003). Parc: a cytoplasmic anchor for p53. Cell 112, 29-40. O'Leary, K. A., Mendrysa, S. M., Vaccaro, A., and Perry, M. E. (2004). Mdm2 regulates p53 independently of p19(ARF) in homeostatic tissues. Mol Cell Biol 24, 186-191. Oda, E., Ohki, R., Murasawa, H., Nemoto, J., Shibue, T., Yamashita, T., Tokino, T., Taniguchi, T., and Tanaka, N. (2000a). Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 288, 1053-1058. Oda, K., Arakawa, H., Tanaka, T., Matsuda, K., Tanikawa, C., Mori, T., Nishimori, H., Tamai, K., Tokino, T., Nakamura, Y., and Taya, Y. (2000b). p53AIP1, a potential mediator of p53-dependent apoptosis, and its regulation by Ser-46-phosphorylated p53. Cell 102, 849-862. Ohki, R., Nemoto, J., Murasawa, H., Oda, E., Inazawa, J., Tanaka, N., and Taniguchi, T. (2000). Reprimo, a new candidate mediator of the p53-mediated cell cycle arrest at the G2 phase. J Biol Chem 275, 22627-22630.

Page 127: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

118

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. Olive, K. P., Tuveson, D. A., Ruhe, Z. C., Yin, B., Willis, N. A., Bronson, R. T., Crowley, D., and Jacks, T. (2004). Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome. Cell 119, 847-860. Onel, K., and Cordon-Cardo, C. (2004). MDM2 and prognosis. Mol Cancer Res 2, 1-8. Palmero, I., Pantoja, C., and Serrano, M. (1998). p19ARF links the tumour suppressor p53 to Ras. Nature 395, 125-126. Parada, L. F., Land, H., Weinberg, R. A., Wolf, D., and Rotter, V. (1984). Cooperation between gene encoding p53 tumour antigen and ras in cellular transformation. Nature 312, 649-651. Parant, J., Chavez-Reyes, A., Little, N. A., Yan, W., Reinke, V., Jochemsen, A. G., and Lozano, G. (2001). Rescue of embryonic lethality in Mdm4-null mice by loss of Trp53 suggests a nonoverlapping pathway with MDM2 to regulate p53. Nat Genet 29, 92-95. Pickart, C. M. (2001). Mechanisms underlying ubiquitination. Annu Rev Biochem 70, 503-533. Pickart, C. M. (2004). Back to the future with ubiquitin. Cell 116, 181-190. Pickart, C. M., and Eddins, M. J. (2004). Ubiquitin: structures, functions, mechanisms. Biochim Biophys Acta 1695, 55-72. Pomerantz, J., Schreiber-Agus, N., Liegeois, N. J., Silverman, A., Alland, L., Chin, L., Potes, J., Chen, K., Orlow, I., Lee, H. W., et al. (1998). The Ink4a tumor suppressor gene product, p19Arf, interacts with MDM2 and neutralizes MDM2's inhibition of p53. Cell 92, 713-723. Puig, O., Caspary, F., Rigaut, G., Rutz, B., Bouveret, E., Bragado-Nilsson, E., Wilm, M., and Seraphin, B. (2001). The tandem affinity purification (TAP) method: a general procedure of protein complex purification. Methods 24, 218-229. Rajendra, R., Malegaonkar, D., Pungaliya, P., Marshall, H., Rasheed, Z., Brownell, J., Liu, L. F., Lutzker, S., Saleem, A., and Rubin, E. H. (2004). Topors functions as an E3 ubiquitin ligase with specific E2 enzymes and ubiquitinates p53. J Biol Chem 279, 36440-36444. Rao, H., and Sastry, A. (2002). Recognition of specific ubiquitin conjugates is important for the proteolytic functions of the ubiquitin-associated domain proteins Dsk2 and Rad23. J Biol Chem 277, 11691-11695.

Page 128: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

119

Resnick-Silverman, L., St Clair, S., Maurer, M., Zhao, K., and Manfredi, J. J. (1998). Identification of a novel class of genomic DNA-binding sites suggests a mechanism for selectivity in target gene activation by the tumor suppressor protein p53. Genes Dev 12, 2102-2107. Rigaut, G., Shevchenko, A., Rutz, B., Wilm, M., Mann, M., and Seraphin, B. (1999). A generic protein purification method for protein complex characterization and proteome exploration. Nat Biotechnol 17, 1030-1032. Riley, T., Sontag, E., Chen, P., and Levine, A. (2008). Transcriptional control of human p53-regulated genes. Nat Rev Mol Cell Biol 9, 402-412. Rodriguez, M. S., Desterro, J. M., Lain, S., Lane, D. P., and Hay, R. T. (2000). Multiple C-terminal lysine residues target p53 for ubiquitin-proteasome-mediated degradation. Mol Cell Biol 20, 8458-8467. Rodriguez, M. S., Desterro, J. M., Lain, S., Midgley, C. A., Lane, D. P., and Hay, R. T. (1999). SUMO-1 modification activates the transcriptional response of p53. EMBO J 18, 6455-6461. Roth, J. A. (2006). Adenovirus p53 gene therapy. Expert Opin Biol Ther 6, 55-61. Rouault, J. P., Falette, N., Guehenneux, F., Guillot, C., Rimokh, R., Wang, Q., Berthet, C., Moyret-Lalle, C., Savatier, P., Pain, B., et al. (1996). Identification of BTG2, an antiproliferative p53-dependent component of the DNA damage cellular response pathway. Nat Genet 14, 482-486. Sabbatini, P., and McCormick, F. (1999). Phosphoinositide 3-OH kinase (PI3K) and PKB/Akt delay the onset of p53-mediated, transcriptionally dependent apoptosis. J Biol Chem 274, 24263-24269. Sablina, A. A., Budanov, A. V., Ilyinskaya, G. V., Agapova, L. S., Kravchenko, J. E., and Chumakov, P. M. (2005). The antioxidant function of the p53 tumor suppressor. Nat Med 11, 1306-1313. Saha, A., and Deshaies, R. J. (2008). Multimodal activation of the ubiquitin ligase SCF by Nedd8 conjugation. Mol Cell 32, 21-31. Saito, S., Yamaguchi, H., Higashimoto, Y., Chao, C., Xu, Y., Fornace, A. J., Jr., Appella, E., and Anderson, C. W. (2003). Phosphorylation site interdependence of human p53 post-translational modifications in response to stress. J Biol Chem 278, 37536-37544. Samuels-Lev, Y., O'Connor, D. J., Bergamaschi, D., Trigiante, G., Hsieh, J. K., Zhong, S., Campargue, I., Naumovski, L., Crook, T., and Lu, X. (2001). ASPP proteins specifically stimulate the apoptotic function of p53. Mol Cell 8, 781-794.

Page 129: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

120

Schmitt, C. A., Fridman, J. S., Yang, M., Baranov, E., Hoffman, R. M., and Lowe, S. W. (2002). Dissecting p53 tumor suppressor functions in vivo. Cancer Cell 1, 289-298. Schoenfeld, A. R., Apgar, S., Dolios, G., Wang, R., and Aaronson, S. A. (2004). BRCA2 is ubiquitinated in vivo and interacts with USP11, a deubiquitinating enzyme that exhibits prosurvival function in the cellular response to DNA damage. Mol Cell Biol 24, 7444-7455. Schott, A. F., Apel, I. J., Nunez, G., and Clarke, M. F. (1995). Bcl-XL protects cancer cells from p53-mediated apoptosis. Oncogene 11, 1389-1394. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D., and Lowe, S. W. (1997). Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593-602. Sheng, Y., Laister, R. C., Lemak, A., Wu, B., Tai, E., Duan, S., Lukin, J., Sunnerhagen, M., Srisailam, S., Karra, M., et al. (2008). Molecular basis of Pirh2-mediated p53 ubiquitylation. Nat Struct Mol Biol 15, 1334-1342. Sheng, Y., Saridakis, V., Sarkari, F., Duan, S., Wu, T., Arrowsmith, C. H., and Frappier, L. (2006). Molecular recognition of p53 and MDM2 by USP7/HAUSP. Nat Struct Mol Biol 13, 285-291. Shevchenko, A., Wilm, M., Vorm, O., and Mann, M. (1996). Mass spectrometric sequencing of proteins silver-stained polyacrylamide gels. Anal Chem 68, 850-858. Shieh, S. Y., Ikeda, M., Taya, Y., and Prives, C. (1997). DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell 91, 325-334. Shikama, N., Lee, C. W., France, S., Delavaine, L., Lyon, J., Krstic-Demonacos, M., and La Thangue, N. B. (1999). A novel cofactor for p300 that regulates the p53 response. Mol Cell 4, 365-376. Shimada, M., Kitagawa, K., Dobashi, Y., Isobe, T., Hattori, T., Uchida, C., Abe, K., Kotake, Y., Oda, T., Suzuki, H., et al. (2009). High expression of Pirh2, an E3 ligase for p27, is associated with low expression of p27 and poor prognosis in head and neck cancers. Cancer Sci 100, 866-872. Sionov, R. V., and Haupt, Y. (1999). The cellular response to p53: the decision between life and death. Oncogene 18, 6145-6157. Soengas, M. S., Alarcon, R. M., Yoshida, H., Giaccia, A. J., Hakem, R., Mak, T. W., and Lowe, S. W. (1999). Apaf-1 and caspase-9 in p53-dependent apoptosis and tumor inhibition. Science 284, 156-159.

Page 130: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

121

Spence, J., Sadis, S., Haas, A. L., and Finley, D. (1995). A ubiquitin mutant with specific defects in DNA repair and multiubiquitination. Mol Cell Biol 15, 1265-1273. Steen, H., and Mann, M. (2004). The ABC's (and XYZ's) of peptide sequencing. Nat Rev Mol Cell Biol 5, 699-711. Sui, G., Affar el, B., Shi, Y., Brignone, C., Wall, N. R., Yin, P., Donohoe, M., Luke, M. P., Calvo, D., and Grossman, S. R. (2004). Yin Yang 1 is a negative regulator of p53. Cell 117, 859-872. Tang, Y., Luo, J., Zhang, W., and Gu, W. (2006). Tip60-dependent acetylation of p53 modulates the decision between cell-cycle arrest and apoptosis. Mol Cell 24, 827-839. Tang, Y., Zhao, W., Chen, Y., Zhao, Y., and Gu, W. (2008). Acetylation is indispensable for p53 activation. Cell 133, 612-626. Toledo, F., Krummel, K. A., Lee, C. J., Liu, C. W., Rodewald, L. W., Tang, M., and Wahl, G. M. (2006). A mouse p53 mutant lacking the proline-rich domain rescues Mdm4 deficiency and provides insight into the Mdm2-Mdm4-p53 regulatory network. Cancer Cell 9, 273-285. Toledo, F., and Wahl, G. M. (2006). Regulating the p53 pathway: in vitro hypotheses, in vivo veritas. Nat Rev Cancer 6, 909-923. Trompouki, E., Hatzivassiliou, E., Tsichritzis, T., Farmer, H., Ashworth, A., and Mosialos, G. (2003). CYLD is a deubiquitinating enzyme that negatively regulates NF-kappaB activation by TNFR family members. Nature 424, 793-796. Unger, T., Juven-Gershon, T., Moallem, E., Berger, M., Vogt Sionov, R., Lozano, G., Oren, M., and Haupt, Y. (1999a). Critical role for Ser20 of human p53 in the negative regulation of p53 by Mdm2. EMBO J 18, 1805-1814. Unger, T., Sionov, R. V., Moallem, E., Yee, C. L., Howley, P. M., Oren, M., and Haupt, Y. (1999b). Mutations in serines 15 and 20 of human p53 impair its apoptotic activity. Oncogene 18, 3205-3212. Vassilev, L. T., Vu, B. T., Graves, B., Carvajal, D., Podlaski, F., Filipovic, Z., Kong, N., Kammlott, U., Lukacs, C., Klein, C., et al. (2004). In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 303, 844-848. 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 131: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

122

Vaziri, H., Squire, J. A., Pandita, T. K., Bradley, G., Kuba, R. M., Zhang, H., Gulyas, S., Hill, R. P., Nolan, G. P., and Benchimol, S. (1999). Analysis of genomic integrity and p53-dependent G1 checkpoint in telomerase-induced extended-life-span human fibroblasts. Mol Cell Biol 19, 2373-2379. Ventii, K. H., and Wilkinson, K. D. (2008). Protein partners of deubiquitinating enzymes. Biochem J 414, 161-175. Ventura, A., Kirsch, D. G., McLaughlin, M. E., Tuveson, D. A., Grimm, J., Lintault, L., Newman, J., Reczek, E. E., Weissleder, R., and Jacks, T. (2007). Restoration of p53 function leads to tumour regression in vivo. Nature 445, 661-665. Vousden, K. H., and Lu, X. (2002). Live or let die: the cell's response to p53. Nat Rev Cancer 2, 594-604. 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. Proc Natl Acad Sci U S A 96, 3706-3711. Weber, J. D., and Zambetti, G. P. (2003). Renewing the debate over the p53 apoptotic response. Cell Death Differ 10, 409-412. Wertz, I. E., O'Rourke, K. M., Zhou, H., Eby, M., Aravind, L., Seshagiri, S., Wu, P., Wiesmann, C., Baker, R., Boone, D. L., et al. (2004). De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling. Nature 430, 694-699. Wu, W. S., Heinrichs, S., Xu, D., Garrison, S. P., Zambetti, G. P., Adams, J. M., and Look, A. T. (2005). Slug antagonizes p53-mediated apoptosis of hematopoietic progenitors by repressing puma. Cell 123, 641-653. Wu, W. W., Wang, G., Baek, S. J., and Shen, R. F. (2006). Comparative study of three proteomic quantitative methods, DIGE, cICAT, and iTRAQ, using 2D gel- or LC-MALDI TOF/TOF. J Proteome Res 5, 651-658. Wu, X., Bayle, J. H., Olson, D., and Levine, A. J. (1993). The p53-mdm-2 autoregulatory feedback loop. Genes Dev 7, 1126-1132. Xirodimas, D. P., Saville, M. K., Bourdon, J. C., Hay, R. T., and Lane, D. P. (2004). Mdm2-mediated NEDD8 conjugation of p53 inhibits its transcriptional activity. Cell 118, 83-97. Xu, P., Duong, D. M., Seyfried, N. T., Cheng, D., Xie, Y., Robert, J., Rush, J., Hochstrasser, M., Finley, D., and Peng, J. (2009). Quantitative proteomics reveals the function of unconventional ubiquitin chains in proteasomal degradation. Cell 137, 133-145.

Page 132: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

123

Xue, W., Zender, L., Miething, C., Dickins, R. A., Hernando, E., Krizhanovsky, V., Cordon-Cardo, C., and Lowe, S. W. (2007). Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445, 656-660. Yamasaki, S., Yagishita, N., Sasaki, T., Nakazawa, M., Kato, Y., Yamadera, T., Bae, E., Toriyama, S., Ikeda, R., Zhang, L., et al. (2007). Cytoplasmic destruction of p53 by the endoplasmic reticulum-resident ubiquitin ligase 'Synoviolin'. EMBO J 26, 113-122. Yang, H. Y., Wen, Y. Y., Chen, C. H., Lozano, G., and Lee, M. H. (2003). 14-3-3 sigma positively regulates p53 and suppresses tumor growth. Mol Cell Biol 23, 7096-7107. Yang, W., Rozan, L. M., McDonald, E. R., 3rd, Navaraj, A., Liu, J. J., Matthew, E. M., Wang, W., Dicker, D. T., and El-Deiry, W. S. (2007). CARPs are ubiquitin ligases that promote MDM2-independent p53 and phospho-p53ser20 degradation. J Biol Chem 282, 3273-3281. Yao, T., and Cohen, R. E. (2002). A cryptic protease couples deubiquitination and degradation by the proteasome. Nature 419, 403-407. Yin, C., Knudson, C. M., Korsmeyer, S. J., and Van Dyke, T. (1997). Bax suppresses tumorigenesis and stimulates apoptosis in vivo. Nature 385, 637-640. Yin, Y., Tainsky, M. A., Bischoff, F. Z., Strong, L. C., and Wahl, G. M. (1992). Wild-type p53 restores cell cycle control and inhibits gene amplification in cells with mutant p53 alleles. Cell 70, 937-948. Zeghouf, M., Li, J., Butland, G., Borkowska, A., Canadien, V., Richards, D., Beattie, B., Emili, A., and Greenblatt, J. F. (2004). Sequential Peptide Affinity (SPA) system for the identification of mammalian and bacterial protein complexes. J Proteome Res 3, 463-468. Zhang, H., Somasundaram, K., Peng, Y., Tian, H., Bi, D., Weber, B. L., and El-Deiry, W. S. (1998). BRCA1 physically associates with p53 and stimulates its transcriptional activity. Oncogene 16, 1713-1721. Zhang, L., Li, J., Wang, C., Ma, Y., and Huo, K. (2005). A new human gene hNTKL-BP1 interacts with hPirh2. Biochem Biophys Res Commun 330, 293-297. 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. Mol Cell Biol 23, 8902-8912. Zheng, G., Ning, J., and Yang, Y. C. (2007). PLAGL2 controls the stability of Pirh2, an E3 ubiquitin ligase for p53. Biochem Biophys Res Commun 364, 344-350.

Page 133: Characterization of the E3 Ubiquitin Ligase Pirh2 · The p53 gene encodes a transcription factor that regulates gene expression in response to various stresses such as DNA damage

124

Zheng, N., Wang, P., Jeffrey, P. D., and Pavletich, N. P. (2000). Structure of a c-Cbl-UbcH7 complex: RING domain function in ubiquitin-protein ligases. Cell 102, 533-539. Zhong, Q., Gao, W., Du, F., and Wang, X. (2005). Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell 121, 1085-1095.