SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a...

126
SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting with the PML-p53 pathway by Deng Pan A dissertation submitted in partial satisfaction of the requirements for the degree of Doctor of Philosophy in Molecular and Cell Biology in the Graduate division of University of California, Berkeley Committee in charge: Professor Kunxin Luo, Chair Professor Judith Campisi Professor Steven Martin Professor Luan Sheng Professor Astar Winoto Fall 2010

Transcript of SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a...

Page 1: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by

interacting with the PML-p53 pathway

by

Deng Pan

A dissertation submitted in partial satisfaction of the requirements

for the degree of

Doctor of Philosophy

in

Molecular and Cell Biology

in the

Graduate division

of

University of California, Berkeley

Committee in charge:

Professor Kunxin Luo, Chair

Professor Judith Campisi

Professor Steven Martin

Professor Luan Sheng

Professor Astar Winoto

Fall 2010

Page 2: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting
Page 3: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

1

Abstract

SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by

interacting with the PML-p53 pathway

by

Deng Pan

Doctor of Philosophy in Molecular and Cell Biology

University of California, Berkeley

Professor Kunxin Luo, Chair

SnoN, a member of the Ski proto-oncoprotein family, was originally classified as an oncogene based on its ability to transform chicken and quail embryonic fibroblasts. Elevated levels of SnoN have been detected in many human carcinoma cell lines. Furthermore reduction of SnoN expression in breast and lung cancer cell lines impairs their ability to proliferate in culture and form tumors in vivo. Moreover, expression in mouse mammary glands of a SnoN fragment containing residues 1-366 promotes PyVmT-induced (polyomavirus middle T antigen) tumor growth and metastasis. These findings strongly support the idea that SnoN has pro-oncogenic properties. However, there is also evidence that SnoN might be anti-oncogenic as well. Although increased expression of SnoN has been observed in certain cancers and at specific stages of malignant transformation, downregulation of SnoN has been shown in other types of cancers. Loss of one allele of SnoN in mice has been shown to increase their susceptibility to chemical carcinogen-induced tumorigenesis. Overexpression of dSnoN (the Drosophila homologue of SnoN) has negative effects on cell growth. SnoN has also been reported to cooperate with p53 to regulate p53 target gene expression. Together, the evidence suggests that SnoN might play a dual role in tumorigenesis. How can SnoN possess two opposite functions in tumorigenesis? What are the mechanisms underlying the dual role of SnoN in tumorigenesis? And, what are the potential regulators of SnoN expression in different types and stages of cancer? My dissertation research aimed to answer those questions.

I have shown that elevated cellular SnoN expression has anti-oncogenic effects both in vivo and in culture. The levels of SnoN are tightly regulated in cells. SnoN expression is relatively low in normal cells, but is significantly elevated when cells are exposed to certain types of stress. Multiple pathways seem to function cooperatively to regulate the expression of SnoN in response to stress. I have shown that the ATM/ATR kinase, MEK/Erk kinase and PI3K kinase pathways, which are activated by stress signals, are necessary for upregulation of SnoN by stress (Chapter III). The elevated SnoN expression has anti-oncogenic functions by virtue of inducing premature senescence. SnoN-induced senescence is mediated by the interaction of SnoN with the PML-p53 pathway. SnoN interacts with PML and is recruited to PML nuclear bodies (Nbs), resulting in the stabilization and activation of p53 by competitively displacing

Page 4: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

2

Mdm2 from p53. In addition, SnoN enhances post-transcriptional modifications of p53 in PML NBs. Through these mechanisms, SnoN participates in activating the transcription of p53 target genes when recruited to their promoters together with p53. SnoN therefore mediates rapid p53 activation in response to specific cellular stress signals. In the absence of SnoN, p53-dependent stress responses are significantly delayed and early stage responses in stressed cells fail to be activated properly. Since p53 plays a central role in tumor suppression as well as regulation of ageing, activation of p53 by SnoN can prevent carcinogen-induced tumorigenesis and accelerate stress related ageing.

Page 5: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

i

Dedication

To my wife, my parents and my sons who are the center of my life.

Thank you for many years’ accompanying, understanding and encouragement

(献给我的太太,父母和孩子们,你们是我的挚爱

感谢你们多年的陪伴,理解和鼓励。)

Page 6: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

ii

Table of contents

Dedication ........................................................................................................................................ i Table of contents............................................................................................................................. ii List of Figures ................................................................................................................................. v List of Tables ................................................................................................................................. vi List of abbreviations ..................................................................................................................... vii Acknowledgement ......................................................................................................................... ix Chapter 1: Introduction ................................................................................................................... 1

TGFβ signaling pathway......................................................................................................... 2 Role of TGFβ signaling in tumorigenesis............................................................................... 4

Tumor suppression function of TGFβ signaling............................................................. 4 The pro-oncogenic function of TGFβ signaling ............................................................. 4

SnoN, a negative regulator of TGFβ signaling....................................................................... 5 Identification and characterization of SnoN ................................................................... 5 Expression patterns and biological functions of SnoN................................................... 5 Role of SnoN in tumorigenesis. ...................................................................................... 6 Potential mechanism for the role of SnoN in tumorigenesis .......................................... 7

p53 and stress responses ......................................................................................................... 8 Chapter 2: Materials and methods ................................................................................................ 10

Knock-in Mice ...................................................................................................................... 11 Isolation of MEFs (Mouse embryonic fibroblast) ................................................................ 11 Population doubling curve: ................................................................................................... 11 Other cells, antibodies, and constructs.................................................................................. 12 Transfection and Luciferase.................................................................................................. 12 Immunoprecipitation and Western Blotting.......................................................................... 12 Nuclear extraction of MEFs.................................................................................................. 12 Pulse Chase Assay ................................................................................................................ 13 RT-PCR................................................................................................................................. 13 GST pull-down assay............................................................................................................ 13 SA-β-gal staining in cells and tissues ................................................................................... 13 Tunel staining in frozen sections .......................................................................................... 14 Retrovirus replication and infection...................................................................................... 14 Immunofluorescence............................................................................................................. 14 H&E (Haematoxylin and Eosin) staining for frozen tissue sections .................................... 15 Chromatin immunoprecipitation (ChIP) assay ..................................................................... 15 BrdU incorporation assay in attached cells........................................................................... 15 Soft-agar assay ...................................................................................................................... 16 Skin carcinogenesis- DMBA/TPA........................................................................................ 16

Chapter3: SnoN functions as a tumor suppressor by inducing premature senescence ................. 17 Abstract ................................................................................................................................. 18 Introduction........................................................................................................................... 19 Results................................................................................................................................... 21

Generation of knock-in mice expressing a mutant SnoN ............................................. 21 Knock-in mice are resistant to carcinogen-induced skin tumorigenesis....................... 21

Page 7: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

iii

MEF expressing mutant SnoN displayed premature senescence.................................. 22 Premature senescence is caused by the elevated SnoN expression .............................. 22 p53 upregulation is required for SnoN-induced premature senescence........................ 23 PML mediates SnoN-induced p53 stabilization and premature senescence................. 24 SnoN physically interacts with PML to promote premature senescence...................... 25 PML expression is upregulated in the m/m MEF ......................................................... 26 Relationship between SnoN, PML and p53 during senescence.................................... 26 High levels of SnoN inhibit transformation of MEF by oncogenes ............................. 27 Blocking the SnoN-PML interaction or abolishing p53 eliminates the anti-oncogenic activity of SnoN ............................................................................................................ 27

Discussion............................................................................................................................. 29 Materials and methods .......................................................................................................... 31 Figure legends....................................................................................................................... 33 Figures................................................................................................................................... 36

Chapter 4: SnoN, a novel activator of p53.................................................................................... 46 Abstract ................................................................................................................................. 47 Introduction........................................................................................................................... 48 Results................................................................................................................................... 50

SnoN binds to p53 directly. .......................................................................................... 50 Binding of SnoN and p53 is required for SnoN-induced senescence........................... 50 Binding of SnoN to p53 is required for stabilization of p53......................................... 51 SnoN competes with Mdm2 for binding to p53............................................................ 51 SnoN binds to the same site as Mdm2 and enhances the modification of p53............. 51 SnoN competes with Mdm2 for binding to the p53 responsive promoter DNA. ......... 52 SnoN and PML can synergistically activate p53 dependent transcription. .................. 52 The SnoN/p53 interaction is required for premature senescence. ................................ 53 Activation of p53 is required for the anti-oncogenic activity of SnoN......................... 53 Activation of p53 by SnoN has an anti-oncogenic function in vivo............................. 53 The SnoN-p53 pathway also regulates ageing in mice. ................................................ 54

Discussion............................................................................................................................. 55 Materials and methods .......................................................................................................... 58 Figure legends....................................................................................................................... 60 Figures................................................................................................................................... 64 Tables.................................................................................................................................... 75

Chapter 5: SnoN, a stress transducer mediating p53 dependent stress responses ........................ 78 Abstract ................................................................................................................................. 79 Introduction........................................................................................................................... 80 Results................................................................................................................................... 83

Oxidative stress can regulate expression of SnoN........................................................ 83 Induction of SnoN requires multiple pathways ............................................................ 83 SnoN plays an important role in regulating the early responses of p53 under stress conditions...................................................................................................................... 85 SnoN is also important for the activation of p53 in response to stress in epithelial cells........................................................................................................................................ 86

Discussion............................................................................................................................. 87 Materials and methods .......................................................................................................... 89

Page 8: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

iv

Figure legend ........................................................................................................................ 91 Figures................................................................................................................................... 93

Figure 1. Oxidative stress can induce the expression of SnoN..................................... 93 Figure 2. Pathways mediate the upregulation of SnoN by oxidative stress.................. 94 Figure 3: SnoN plays an important role in regulation of p53 dependent stress responses........................................................................................................................................ 95

Chapter 6: Discussion and future direction................................................................................... 96 Chapter 7: Reference................................................................................................................... 100

Page 9: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

v

List of Figures

Chapter 3

Figure 1. Generation of SnoN knock-in mice............................................................... 36 Figure 2. The snoN knock-in mice are resistant to chemical carcinogen-induced skin tumorigenesis. ............................................................................................................... 37 Figure 3. MEF from the snoN knock-in mice displayed premature senescence........... 38 Figure 4. Elevated SnoN expression is responsible for premature senescence of the m/m MEF...................................................................................................................... 39 Figure 5. p53 is required for the premature senescence of m/m MEF......................... 40 Figure 6. Interaction between SnoN and PML is required for premature senescence.. 41 Figure 7. SnoN upregulates PML expression to mediate p53 stabilization.................. 42 Figure 8. Oncogenic transformation of MEF cells ....................................................... 43 Supplemental figure 1 ................................................................................................... 44 Supplemental figure 2 ................................................................................................... 45

Chapter 4

Figure 1. SnoN directly bind to p53 when they are highly expressed in cells.............. 64 Figure 2. Mapping the binding site of p53 in SnoN and SnoN can interact with PML independently of p53..................................................................................................... 65 Figure 3. The binding of SnoN to p53 leads to the stabilization of p53 by competing with Mdm 2 binding...................................................................................................... 66 Figure 4. SnoN binds to the same site as Mdm2 in p53 and also enhances the post-trancriptional modification of p53 ................................................................................ 67 Figure 5. SnoN can ben recruited to the p53 specific binding elment of p21 gene and activate p53 dependent transcription by antagonizing Mdm2 ...................................... 68 Figure 6. Activation of p53 by SnoN can trgger p53 dependent senescence and block oncogene induced transformation in cells..................................................................... 69 Figure 7. Activation of p53 by SnoN prevents chemical carcinogen induced tumorigenesis ................................................................................................................ 70 Figure 8. Premature aging associated phenotypes in knock-in mice with mutant SnoN........................................................................................................................................ 71 Figure 9. Senescence or apoptotic cells are accumulated in aged tissues from knock-in mice............................................................................................................................... 72 Sup_Fig 1: Mapping the binding site of p53 in SnoN and SnoN can bind to Smad proteins independent of p53.......................................................................................... 73 Sup_Fig 2: The half life of SnoN is unaffected by p53 and Mdm2 can not release SnoN from p53........................................................................................................................ 74

Chapter 5

Figure 1. Oxidative stress can induce the expression of SnoN..................................... 93 Figure 2. Pathways mediate the upregulation of SnoN by oxidative stress.................. 94 Figure 3: SnoN plays an important role in regulation of p53 dependent stress responses........................................................................................................................................ 95

Page 10: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

vi

List of Tables

Chapter 4

Table 1: Shorter lifespan in m/m mice.......................................................................... 75 Table 2: Metabolic activities in aged mice ................................................................... 76

Page 11: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

vii

List of abbreviations

ALK Activin receptor like kinase ATM Ataxia telangiectasia mutated protein ATR Ataxia telangiectasia and Rad3 related BMP Bone morphogenic protein bp Base pair Co-Smad Common Smad DMEM Dullbecco’s modified eagle medium DNA Deoxyribonucleic acid DTT Dithiothreitol ECM Extracellular matrix EMT Epitheilal-to-mesenchymal transdifferentiation FBS Fetal bovine serum flox LoxP sites flanking GST Glutathione-S-tranferase HA Hemagglutinin HDAC Histone Deacetylase hr Hour I-Smad Inhibitory Smad kDa Kilodalton LAP Latency-associated peptide LTBP Latent TGFβ binding protein MEF Mouse embryonic fibroblast MEM Minimal essential medium MH domain Mad homology domain min mintute ml milliliter mM millimolar MMP Matrix metalloprotease mSnoN Mutant SnoN deficient in Smad binding mRNA Messenger ribonucleic Acid MTT 3-(4, 5 dimethylthiazol-2yl)2, 5 diphenyl tetrazolium

bromide NES Nuclear export sequence NLS Nuclear localization signal NPC Nuclear pore complex p53 Tumor protein 53 PAGE Polyacrylamide gel electrophoresis, PCR Polymerase chain reaction PML NBs promyelocytic leukemia protein nuclear bodies PBS phosphate buffered saline pM Picomolar RE1 p53 responsive element PMSF phenylmethylsulfonylfluoride

Page 12: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

viii

R-Smad Receptor regulated smad RT-PCR Reverse transcriptase chain reaction SAND domain Domain in Sp100, AIRE-1, NucP41/75 and DEAF1 SARA Smad anchor for receptor activation SBE Smad binding element SDS sodium dodecyl sulphate shRNA Small hairpin RNA siRNA Small interfering RNA Sno Ski related novel protein Ski Sloan kettering virus oncoprotein TGFβ Transforming growth factor β TβRI Type I TGFβ receptor TβRII Type II TGFβ receptor μg microgram μl microlitter μM micromolar wt wildtype +/+ wildtype m/m Homozygous mutant -/- Homozygous knockout

Page 13: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

ix

Acknowledgement

Many people have helped me both academically and personally during the years when I

was studying at UC Berkeley. First, I want to thank my mentor, Dr. Kunxin Luo for all her support throughout all these years and for teaching me how to design good experiments, organize and present the results nicely, the most important things that I have learned in the Ph.D program. Second, I want to give my thanks to my current and former lab-mates with whom I spent most of my time in the lab. I really appreciate for their nice collaboration and help with experiments, and for everything they taught me through the interesting scientific discussions. Especially, Qingwei Zhu, Fangjiu Zhang and Luis Estevez were always willing to teach how to run experiments and answer my questions. Third, I want to thank all members of my thesis committee for their prompt feedback and instructive mentoring. Without these, it is impossible for me to finish the projects. This work was supported by DOE BCRP pre-doctoral fellowship.

Page 14: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

1

Chapter 1: Introduction

Page 15: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

2

Activation of oncogenes and inactivation of tumor suppressor genes are critical events for the multistage tumorigenesis process. Oncogenes are a group of genes which can promote tumor growth when activated by gain of function mutations; while the tumor suppressor genes can promote the transformation only when inactivated by loss of function mutations. The definitions for oncogene and tumor suppressor gene are very distinct. However, it is not easy in reality to distinguish an oncogene or a tumor suppressor gene. It has been well established that some proteins or signaling pathways may function differently in different cell context. Both oncogenic and antioncogenic functions could be possessed by a protein or pathway at the different stages determined by the cell context such as TGFβ pathway (Akhurst and Derynck, 2001; Derynck et al., 2001a) and its negative regulator, SnoN (Deheuninck and Luo, 2009; Jahchan and Luo, ; Pan et al., 2009). The interaction of SnoN with TGFβ signals is important for its functions in tumorigenesis but interestingly, SnoN can also play a role independently of TGFβ.

TGFβ signaling pathway

Transforming growth factorβ (TGFβ) signaling pathway has pleiotropic functions regulating many processes ranging from embryonic patterning, organ development, immune regulation and tissue homeostasis (Massague, 1998). TGFβ is the founding member of a larger family of structurally related cytokines, which was first identified from human platelets and human placenta based on its ability to induce anchorage-independent growth in non-transformed rat kidney fibroblast (Frolik et al., 1983; Roberts et al., 1982). The active form of TGFβ exists as a homodimer of two 112 amino acid subunits produced from proteolytic cleavage of a longer precursor polypeptide (Derynck et al., 1985). Since the first discovery and cloning of TGFβ, the other members of the TGFβ superfamily have been identified in mammals including Bone morphogenic protein (BMP), Activin, Nodal, Inhibin and Vg1 subfamilies, all of which share varying degrees of homology in their cysteine-rich C-termial bioactive domains (Massague, 1990).

The properties of TGFβ in regulation of growth and proliferation of normal cells were soon uncovered after its purification. Although TGFβ was identified based on its transformation ability, a growth inhibitory effect of this growth factor was uncovered from its growth inhibitory function on African green monkey kidney cells (Tucker et al., 1984). The inhibitory effects of TGFβ on growth and proliferation were subsequently documented in different types of cells including epithelial, endothelial, fibroblast, lymphoid and hematopoietic cells (Anzano et al., 1986; Hayashi and Carr, 1985; Kehrl et al., 1986; Shipley et al., 1986). In fact, the transforming activity of TGFβ is only limited to certain selected fibroblast cells in the presence of other additional factors (Roberts et al., 1985).

TGFβ signal transduces from extracellular space to inside of cell through different sets of TGFβ receptors, a family of structurally related transmemberane serine/threonine kinases. Two major classes of receptors, TβRI (TGFβ type I) abd TβRII (TGFβ type II) receptors have been characterized to have a high affinity with TGFβ ligands (Massague, 1998). Type I and type II receptors are both required for TGFβ activation. Upon the active ligand binding to type II receptors whose kinase domains are constitutively active, the complex recruits the type I receptors and begin to transduce the signal into the cell’s interior (Wrana et al., 1994). Type I receptors recognize ligand only when complexed with type II receptors. At this point, TGFβ ligands act as a bridge to mediate the formation of receptor heteromeric complex. When type I

Page 16: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

3

receptors are recruited to type II receptors, they can be phosphorylated in the GS domain by the constitutively active type II receptors. Activation of type I receptors by phosphorylation can activate its kinase activity and propagate the signals to downstream substrates (Attisano et al., 1993; Bassing et al., 1994; Inagaki et al., 1992; Luo and Lodish, 1996; Massague et al., 1992; Wrana et al., 1992).

Genetic studies in Drosophila and C. elegans lead to the identification of the cytoplasmic signal transducers of TGFβ pathway in mammalian cells (Derynck et al., 1998; Itoh et al., 2000; Massague, 2000; Massague and Wotton, 2000). Three categories of Smad proteins have been identified based on their functions in the signaling transduction of TGFβ. Receptor regulated Smads (R-Smads) are the proteins which can be directly phosphorylated by type I receptors. Different TGFβ family ligands can cause the activation of different subtypes of R-Smads. Smad2 and Smad3 are activated by the signals from TGFβ or activin; while Smad1, Smad5 and Smad8 are activated specifically by BMP ligands. Once phosphorylated, R-Smads form heteromeric complex with the common Smad (Co-Smad) Smad4, another class of Smad proteins. Importantly, Smad4 is the only Co-Smad which is required by the signaling transduction of all TGFβ family members. The third class of Smads are called as inhibitory Smads (I-Smads) including Smad6 and Smad7. The I-Smads inhibit the signaling transduction of TGFβ family members by several potential mechanisms (See Turn off the signals). Both Smad6 and Smad7 are induced by TGFβ signals and repress the signals through negative feedback loops (Derynck et al., 1998; Itoh et al., 2000; Massague, 2000; Massague and Wotton, 2000).

Genetic, biochemical and structural studies have provided the detailed mechanism of the activation of Smad proteins through the interaction of TGFβ ligand and receptor complex. Upon ligand activation, receptor complexes are formed and subsequently, type I receptors are activated. Activated type I receptor kinases directly phosphorylate R-Smad proteins on two serine residues in their SSXS motif further propagating the TGFβ signals inside the cells (Abdollah et al., 1997; Macias-Silva et al., 1996; Souchelnytskyi et al., 1997; Zhang et al., 1996). Phosphorylated R-Smad proteins are released from the receptor complex and subsequently form a complex with Co-Smad (Tsukazaki et al., 1998; Wu et al., 2000). After phosphorylation and formation of the Smad complex, the affinity of Smad proteins with these cytoplasmic anchors is decreased, the nuclear localization signals (NLS) are exposed and the nuclear export signals (NES) are inhibitor (Kurisaki et al., 2001; Xiao et al., 2000a; Xiao et al., 2000b; Watanabe et al., 2000; Xiao et al., 2000a).. The Smad complexes undergo efficient nuclear translocation, leading to accumulation of Smad proteins in the nucleus.

Upon ligand stimulation, Smad complex is rapidly formed and accumulated in the nucleus. When the Smad complex migrates to the nucleus, it can bind to a Smad-binding element within the promoters of TGFβ responsive genes and regulate the expression of these target genes by association with a variety of transcriptional factors (Massague and Wotton, 2000). For years of study, more than 40 different structurally diverse transcription factors have been reported to interact with Smad proteins including the basic helix-loop-helix, bZip, homeodomain, forehead, nuclear receptor, Runx and zinc finger families (Feng and Derynck, 2005). The interaction of Smads with different co-factors leads to either transcription activation or repression depending on the specific promoter context and other associated transcription factors. By recruitment of co-factors, Smad proteins can modify the transcriptional outcomes through regulating promoter proximity to RNA polymerase II or altering the modification of core Histone tails on chromosomes or by other mechanisms (Feng and Derynck, 2005). However, the mechanism

Page 17: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

4

underlying the precise control of TGFβ signaling in vivo in a temperal and spatial manner is still very limited, and requires further investigation.

Role of TGFβ signaling in tumorigenesis

Tumor suppression function of TGFβ signaling

The tumor suppression function of TGFβ signaling is rooted in anti-proliferating effects of TGFβ in different cell types. Appropriately, mutations in the major components of TGFβ pathways have been detected in many human tumor types and mouse models of tumorigenesis. Mutations in TGFBR2, the gene encoding TβRII, are commonly detected in patients with HNPCC (Hereditary non-polyposis colorectal cancer), and frequently observed in most colorectal and gastric carcinomas with microsatellite instability (Markowitz et al., 1995; Myeroff et al., 1995; Ohue et al., 1996; Park et al., 1994). Mutations that inactivate TβR1 genes have also been detected in ovarian, breast and pancreatic cancers and T cell lymphomas (Chen et al., 1998; Goggins et al., 1998; Schiemann et al., 1999). Inactivating mutations of Smad4 locus is observed to occur in half of human pancreatic cancers and one third of colorectal cancers (Blobe et al., 2000; Hahn et al., 1996; Schutte et al., 1996). Mutations in Smad2 have also been detected in certain colorectal cancers even it is rare (Eppert et al., 1996; Ohtaki et al., 2001). Genetic mouse models from mutations or deletions of TGFβ, receptors and Smad proteins have further supported the tumor suppressing ability of TGFβ signaling in vivo. (Engle et al., 1999; Tang et al., 1998; Bottinger et al., 1997a; Bottinger et al., 1997b; Gorska et al., 1998; Siegel et al., 2003; Xu et al., 2000). The accumulation of these studies strongly suggests that TGFβ signals have an anti-oncogenic function in vivo.

The pro-oncogenic function of TGFβ signaling

Although mutations in TGFβ receptors or Smads are detected in certain types of tumors, many human cancers and most cancer cell lines posses an intact TGFβ signaling pathway with functional core components (Derynck et al., 2001a; Siegel and Massague, 2003). Despite the tumor suppressor function of TGFβ in untransformed cells, it is well established that TGFβ can also elicit pro-oncogenic effects on transformed tumor cells and tumor micro-environments (Derynck et al., 1987; Dickson et al., 1987; Gorsch et al., 1992). In the process of tumor development, transformed cells selectively lose the anti-proliferation effects of TGFβ signaling while the remaining pro-oncogenic function of TGFβ actually facilitates tumor progression (Derynck et al., 2001b; Siegel and Massague, 2003; Wakefield and Roberts, 2002). TGFβ signaling plays an important role in the epithelial-to-mesenchymal trans-differentiation (EMT) in both embryogenesis and tumor progression (Grunert et al., 2003; Thiery, 2002). TGFβ can promote EMT, leading to the acquisition of a migrative, invasive phenotype that is normally associated with the metastatic capability of cancer cells. In mouse models, TGFβ has been reported to enhance the metastasis of breast cancer cells to bone and lung tissues (Muraoka-Cook et al., 2004). In metastatic breast cancer cell lines, osteolytic capability was blocked by introduction of a dominant negative TβRII and enhanced by introduction of a constitutively active TβRI (Kakonen et al., 2002; Yin et al., 1999).

Page 18: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

5

The oncogenic functions of TGFβ signals not only depend on the effects of TGFβ on cancer cells but also are contributed by the effects of TGFβ on the tumor micro-environment. First, TGFβ promotes angiogenesis in tumor tissues facilitating tumor growth and metastasis by increasing the availability of oxygen and nutrients to tumor cells. (Chang et al., 1999; Oh et al., 2000; Oshima et al., 1996; Stearns et al., 1999; Ueki et al., 1992). Additionally, TGFβ also plays a role in the evasion of tumor cells from anti-tumor immune surveillance leading to malignant progression. (Gorelik and Flavell, 2000; Gorelik and Flavell, 2002; Chen and Wahl, 2003; Fantini et al., 2004; Huber et al., 2004; Rao et al., 2005; Yamagiwa et al., 2001). Furthermore, TGFβ can have a direct effect on the extracellular matrix components of tumor cells. TGFβ has an ability to regulate both deposition and degradation of ECM, thus regulating the dynamic processes of ECM remodeling. (Dean et al., 1988; Ignotz and Massague, 1986; Massague, 1990; Roberts et al., 1988; Agarwal et al., 1994; Delany and Canalis, 2001; Sehgal and Thompson, 1999; Shimizu et al., 1996) By regulating deposition and degradation of ECM, TGFβ signaling is capable of facilitating both tumor cell invasion as well as angiogenesis.

SnoN, a negative regulator of TGFβ signaling

Identification and characterization of SnoN

SnoN is a member of Ski pro-oncoprotein family including two isoforms of Ski and four isoforms of Sno (SnoN, SnoN2, SnoA and SnoI). Ski, the prototypical member, was first identified as viral oncogene (v-Ski) from the Sloan-Kettering avian retrovirus (Li et al., 1986; Stavnezer et al., 1986). Based on the homology to ski gene, SnoN and its isoforms (SnoN2 and SnoA) were identified and cloned (Nomura et al., 1989; Pearson-White, 1993; Pearson-White and Crittenden, 1997; Stavnezer et al., 1989; Sutrave et al., 1990; Sutrave and Hughes, 1989). Overexpression of Ski and SnoN can transform chicken embryonic fibroblast and induce terminal muscle differentiation of quail embryonic cells (Boyer et al., 1993; Colmenares and Stavnezer, 1989; Colmenares et al., 1991). All members of this family share a conserved N-terminal domain that is necessary and sufficient for their transformation and differentiation capabilities (Cohen et al., 1998; Nomura et al., 1989; Pearson-White, 1993; Zheng et al., 1997). Like Ski protein, overexpression of SnoN results in its nuclear localization (Barkas et al., 1986; He et al., 2003; Sutrave et al., 1990). However, at the physiological conditions, SnoN is likely both nuclear and cytoplasmic depending on the cell type and stage of cell development (Krakowski et al., 2005; Zhang et al., 2003). In certain cases, SnoN can localize to very distinct cellular locations, such as the localization of SnoN to promyelocytic leukemic nuclear bodies (PML NBs) in senescence cells as I have demonstrated (Pan et al., 2009).

Expression patterns and biological functions of SnoN

SnoN in embryogenesis SnoN is ubiquitously detected in all embryonic and adult tissues although the expression

level of SnoN is relatively low in most cases. However, elevated expression levels of SnoN were observed in certain stages of embryogenesis and adult tissues as well as in some cancer cell lines (Nomura et al., 1989, Pearson-White, 1997 #5011; Pelzer et al., 1996).

Upregulation of snoN transcript in moue embryos is first observed at 9.5 days p.c (post coitum) in neural tissues, neural crest cells, developing limb bud, vasculatures and mandibular

Page 19: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

6

arches. snoN transcript was induced in the developing skeletal muscle after 11.5 days p.c, and started to increase in tissues with high proliferative potentials (Pelzer et al., 1996). However, the function of SnoN in embryogenesis is still unclear. There are three existing lines of snoN knock-out mice, two with a deletion in exon 1 and one targeted to promoter of snoN. Interestingly, only one line of snoN knock-out mice showed embryonic defects and all embryos dies at E3.5 probably due to a defect in blastocyte formation (Shinagawa et al., 2000). The other two lines are completely viable and fertile although mildly defected in T cell activation due to reduced IL-2 and IL-4 expression (Pearson-White and McDuffie, 2003). Definitely, more studies are required to further characterize the role of SnoN in embryogenesis. Expression of SnoN in adult tissues The expression of SnoN was observed at certain stages of adult tissue development. In mammary glands, SnoN is detected at a low level in luminal epithelial cells. Its expression is significantly elevated during late pregnancy, reaching a maximal level at around 18.5 days of gestation and maintaining its elevated level through the beginning of lactation. Then, SnoN expression starts to decrease at the 3rd day of lactation and returns to the basal level in mature virgin gland (Jahchan et al.). The dynamic expression pattern of SnoN in mammary gland development suggests that it may play an important role in regulating the proliferation and differentiation of the secretory alveolar epithelial cells. Indeed, studies in a transgenic mouse line expressing a fragment of SnoN under a MMTV (mammary gland tumor virus) promoter and SnoN null mice, have showed that SnoN might promote side-branching and lobular-alveolar proliferation in virgin gland (Jahchan et al.). In the adult neuronal system, snoN transcript is enriched in both the cerebrum and cerebellum and its protein product is detected in the nucleus of the granule neurons of the cerebellum and in both granule neurons and Purkinje cells in the cerebellar cortex (Pelzer et al., 1996; Stegmuller et al., 2006). It suggests that SnoN might be important for the development of granule neuron. Reducing expression of SnoN in the cerebellar cortex of rat markedly impaired axon growth (Stegmuller et al., 2006). This activity appears to be linked with the actin binding protein Ccd1 which activates JNK kinase at axon termini to promote axon growth. SnoN is a direct inducer of Ccd1 transcription by cooperating with p300/CBP and plays an important role in the regulation of axon morphogenesis (Ikeuchi et al., 2009). SnoN is also expressed in both the nucleus and cytoplasm of keratinocytes in the outer suprabasal layers but not in the basal cell layers. The expression of SnoN can be detected in benign seborrheic keratosis and is elevated in non-metastastic intraepidermal squamous cell carcinoma but absent in undifferentiated cells in invasive squamous cell carcinoma (Zhang et al., 2009). These suggest that SnoN might regulate the differentiation of epidermis as well. SnoN has also been reported to have a role in regulating hematopoietic lineage and muscle differentiation (Pearson-White and Crittenden, 1997). However, the function of SnoN in these tissues has not yet been clearly clarified.

Role of SnoN in tumorigenesis.

SnoN has both oncogenic and anti-oncogenic functions. Overexpression of SnoN can transform chicken embryonic fibroblast cells suggesting that SnoN might have a pro-oncogenic function (Boyer et al., 1993). Actually, elevated SnoN expression is detected in many human cancer cell lines including melanoma and carcinoma cells of the lung breast, ovary, vulva, prostate, esophagus and thyroid (Deheuninck and Luo, 2009; Jahchan and Luo). Additionally, the locus of human snoN gene is at chromosome 3q26.2, a frequent amplified region in many cancer types including esophagus, lung, ovary, cervix, head and neck and prostate. It is reported

Page 20: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

7

by Qingwei in our group several years ago that reducing SnoN expression by shRNA in invasive breast cancer and lung cancer cell lines impaired tumor growth both in vivo and in vitro (Zhu et al., 2006). Moreover, another report by Nadine in our group have shown that overexpression of SnoI, a C-terminal truncated SnoN in mouse mammary gland promotes tumor formation and metastasis induced by the polyoma middle T antigen (PyVmT) (Jahchan et al.). Both reports give strong evident that SnoN has pro-oncogenic activity in vivo.

Recently, there is emerging evidence to support that SnoN might have anti-oncogenic activity as well. Although the overexpression or amplification of snoN gene is observed in many cancer types, the downregulation of SnoN expression has also been observed in certain cancer types. In a recent study of tissue samples from patients with Barrett's esophagus, a precancerous condition that is associated with an increased risk of adenocarcinoma, the highly expression of SnoN was only detected in tissues with low grade dysplasia but either entirely absent or expressed only at very low levels in those tissues with high grade dysplasia or adenocarcinoma (Villanacci et al., 2008). In colorectal cancer with microsatellite instability SnoN is found to be downregulated in 39% of samples and upregulated in 33% samples (Chia et al., 2006). The dynamic expression of SnoN in different stages of cancer suggest that high levels of SnoN, while beneficial to certain stages of tumor development, may not always be advantageous for malignant progression. In mouse model, loss of one allele of SnoN in mice results in a slightly higher incidence of tumor formation and increased susceptibility to chemical induced carcinogenesis (Shinagawa et al., 2000). Additionally, reducing SnoN expression in human lung and breast cancer cells enhances EMT and promotes tumor metastasis in vivo (Zhu et al., 2006). Furthermore, SnoN has been shown to cooperate with p53 in silencing of the alpha-fetoprotein gene, which is aberrantly overexpressed in liver cancer cells (Wilkinson et al., 2005).

Potential mechanism for the role of SnoN in tumorigenesis

Interaction of SnoN with TGFβ signaling and the PML-p53 pathway Sequence analysis reveals that there is not any enzymatic domain in the amino acid

sequence of SnoN and no intrinsic catalytic activities are identified so far. Moreover, SnoN is incapable of directly binding to DNA but is found to be incorporated into DNA complexes by association with other DNA-binding cofactors. It seems that the effects of SnoN in cell are reliant upon the interaction with other factors in cells.

We and others have shown that SnoN physically interacts with R-Smad and Co-Smad and functions as a negative regulator of TGFβ signaling. The interactions of SnoN with Smad proteins have been identified by using a wide array of approaches including affinity purification with Smad proteins and yeast two hybrid screens for Smad associated factors. The binding of SnoN with Smad proteins is mediated by residues in MH2 domain of the Smad proteins and the N-terminal half of SnoN (Akiyoshi et al., 1999; Stroschein et al., 1999; Sun et al., 1999a).

When overexpressed, SnoN represses Smad transactivation and blocks TGFβ induced growth arrest in cells (Akiyoshi et al., 1999; Stroschein et al., 1999; Sun et al., 1999a). The interaction of SnoN with the Smad complex represses their transactivation activity through several redundant mechanisms. The binding of SnoN to the Smad complex leads to the disruption of a functional heteromeric Smad complex, recruitment of transcription a co-repressor complex including N-CoR and HDAC to the Smad complex and releasing of the binding of transcriptional co-activators from the Smad complex. When localized to cytoplasm, SnoN can sequester Smad proteins and prevent Smad from undergoing nuclear translocation in response to

Page 21: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

8

TGFβ stimulation. Because of these capabilities, SnoN has been characterized as a repressor of the Smad proteins (Akiyoshi et al., 1999; Krakowski et al., 2005; Nomura et al., 1999; Stroschein et al., 1999; Wu et al., 2002).

On the other hand, the expression of SnoN is tightly controlled by TGFβ signaling as well. Post 30 minutes of TGFβ stimulation, SnoN is capable of rapidly degradation through ubiquitin mediated protein degradation. The polyubiquitination of SnoN is elicited by the binding of R-smad with SnoN leading to recruitment of several E3 ubiquitin ligases to SnoN including Smurf, APC and Arkadia. Within two hours of stimulation, TGFβ induce SnoN expression in a Smad dependent upregulation of snoN mRNA (Deheuninck and Luo, 2009; Luo, 2004; Stroschein et al., 1999; Zhu et al., 2005). However, this latter induction of SnoN by TGFβ signals has yet been functionally characterized. It might function to shut off the TGFβ response at a later stage in a negative feedback.

This ability to antagonize TGFβ signaling may be responsible for the pro-oncogenic activity of SnoN. Indeed, mutant SnoN incapable of binding Smad proteins, loses its transformating capability (He et al., 2003). In a previous study from our group, reduction of SnoN expression in human lung and breast cancer cells markedly impaired the ability of tumor cells to undergo anchorage-independent growth and tumorigenesis in nude mice. However, these tumor cells with reduced SnoN displayed enhanced EMT and showed more metastatic potential than their parental cells in vivo (Zhu et al., 2006). It supports that SnoN acts as both an oncogene and tumor suppressor with regard to different aspects of tumorigenesis. These are reminiscent of the abilities of TGFβ signals to suppress or promote tumor cell progression at the different stages of tumorigenesis. Indeed, the tumor suppressive activities in tumor cells with reduced SnoN expression are due to restoring TGFβ-induced growth arrest in tumor cells while the promoting activities at later stages are partially through activating TGFβ-enhanced EMT (Zhu et al., 2006).

To understand the role of SnoN in tumorigenesis, knock-in mice with a mutant snoN gene deficient in binding to R-Smad proteins and Smad4 were generated in lab. These mice allow us to study the role of SnoN and Smad interaction in vivo and determine whether SnoN can play a role in tumorigenesis independent of Smad functions. From studying these knock-in mice, I have dissected a novel pathway, PML-p53 pathway which can interact with SnoN and mediate its anti-oncogenic properties.

p53 and stress responses

p53, the guardiam of the genome, acts as a stress responder to a wide array of intrinsic and extrinsic stress signals including oxidative stress, DNA damage, hypothexia, abberantly activated oncogenes and metabolic abnormality (Ljungman, 2007; Vousden, 2002). p53 functions as a node for organizing the cellular responses such as growth arrest, DNA repair, senescence or apoptosis to various types and levels of stress signals (Balint and Vousden, 2001; Vousden and Lu, 2002). Stress activated p53-controlled transactivation of target genes is an essential feature of each stress response. p53 is a sequence-specific DNA binding protein which contains a typical DNA binding domain and transactivation domain. As a transcription factor that regulates a broad range of target genes, p53 requires a complicated network to control the precise responses to the various stress signlas encounted by cells. p53 is mainly regulated at post-transcriptional levels by an array of post-transcriptional modifications or associating with other binding partners. Based on the recent proposed model of p53 regulation, the activation of p53

Page 22: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

9

can be devided into three key steps consisting of p53 stabilization, derepression and promoter-specific activation (Kruse and Gu, 2009). In unstressed cells, p53 is bound by an E3 ubiqutin ligase, Mdm2, which promotes its rapid proteosomal degradation. Many stress pathways can lead to the stabilization of p53 through affecting the ability of Mdm2 to ubiquitinate p53. Although p53 might have intrinsic transactivation abilities, the stabilized p53 still remains inactive due to the repression by its key negative regulator, Mdm2 and MdmX at promoters. The release of p53 from the repression mediated by Mdm2 and MdmX requires the modifications of p53 or recruitment of other facotors to p53. For full activation of p53 on some specific promoters, p53 gets further modifications and interacts with numberous cofactors regulating the transcription of specific groups of p53 target genes for different cellular stress responses (Kruse and Gu, 2009).

Once p53 becomes activated by stress signals such as DNA damage, a complex signaling network is enaged to result in a long-term cell fate decision. In all cases, initial processing of the DNA damge immediately activates p53 controlled cell cycle checkpoints leading to transient cell growth arrest. p53 physically localizes to sites of DNA damage to promote repair and simultaneously induces the transcription of effectors for cell growth arrest and efficient DNA repair. If the DNA damage is transient and limited, it is rapid repaired and p53 then return to the inactive states. If the DNA damage is highe or is continuous, p53 activation can be extendent and induced more permanent outcomes including senescence and apoptosis depending on cell type and physiological context. Senescence is a complex final fate decision that estabilishes permant cell cycle arrest. In most cell types, p53 is crucial for initiating or maintaining senescence programs. However, the exact downstream events of p53 activaiton in senescence response are not completely understood probably involving in both genetic and epigenetic regulations. In severe damage condition, the activation of p53 may induce apoptosis rather than senescence by upregulating the pro-apoptotic genes and downregulating the anti-apoptotic genes. Both senescence and apoptosis are potent tumor suppressor mechanism in organism by emiliminating the stressed or damaged cells with high risk giving rise to cancerous cells. Through these various mechanisms, p53 considered as one of the most important and powerful tumor suppressor genes, imposed an important barrier againise tumor development. Moreover, the irreversibly removal of cells from tissues can also deplete stem cells or progenitor cells from tissue or change the microenviroment of tissues contributing to organ degeneration and ageing. p53 is the important node to control the balance between the beneficial effects of tumor suppression and the detrimental effects of tissue degeneration. Genetic engineered mouse models have provided evidence to support the profound impacts of p53 in the longevity. The activaion of p53 reduces the somatic mutations and removes the damage and stressed cells to prevent early death through cancer and promote the longevity. In the mean time, continueous chronic activation of p53 has a deleterious effect on ageing processes by declining in stem/progenitor cells self-renewal and differentiation through its anti-proliferative effects. SnoN which can activate p53 by interacting with the PML-p53 pathway can also regulate p53 effects in tumorigenesis and ageing processes.

,

Page 23: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

10

Chapter 2: Materials and methods

Page 24: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

11

Material and method

Knock-in Mice

The target vector was introduced into the E14 mouse embryonic stem (ES) cells derived from 129P2/OlaHsd mice followed by selection of G418 to get the positive ES cells. After confirmed by southern blotting, positive ES cells were injected into the blastocysts of C57BL/6 background. The chimera were crossbred with a CMV (Cytomegalovirus immediate-early promoter)-cre transgenic mice to generate the knock-in mice and backcrossed to C57BL/6 for additional 5 generations. Knockout SnoN mice in C57BL/6 background were obtained from Dr. S. Pearson-White. Knockout p53 mice were obtained from The Jackson Laboratory.

Isolation of MEFs (Mouse embryonic fibroblast)

MEFs are primary cells which have finite growth ability usually about 10 passages but dependent on how healthy the cells are when they are isolated. MEFs were cultured in 5% CO2 with DMEM containing 10% FBS and antibiotics (During isolation process, you can add fungizone to prevent contamination but do not add it in the regular culture medium).

Basically, the pregnant female mice are scarified at E13.5. The uterine horns are dissected out and briefly rinsed in 70% ethanol. Then, they are put into dishes with sterilized PBS in the culture hood. Separate each embryo from its placenta and remove the heads and all red organs (Keep the head for the genotyping). Tear the remaining embryos into small pipettable pieces by using tweezers and transfers the pieces into l ml of 0.1% trpsin in eppendorf tubes. Incubate the embryos for 25 min at 37°C. After incubation, the suspension is pipetted 4-5 times. Ideally, the suspension should be free of visiable larger pieces of tissue. Then, the cells are transferred into 15 ml of tube filled with 9 ml of culture medium. Let the remaining pieces of tissue settle down to the bottom of the tube within 2 min and carefully take off the supernatant to spin down the released cells. After resuspend the cell pellet in culture medium, they are plated out at 1 embryo per 10 cm dish (This is passage No. 0). When cells become confluent (normorlly in 1-2 days), 106 of MEFs were frozen down in one tubes. They can be thawed and placed out in 50 cm dish (passage No. 1) when needed. (Serrano et al., 1997).

Population doubling curve:

3T9 protocol (3x105 cells passaged every 3 day) was used to compare the population doubling number at different passages (Marı´a A. Blasco Et, 1997). Basically, primary MEFs were placed out in 50 cm dishes at a density of 106 of cells (5 dishes for each line to the statistic numbers). Every 3 days, the cell number is counted and recorded. Then, the same amount of cells (106) is replated. Repeat passageing cells and cells eventually stop growing entering senescence (10 passages for wt MEFs). Please continue to passage the cells even they are senescent until they become immortalized. The population doubling number for each passage is based on the formula of PDn=PDn-1+Ln NOn/NOn-1, (Assume PD 1=2).

Page 25: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

12

Other cells, antibodies, and constructs

293T and RIE cells were maintained in DMEM with 10% serum. Hep3B cells were maintained in MEM with 10% serum. Primary mammary gland epithelial cells were obtained from Dr. Martha Stampfer. An anti-SnoN rabbit polyclonal antibody was raised against SnoN N-terminus fragment. Antibodies for Flag and HA were purchased from Sigma; antibodies for p53 (ab26); p19Arf (ab 80) from Abcam; antibody for mouse PML (Mab 3738) from Chemicon; anti-body for acetyl-p53 from Upstate; antibodies for phospho-ATM and phospho-Chk1 from Cell Signaling; antibodies for p16Ink4a, Samd3 (FL245), Mdm2 and Tubulin from Santa Cruz biotechonology; antibody for p21 (ab 5) from Oncogene Science; antibody for cleaved Caspase 3 from abcam. shRNA was designed based on the retrovirus vector of pSuper-Retro-puro (Oligoengine) for infection. shSmad3, was synthesized following the sequence of . siRNAs for mouse Smad2 or Smad3 was purchased from DHARMACON and transfected into MEFs by Nucleofector system. shRNA for p19Arf was designed based on the sequence of 5’ caccggaatcctggaccag 3’. shRNA for p53 was based on the sequence of 5’gtactctcctcccctcaat 3’; shRNA for murine PML was based on 5’ cccacttagaagacaggac3’ ; shRNA for human PML was based on 5’ ggaggagttccagtttctg 3’; shRNA for SnoN was based on 5’ctccattctgcagaggaag 3’

Transfection and Luciferase

293T Hep3B and MEF cells were transiently transfected using Lipofectamine and Plus (Invitrogen) following manufacturer's recommendations. In the luciferase assay for TGFβ induced transcription, 0.5 μg of p3TP-Lux was co-transfected with or without 1 μg of Flag-SnoN or Flag-mSnoN or Flag-SnoN Δ322-366 or Flag-SnoN Δ255-258. Luciferase activity was measured 16 hr after stimulation with 50 pm TGFβ1. 0.5 μg of p21-Lux and 0.5 μg of Flag-p53 were contranfected with or without 1 μg of Flag-SnoN in presence or absence of 0.5 μg of Flag-Mdm2 or His-PML. For H2O2 induced transcription, 0.3 μg of pSnoN-Lux was transfected into MEFs by using a tranfection condition similar as Hep3B cells. Luciferase activity was measured 24hr after stimulation with 70 μM of H2O2.

Immunoprecipitation and Western Blotting

Flag tagged SnoN variants were transfected into 293T cells with Histidine tagged PML. SnoN was isolated from transfected 293T by immunoprecipitation with anti-Flag beads followed by elution with the Flag peptide and analyzed by Western blotting as described previously. Endogenous proteins were isolated by immunoprecipitation with 30μg of antibodies.

Nuclear extraction of MEFs

Attached cells were treated with 0.1% trypsin at 37℃ for 10 min. Scrape off and pellet cells by spinning at 4,000rpm in the microfuge for 2 min. Pellet cells were resuspended in PBS and transfer to eppendorf tube. Spin cells again in eppendorf tube at 4,000 rpm for 2 min. The packed cell volume (PCV) was recorded. Add 5 times of PCV of BufferA (10mM Hepes pH7.9;

Page 26: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

13

1.5mM MgCl2; 10mM KCl; 1mM DTT; 0.5 mM PMSF) to resuspend the cell pellet and leave the samples on ice for 15 min. Then, the cells were spinned down at 3,000rpm for 8 min. After removal of the supernatant, resuspend pellet with 2 times of PCV of Buffer A followed by douncing cells for 20 times with 1ml syringe (25G 5/8 needle). Monitor the breakage of cells under a microscopy to ensure the release of nuclei from most of cells. Spin down the nucleis at 3,000rpm for 10 min. After removal of the cytoplasmic fraction, nuclei is lyzed in IP lysis buffer (50mM Hepes pH7.8; 5mM EDTA; 150mM NaCl; 1% NP40) in order to run endogenous SnoN IP.

Pulse Chase Assay

Transfected 293T cells growing in 6-well plates were washed with 2 ml of DMEM lacking Met and Cys, pulsed with 0.4 mCi/ml 35S-express (Roche) for 30 min and chased for different periods of time. Lyse the labeled cells and immunoprecipitate the target proteins. The proteins are isolated SDS-PAGE and detected by radioautography.

RT-PCR

Total RNA extraction, cDNA preparation and PCR were performed using the standard protocol. The primers used to amplify the different genes by PCR are: p53: 5’actgccatggaggagtcac3’/5’tcagtctgagtcaggcccc3’; PML: 5’agcaggaggcttctcagacagt3’/ 5’cttgatgatcttcctggagcaa3’; p19ARF: 5’gttttcttggtgaagttcgtgc3’/5’tcatcacctggtccaggattc3’

GST pull-down assay

Recombinant p53 was expressed in and purified from E. coli as GST fusion proteins. Recombinant SnoN and Mdm2 were similarly expressed as GST fusion proteins followed by Thrombin cleavage. For binding experiments, 1.5 μg of GST-p53 immobilized on the glutothione Sepharose were incubated with 2 μg of recombinant SnoN for 1 hour at 4℃. After intensive washing, the proteins were eluted by boiling with SDS-PAGE sample buffer. For the competition experiments, 1.5 μg of GST-p53 immobilized on the glutothione Sepharose were incubated with indicated amounts of recombinant SnoN or Mdm2. After extensive washing, the proteins were eluted by boiling in SDS-PAGE sample buffer and detected by antibodies or visualized by coomasie staining.

SA-β-gal staining in cells and tissues

The senescence detection kit (Calbiochem) was implied stain senescent cells following the instruction. Basically, cells growing on 12-well plate are washed once with 1ml of PBS. Then, cells are fixed with 0.5ml of fixative solution provided in the kit. Wash out the fixative solution with 1 ml of PBS and add 0.5 ml of the staining solution (fresh made, X-gal) provided in the kit to each wells. Incubate at 37℃ in a humid condition for 16-24 hrs. Visualize and count cells

Page 27: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

14

under a microscope. To store the stained plates for a long-term at 4℃, remove the staining solution and overlay the cells with 70% glycerol.

Senescence staining can be carried out in frozen tissue section. Fresh tissues are embedded in OCT compound in cryomolds and frozen on dry ice. Store frozen blocks at -80℃.

Cut 6μM thick cryostat sections and mount on superfrost plus slides or gelatin coated slides. Store slides at -80℃ until staining. Before staining, warm slides at room temperature for 30 min. The tissue sections are stained for SA-β-gal on the slides following the same protocol as cells. The fixative and staining solution can be overlaid on the slides in order to cover the sections. After SA-β-gal staining, the samples are counter-staining with Eosin. Basically, wash the stained slides with PBS for 3 times and then immerse them into 0.5% Eosin (in 95% Ethanol) for 2 min. Dip in water for several times until the eosin stops streaking. Wash the slides by serial concentrations of Ethanol (50%, 70%, 95% and 100%) and finish the washing in Xylene. Mount and coverslip the sample to visualize under a microscope.

Tunel staining in frozen sections

DeadEnd TUNEL system (Promega) was implied to label the apoptotic cells in tissues. Warm and dry the 6μM thick cryostatic tissue sections at room temperature for 30 min. Fix samples in 3.7% formaldehyde solution for 10 min. Then, wash the slides 2 times for 5min in PBS. Place 50μl of protein K solution onto each sample and incubate at room temperature for 15 min. After permeabilization, wash the slides twice for 2 min in PBS and then place 50μl of labeling reaction mix provided in the kit onto each samples to label cells for 1 hr at 37℃. After labeling, transfer the slides into TdT stop buffer provided in the kit for 5 min and wash 2 times in PBS for 2 min each. Then, the samples are stained with DAPI and visualized under a fluorescent microscope.

Retrovirus replication and infection

Retrovirus vector was transfected into phoenix ecotropic or amphotropic cells to generate the high titer of virus as described previously (Zhu et al., 2005). The virus supernatants are used to incubate with proliferating MEFs at P3 in presence of 8μg/ml of polybrene. (If second virus need to be introduced to cells, the virus supernatant is implied to the cells after 48-hour infection.) 48 hours later, infected cells can be used directly to soft agar assay or be splitted and selected by 2 μg/ml puromycin for 2 days or 400μg/ml of G418 for 4 days. The survival cells are used for further analysis.

Immunofluorescence

MEFs were grown on glass cover-slips, fixed with ice-cold methanol for 20 min, permeabilized with 0.1% Triton X-100 for 5mins and stained with the appropriate primary and secondary antibodies. Nuclei were detected by 4,6-diamidino-2-phenylindole (DAPI) staining. Cells were visualized by a Zeiss 510 confocal microscope. Immunofluorescence staining can be

Page 28: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

15

carried out in 6μM thickness of frozen tissue sections on coated slides following the same protocol except the permeabilization with 0.5% Triton X-100 for 15 min.

H&E (Haematoxylin and Eosin) staining for frozen tissue sections

Warm and dry the frozen sections at room temperature for 30 min. Stain the sections with 0.1% Mayer Hematoxylin for 10 min. After staining, wash the slides in water for 5 times and then immerse them into 0.5% Eosin (in 95% Ethanol) for 2 min. Dip in water for several times until the eosin stops streaking. Wash the slides by serial concentrations of Ethanol (50%, 70%, 95% and 100%) and finish the washing in Xylene. Mount and coverslip the samples to visualize them under a microscope.

Chromatin immunoprecipitation (ChIP) assay

Cells were cross-linked with formaldehyde for 10 min and stopped by the addition of glycine to a final concentration of 0.125 M. Then, plates were rinsed with PBS, incubated with

Tripsin and scraped to harvest cells. Cells were collected by centrifugation and washed in PBS with protease inhibitors. Pellets were resuspended in 3 ml of ChIP lysis buffer (5 mM PIPES pH 8.0, 85 mM KCl, 0.5% NP-40, 0.5 mM PMSF, and 5 l of aprotinin  per ml) and incubated on ice for 20 min. Then, cells were Dounce through 18G needle for 20 times. Nuclei were collected by microcentrifugation at 5,000 rpm, suspended in 1 ml of sonication buffer (1% sodium dodecyl sulfate [SDS], 10 mM EDTA, 50 mM Tris-HCl [pH 8.1], 0.5 mM PMSF, and 5 l of aprotinin  per ml), and incubated on ice for 10 min. Samples were sonicated with an Ultrasonics sonicator at full power for three 30-s pulses on ice to an average length of 200 to 600 bp and then microcentrifuged at 14,000 rpm. The chromatin solution was precleared with the addition of protein A beads for 30 min at 4°C. Precleared chromatin was incubated with protein A beads coated with antibodies for SnoN or p53 or Flag or no antibody and rotated at 4°C for 12 h. After extensive washing, the complexes were eluted with elution buffer (50mM NaHCO3, 1% SDS). Associated DNA were extracted by phenol/chloroform and solved in 30ml of TE buffer. The primer pairs used to amplify RE1 regions of p21 promoters are 5’ cttatatctcccttggtcc 3’ and 5’ tattctgctggcaaagtggg 3’; for SBE regions of p21 promoter, the primers are 5’ acaggatgaggcttttgagg 3’ and 5’ cggcatacatctgtaatccc 3’

BrdU incorporation assay in attached cells

BrdU labeling and detection kit (Roche) was implied to stain the proliferating cells. Cells growing on cover slips were incubated with medium containing 0.1mg/ml BrdU for 6 hours. Aspirate cell the BrdU labeling medium and wash the cover slips three times in washing buffer provided in the kit. After fixation in ice-cold methanol for 20 min at -20°C and then washed in washing buffer, cells were incubated with anti-BrdU working solution provided in kit for 30 min at 37°C. Wash cells with washing solution for 3 times ant then stain cells with Alexa Fluor 488-conjugated goat anti-mouse-IgG (Invitrogen). After washed with washing buffer for 3 times, cells were stained with DAPI for 5 mins and visualized by fluorescent microscopy.

Page 29: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

16

Soft-agar assay

3 ml of growth medium containing 0.66% Bacto-agar was added to a 50mm dish for the bottom agar layer. 6x103 MEFs were resuspended in 2 ml medium containing 0.4% agar and overlaid on the hardened bottom layer. 2ml of fresh medium containing 0.4% agar was added to the dish once a week for 4 weeks. The colonies were visualized by staining with 0.5 mg/ml 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) (Sigma) in PBS for 4 hrs at 37°C. After washing with PBS to remove the color solution, colonies were counted and pictured under a microscopy.

Skin carcinogenesis- DMBA/TPA

The dorsal skins of 8-week-old mice were shaved and spread with 7,12-dimethylbenz(a)anthracene (DMBA; 100 μg/200 μl in acetone). One week later, 12-O-tetradecanoyl-phorbol-13-acetate (TPA; 20 nmol/200 μl in acetone) was applied twice weekly to the same area for 30 weeks (Balmain et al., 1988). Numbers and sizes of tumors growing on the treated mice were recorded every other week. Mice were euthanized at the end of 30 weeks. Surgically removed tumors were immediately snap-frozen in OCT solution, and stored at -80°C for senescence assays. Alternatively, dissected skin and tumor samples were fixed in 10% neutral-buffered formalin at 4°C overnight, embedded in paraffin, sectioned to 6-µm thickness, and stained with hematoxylin and eosin (H&E). A small piece of tumor samples was minced and boiled in SDS-PAGE sample buffer to analyze protein levels.

Page 30: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

17

Chapter3: SnoN functions as a tumor suppressor by inducing premature senescence

(Published paper: EMBO J. 2009 Nov 18;28(22):3500-13. 2009 Sep 10; PMID: 19745809;

http://www.nature.com/emboj/journal/v28/n22/full/emboj2009250a.html)

Page 31: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

18

Abstract

SnoN represses TGFβ signaling to promote cell proliferation and has been defined as a proto-oncogene partly due to its elevated expression in many human cancer cells. Although the anti-tumorigenic activity of SnoN has been suggested, the molecular basis for this has not been defined. We showed here that high levels of SnoN exert anti-oncogenic activity by inducing senescence. SnoN interacts with the Promyelocytic leukemia (PML) protein and is recruited to the PML nuclear bodies where it stabilizes p53, leading to premature senescence. Furthermore, overexpression of SnoN inhibits oncogenic transformation induced by Ras and Myc in vitro and significantly blocks papilloma development in vivo in a carcinogen-induced skin tumorigenesis model. The few papillomas that were developed displayed high levels of senescence and spontaneously regressed. Our study has revealed a novel Smad-independent pathway of SnoN function that mediates its anti-oncogenic activity.

Page 32: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

19

Introduction

Tumor development involves inactivation of tumor suppressors and activation of proto-oncogenes. Some oncogenes have been shown to also possess anti-oncogenic activities, which makes it difficult to design targeted drugs. SnoN is a member of the Ski family of proteins identified based on sequence homology with v-Ski, the transforming protein of Sloan Kettering virus (Nomura et al., 1989; Pearson-White, 1993). It is expressed ubiquitously in most cell types with a relatively high level of expression in the embryos but at a lower level in adult cells (Pelzer et al., 1996). It was classified as an oncoprotein based on its transformation ability when overexpressed in chicken or quail embryo fibroblast (Boyer et al., 1993). The role of SnoN in mammalian tumorigenesis, however, is much more complex and even controversial. While elevation in SnoN expression is often detected in many human carcinoma tissues and cell lines due either to gene amplification, transcriptional activation or enhanced protein stability (Bravou et al., 2009; Buess et al., 2004; Guzman-Ayala et al., 2009; Imoto et al., 2001; Zhang et al., 2003), (Edmiston et al., 2005; Zhu et al., 2006), downregulation of SnoN expression has also been observed in human cancers. In tissue samples from patients with Barrett's esophagus, a precancerous condition that associates with an increased risk of adenocarcinoma, SnoN was found at high levels in low grade dysplasia but absent in high grade dysplasia or adenocarcinoma (Villanacci et al., 2008). In colorectal cancer with microsatellite instability, SnoN is downregulated in 39% of samples and upregulated in 33% samples (Chia et al., 2006). These observations suggest that high levels of SnoN, while beneficial to certain stages of tumor development, may not always be advantageous for malignant progression. More importantly, these studies enforce the hypothesis that SnoN may possess both pro-oncogenic and anti-oncogenic activities in mammalian carcinogenesis. The strongest support for a pro-oncogenic activity of SnoN comes from the observation that reducing SnoN expression in human lung and breast cancer cells inhibits cancer cell growth in vitro and in vivo (Zhu et al., 2006). However, other studies also supported an anti-oncogenic activity of SnoN. Loss of one allele of SnoN in mice results in a slightly higher incidence of tumor formation and increased susceptibility to chemical carcinogen-induced tumorigenesis (Shinagawa et al., 2000), and reducing SnoN expression in human lung and breast cancer cells enhances EMT and tumor metastasis in vivo (Zhu et al., 2006). In Drosophila, overexpressed dSnoN inhibits cell growth (Ramel et al., 2007). Finally, SnoN can cooperate with p53 in silencing of the alpha-fetoprotein gene, which is aberrantly overexpressed in liver cancer cells (Wilkinson et al., 2008). The mechanisms by which SnoN exerts these differential effects on malignant progression have not been defined.

SnoN is an important negative regulator of TGFβ signaling. TGFβ is a pluripotent cytokine that regulates many cellular processes including cell proliferation, survival, cell-matrix interaction and differentiation (Schilling et al., 2008). Smad proteins are critical mediators of many TGFβ-induced signals. Upon binding of TGFβ to its receptor complex, the receptor-activated Smads (R-Smad), Smad2 and Smad3 become phosphorylated by the active receptor kinase complex, form a heteromeric complex with Smad4 and translocate into the nucleus. In the nucleus, the Smad heteromeric complex binds to TGFβ responsive promoters either directly or through association with other sequence-specific transcriptional factors and regulate the expression of TGFβ responsive genes (Feng and Derynck, 2005), (Shi and Massague, 2003), (Moustakas et al., 2001). We and others have shown that SnoN interacts with the Smad proteins and represses their transactivation activity through disrupting the functional heteromeric Smad

Page 33: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

20

complexes, recruiting transcription co-repressor complex and blocking the binding of transcriptional co-activators to the Smads, (Stroschein et al., 1999), (Sun et al., 1999b), (Wu et al., 2002). Through these, SnoN acts as a co-repressor of the Smad proteins. Indeed, overexpression of SnoN blocks the growth inhibitory responses to TGFβ. This ability to antagonize TGFβ signaling may be responsible for the oncogenic activity of SnoN at early stages of tumorigenesis (He et al., 2003).

While SnoN represses the activity of the Smads, its own expression is also tightly regulated by the R-Smads. Shortly after TGFβ stimulation, R-Smads bind to SnoN and recruit various E3 ubiquitin ligases including the anaphase promoting complex (APC/C), Smurf2 or Arkadia to SnoN to induce its polyubiquitinylation and degradation (Stroschein et al., 2001), (Wan et al., 2001), (Bonni et al., 2001), (Nagano et al., 2007; Stroschein et al., 1999), (Levy et al., 2007), allowing activation of Smad-mediated transcription. Interestingly, the snoN gene itself is a transcription target of the Smads and its expression is upregulated 2hr after TGFβ stimulation (Stroschein et al., 1999), (Zhu et al., 2005). This later increase in SnoN expression may turn off TGFβ signaling in a negative feedback manner or regulate cell proliferation and differentiation in a TGFβ-independent manner (Zhu et al., 2005).

To elucidate the physiological function of the SnoN-Smad interaction, we generated knock-in mice substituting the endogenous snoN gene with a mutant deficient in binding to both the R-Smads and Smad4. Mice expressing the mSnoN gene are resistant to chemical carcinogen-induced tumorigenesis due probably to the accumulation of senescence cells in tumors. Accordingly, mouse embryonic fibroblasts (MEF) prepared from the knock-in mice also exhibit premature senescence. We showed here that the ability of SnoN to promote premature senescence is dependent on p53 and PML proteins and functions to block oncogenic transformation in vitro and tumor development in vivo. Our study therefore revealed a novel function of SnoN in promoting senescence and provided a potential mechanism to understand the tumor suppressor functions of SnoN.

Page 34: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

21

Results

Generation of knock-in mice expressing a mutant SnoN

To understand the functions of the SnoN/Smad interaction, we generated a knock-in mouse replacing the endogenous snoN gene with a snoN mutant (mSnoN) containing point mutations that alter amino acid residues 88-92 and 267-277 to alanine via homologous recombination. These mutations disrupt the interaction of SnoN with both R-Smads and Smad4 and abolish the ability of SnoN to repress TGFβ signaling (Wu et al., 2002), (He et al., 2003), (Zhu et al., 2006). The mSnoN gene can be distinguished from the WT allele by the introduction of the Swa I and Sph I restriction enzyme cleavage sites with the point mutations (Fig. 1A). Mice carrying a targeted allele of mutant exon 1 were crossed with a CMV-Cre transgenic mice to yield the knock-in mice. Expression of the knock-in allele was confirmed by isolating the exon 1 of the snoN gene from the genomic DNA by PCR followed by Sph I digestion (Fig. 1B). While WT snoN gene yielded a 1 kb fragment, the mutant allele produced a 0.8 kb fragment. Since it is not possible to distinguish the WT and mutant SnoN protein by western blotting, we resorted to functional assays to confirm the expression of mSnoN. MEF from multiple E13.5 embryos were derived from both the knock-in mice and WT littermates and subjected to various assays to measure TGFβ responsiveness. As expected, the homozygous mutant (m/m) MEF cells showed increased transcription responses to TGFβ in a luciferase reporter assay (Fig. 1C) and were more sensitive to TGFβ-induced growth arrest (Fig. 1D), consistent with the elevated Smad activity in m/m MEF due to the lack of antagonism by SnoN.

Knock-in mice are resistant to carcinogen-induced skin tumorigenesis

Among the pups that were born, 15.7% were homozygous for the knock-in allele, 52.4% were heterozygous and 31.9% WT, indicating that approximately 37.2% of the homozygous embryos died before birth and 62.8% survived. The survived homozygous mice lived up to 24 months without any apparent defects. No increase in spontaneous tumor development was observed in these mice for up to 24 months (data not shown).

To determine whether the knock-in mice are more or less susceptible to chemical-induced carcinogenesis, a two-step skin tumorigenesis protocol was employed (Balmain et al., 1988) (Fig. 2A). 8-week old m/m or WT mice were administered with one dose of DMBA followed by twice weekly treatment of TPA for 30 weeks. Development of papilloma was monitored for 30 weeks. Under this regime, papilloma started to be detected at 14-15 weeks after the initial DMBA treatment in both +/+ and m/m mice. However, while more than 80% of +/+ mice developed tumor by 30 weeks, less than 40% of m/m mice did (Fig 2B). Of those m/m mice that developed tumors, the average number of papillomas per mouse was significantly reduced (Fig 2C). More importantly, most tumors in m/m mice ceased growth after only a short period of time and spontaneously regressed, and few reached a size larger than 2mm in diameter while papillomas in WT mice continued to grow to larger than 10mm (Fig 2D). This strongly suggests that mSnoN blocked papilloma development in vivo.

Cellular senescence is a permanent non-proliferative state that can be triggered by telomere shortening or accumulation of physiological stress (Hayflick, 1965), (Campisi, 2001), (Collado et al., 2007). It has been shown to be an important tumor suppressive mechanism in

Page 35: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

22

mouse models of human cancer (Chen et al., 2005), (Braig et al., 2005), (Michaloglou et al., 2005), (Xue et al., 2007) and can cause tumor regression. To determine whether papillomas from the m/m mice exhibited increased senescence, sections from papilloma of 2 mm in diameter harvested from WT or m/m mice were subjected to H&E analysis and staining of senescence markers. Papillomas from these mice were indistinguishable histologically from each other (Fig. 2E) and exhibited similar levels of phospho-ERK (Fig. 2F), indicating that mSnoN did not affect Ras signaling. However, while no senescence associated β-gal (SA-β-gal) staining could be detected in WT papillomas, they were readily observed in m/m papillomas (Fig 2G). In addition, the expression of another senescence maker, p19ARF was also detected in the nucleolus in m/m tumors but not in +/+ tumors (Fig. 2H). Consistent with this, p53, but not p16INK4a expression was significant increased in m/m tumors (Fig. 2F). Thus, expression of mSnoN induces senescence in tumor tissues, and this may contribute to the resistance of the m/m mice to chemical carcinogen-induced tumorigenesis.

MEF expressing mutant SnoN displayed premature senescence

To determine the molecular mechanism underlying the senescence phenotype, we isolated primary MEF cells from WT and m/m mice. During the initial passages, the m/m MEF were indistinguishable from the WT MEF in morphology and growth under the normal serum concentration. When cultured by a 3T9 protocol, WT MEF cells gradually lost their growth capability and entered senescence around passage 10 (P10) (Todaro and Green, 1963), (Sherr and DePinho, 2000). The cells then remained in senescence for another 8 passages until a small group of cells became spontaneously immortalized at around P18 (Fig. 3A). The m/m MEF proliferated and incorporated BrdU at a similar rate as WT MEF during the first 3 passages (Fig. 3A-B). The BrdU incorporation by the m/m MEF decreased gradually after P4 and by P6, more than 80% of the cell population was negative for BrdU (Fig. 3B). At this passage, more than 80% of m/m MEF entered senescence prematurely (Fig. 3A) and were positive for SA-β-Gal staining (Fig 3C). These m/m MEF stayed in senescence for a longer duration and only became immortalized after P25. This premature senescence was observed for all independently established m/m MEF lines (data not shown).

Premature senescence is caused by the elevated SnoN expression

The premature senescence found in m/m MEF could be attributed to either the increased Smad activity due to the lack of repression by mSnoN or the elevated expression of mSnoN. Previously we have shown that interaction of SnoN with R-Smads results in polyubiquitination and degradation of SnoN (Stroschein et al., 1999), (Sun et al., 1999b), (Stroschein et al., 2001), (Bonni et al., 2001), (Nagano et al., 2007), (Levy et al., 2007). mSnoN defective in Smad binding is not polyubiquitinated (Stroschein et al., 2001) and as a result, accumulated to a higher level in the m/m MEF at P6 (Fig. 4A).

To determine whether elevated Smad activity might be responsible for the observed premature senescence, we first prepared MEF from snoN knockout (-/-) embryos and subjected them to the 3T9 protocol. The -/- MEF also display enhanced Smad signaling but do not express the SnoN protein. Interestingly, the -/- MEF did not exhibit premature senescence at P6 (Fig. 4B). Rather, they displayed a slightly reduced senescence than WT MEF even at P13. The lack of premature senescence in the -/- MEF is not due to any defects in the senescence machinery

Page 36: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

23

because they still respond to UV- and TGFβ-induced senescence (data not shown). Thus, elevated Smad signaling is not sufficient to cause premature senescence.

To directly confirm that increased Smad signaling does not induce premature senescence, a siRNA mixture targeting both mouse Smad2 and Smad3 was introduced into the WT or m/m MEF at P4 by transfection. The transfected cells were then examined for senescence at P6. As shown in Fig. 4C, the siRNA mixture effectively reduced the expression of Smad2 and partially decreased the level of Smad3. Consistent with this, the expression of Smad7, a transcription target of Smad2 and Smad3, was significantly reduced (Fig. 4C). However, premature senescence in the m/m MEF still occurred efficiently (Fig. 4D), suggesting that elevated Smad activity is not responsible for premature senescence. Similarly, introduction of a shRNA specific for mouse Smad3 into m/m MEF by retroviral infection effectively reduced the expression of Smad3 and its target Smad7 (Fig. 4E), but did not affect the senescence of these cells at P6 (Fig. 4F). Finally, treatment of m/m MEF with SB431542, a pharmacological inhibitor of type I TGFβ receptor, effectively blocked Smad3 phosphorylation and activation but did not affect premature senescence (Supplemental Fig. 1). Taken together, our data suggested that elevated Smad activity in the m/m MEF is not responsible for the observed premature senescence.

To determine whether the elevated mSnoN expression is responsible for premature senescence, we introduced shRNA for murine SnoN into the m/m MEF. As shown in Fig. 4G and 4H, when SnoN level was reduced by more than 80%, premature senescence was blocked significantly with less than 10% SA-β-gal positive cells. This suggests that senescence of MEF is sensitive to the expression level of SnoN. Consistent with this, ectopic overexpression of WT SnoN or mSnoN in WT MEF induced premature senescence (Fig. 4G-H and Fig. 6I). Thus, elevated SnoN expression in the m/m MEF, independent of its ability to antagonize Smad signaling, is responsible for the observed premature senescence.

p53 upregulation is required for SnoN-induced premature senescence

Stress-induced senescence of human fibroblasts is controlled by both p53 and Rb pathways but that of mouse fibroblasts is mostly regulated by the p53 pathway (Wadhwa et al., 2004), (Campisi, 2005), (Rodier et al., 2007), (Courtois-Cox et al., 2008)). Inactivation of p53 either by homologous deletion (p53-/-) or by deleting p19ARF (p19ARF-/-) in MEF bypasses senescence and results in spontaneous immortalization but p16INK4A-/- MEF undergo senescence in the same manner as WT MEF (Sharpless et al., 2001). To determine which pathways are involved in SnoN-induced senescence, we compared the expression of p53 and p16INK4A at passages representing pre-senescence (P4), m/m senescence (P6), senescence for both (P13) and after immortalization (Im) between WT and m/m MEF. At the pre-senescence stage, both proteins were maintained at an undetectable level. Interestingly, at P6 when most m/m MEF have entered senescence prematurely, the level of p53 was significantly elevated in m/m MEF but to a much lesser extent in WT MEF, most of which have not entered senescence (Fig. 5A). The elevated p53 expression was maintained through senescence and returned to the background level upon immortalization. Thus the profile of p53 expression correlated well with the senescence status of m/m and WT MEF. In contrast, the level of p16INK4A did not change appreciably during this process, nor did Rb phosphorylation (at residues 780 and 795) and expression (Fig. 5A). Thus, p53 appears to be the major regulator in SnoN-induced premature senescence. This is consistent with our earlier observation that p53 but not p16Ink4a was upregulated in the tumor samples derived from the m/m mice (Fig. 2F).

Page 37: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

24

The increase in p53 level is not due to an increase in transcription (data not shown), but most likely due to stabilization of the p53 protein. p53 stability can be regulated by many proteins such as p19ARF, ATM/ATR and Chk1/Chk2 kinases (Efeyan and Serrano, 2007), (Rodier et al., 2007). While the activities of ATM/ATR and Chk1/Chk2 as represented by the phosphorylation of these proteins did not change in either m/m or WT MEF, the expression of p19ARF was elevated as soon as the m/m MEF entered senescence at P6 and remained high thereafter (Fig. 5A). This increase in p19ARF expression occurred at the transcription level as detected by RT-PCR (data not shown).

To determine whether the increased p19ARF and p53 levels are necessary for SnoN-induced senescence, we introduced shRNA for p19ARF or p53 into m/m MEF by retroviral infection at P3 and examined senescence at P6. An efficient knockdown of p53 (Fig. 5B) effectively blocked premature senescence of the m/m MEF at P6 (Fig. 5C), confirming that p53 is indeed a critical mediator of SnoN-induced senescence. Surprisingly, reducing p19ARF expression by shRNA did not have any effect on premature senescence (Fig. 5C) or p53 expression even though more than 90% of p19ARF was eliminated by the shRNA (Fig. 5D). This indicates that although p19ARF is upregulated in the m/m MEF, it is not primarily responsible for the increased p53 expression or the premature senescence of the cells. The upregulation of p19ARF expression most likely occurred as a consequence but not as a cause of the premature senescence.

Taken together, our studies have established p53 as a critical mediator of SnoN-induced premature senescence.

PML mediates SnoN-induced p53 stabilization and premature senescence

To determine how SnoN induces upregulation of p53, we examined the intracellular localization of SnoN before and during senescence. In pre-senescence WT and m/m MEF, endogenous SnoN was distributed throughout the nucleus (Fig. 6A: +/+ MEF at P3 and P6 and m/m MEF at P3). However, upon entry into senescence, SnoN was found to accumulate in numerous small nuclear speckles (Fig 6A: +/+ MEF at P13 or m/m MEF at P6 and P13). These senescence-associated SnoN speckles are reminiscent of Promyelocytic leukemia (PML) nuclear bodies in size and morphology. Indeed, using markers of various nuclear domains, SnoN was found to co-localize only with the PML protein in the PML bodies (Fig 6A), but not in heterochromatic foci or DNA damage/repair foci (data not shown). Localization of SnoN in the PML bodies occurred in both WT and m/m MEF during senescence, but not before senescence or after immortalization (Fig. 6A and data not shown). Finally, in WT MEF that undergo premature senescence due to overexpression of SnoN, SnoN also localized in the PML bodies (Fig. 6B). Thus, accumulation of SnoN in the PML nuclear bodies appears to correlate with the senescence status of MEF.

The PML nuclear bodies are discrete nuclear domains consisting of large number of proteins and have been linked to many fundamental cellular processes including transcriptional control, anti-viral response, DNA repair, apoptosis and senescence (Pearson and Pelicci, 2001), (Salomoni and Pandolfi, 2002). The PML tumor suppressor protein is essential for the formation of the PML nuclear bodies and for recruitment of diverse proteins to the nuclear domain (Goddard et al., 1995), (Seeler and Dejean, 1999), (Dellaire et al., 2006), (Bernardi and Pandolfi, 2007), (Shen et al., 2006). Of particular interest is its role in p53 stabilization in response to

Page 38: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

25

different stimuli. PML bodies may serve as scaffolds to increase the local concentrations of factors implicated in p53 stabilization by enhancing phosphorylation and acetylation or regulating the ubiquitination and proteasome-dependent degradation of p53 (Pearson et al., 2000), (Pearson and Pelicci, 2001), (Bode and Dong, 2004), (Takahashi et al., 2004). Loss of PML allows MEF to bypass senescence due to lack of p53 activation (Pearson et al., 2000), (de Stanchina et al., 2004), (Ferbeyre et al., 2000), (Marcotte and Wang, 2002). Thus, it is possible that SnoN localizes to the PML nuclear bodies to allow stabilization of p53, leading to premature senescence.

To test whether PML is a critical intermediate for SnoN-induced p53 stabilization and premature senescence, we introduced shRNA for PML into the m/m MEF by retroviral infection. Interestingly, efficient reduction of PML expression (Fig. 6C) completely inhibited premature senescence (Fig 6D) and prevented stabilization of p53 in the m/m MEF (Fig 6C). Thus, PML is required for SnoN-induced p53 stabilization and premature senescence.

SnoN physically interacts with PML to promote premature senescence.

Since SnoN is localized in the PML nuclear bodies during senescence, we hypothesized that PML may physically interact with SnoN to recruit it to the PML bodies. To test this, Flag-SnoN was co-transfected with His-PML into the 293T cells and isolated by immunoprecipitation with anti-Flag. The PML protein that is associated with SnoN was then detected by western blotting with anti-PML. As shown in Fig. 6E, PML associated with both WT SnoN and mSnoN to the same extent, indicating that SnoN interacts with PML independently of its ability to antagonize the Smad proteins. This interaction also occurred at the endogenous level in m/m MEF at P6 as endogenous SnoN was found to co-precipitate with PML efficiently (Fig. 6F). In WT MEF, endogenous WT SnoN also interacted with PML but to a much lesser extent, probably due to relatively lower level of SnoN in WT MEF (Fig. 6F).

We next mapped the domain in SnoN that is required for interaction with PML. Various SnoN deletion mutants were co-transfected into 293T cells with PML, and their ability to interact with PML was evaluated by co-immunoprecipitation assay. As shown in Fig. 6G, the amino-terminal 366 amino acids of SnoN encoded by exon 1 was sufficient for binding to PML, while the C-terminal 367-684 fragment failed to bind. Further deletional analysis identified a short region immediately after the SAND-like domain between residues 322-366 as being critical for binding to PML. Deletion of residues 322-366 (SnoNΔ322-366) abolished the SnoN-PML interaction (Fig. 6G), but did not affect the binding of SnoN to Smad4 (data not shown) nor its ability to repress TGFβ-induced transcription (data not shown). This suggests that the deletion did not disrupt the structural integrity of SnoN but specifically block the interaction between SnoN and PML. More importantly, binding of SnoN to PML is independent of the SnoN-Smad interaction and does not interfere with the ability of SnoN to antagonize Smad signaling.

Next we examined the ability of this deletion mutant to be recruited to the PML bodies and to induce p53 stabilization and premature senescence. As shown earlier, ectopic expression of WT SnoN in WT MEF resulted in stabilization of p53, premature senescence and localization of SnoN in the PML bodies. In contrast, ectopic expression of SnoNΔ322-266 did not lead to p53 stabilization and premature senescence (Fig. 6I). Furthermore, this mutant SnoN∆322-366 was distributed throughout the nucleocytoplasm and failed to accumulate in PML bodies (Fig 6J). These results strongly indicate that the interaction between SnoN and PML is critical for the

Page 39: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

26

recruitment of SnoN to the PML bodies and the subsequent p53 stabilization and premature senescence.

PML expression is upregulated in the m/m MEF

During the course of our investigation, we noticed that m/m MEF seemed to express a higher level of PML than WT MEF. This prompted us to compare the expression of PML between WT and m/m MEF. By RT-PCR and western blotting, PML mRNA and protein levels were both increased in the m/m MEF (Fig. 7A and 7B). Interestingly, SnoN is necessary for the upregulation of PML in the m/m MEF. Knocking down SnoN by shRNA significantly decreased the level of PML (Fig 7B).

Relationship between SnoN, PML and p53 during senescence

Despite a higher level of SnoN and PML proteins in the m/m MEF, it still takes 6 passages for the cells to enter senescence. Consistent with this, the level of p53 did not peak until P6 in m/m MEF (Fig. 4A and 7C). To investigate the cause of this delay in entering senescence by the m/m MEF, we examined the expression levels of endogenous SnoN, PML and p53 as well as the interactions among these proteins in WT and m/m MEF cells at P1, P6 and P13. As shown in Figure 4A and 7C, SnoN expression was at a relatively low level in m/m MEF at early passages (P1-P3) and increased with the passage of cells and reached a high level at P6. Correlating with this increase in mSnoN expression, PML and p53 levels were also elevated progressively and reached maximal level at P6 in m/m MEF. In WT MEF, the levels of PML and p53 at P6 were still low and did not peak until P13. Interestingly, PML and p53 levels in WT MEF at P13 were comparable to that in the m/m MEF at P6, suggesting that PML and p53 may need to accumulate to certain levels to induce cells to enter senescence. In these cells that have entered senescence, SnoN and p53 were found to form a complex and co-localize in the PML NBs as shown by both co-immunoprecipitation assay (Fig. 7C) and by immunofluorescence staining (Fig. 7D).

To determine whether the accumulation of mSnoN is required for p53 expression, we examined p53 levels in SnoN knockdown cells by western blotting (Fig. 7B). When SnoN level was reduced, accumulation of PML and p53 in m/m MEF was stalled, indicating that elevation in SnoN expression precedes PML and p53 accumulation. In contrast, when PML expression was reduced by shRNA, SnoN accumulation was not affected, suggesting that SnoN acts upstream of PML (Fig 7E).

Since mSnoN accumulation is a pre-requisite for elevation of PML and p53 expression, and the peak mSnoN level is not reached until P6, senescence of the m/m MEF was not observed until P6. This gradual accumulation of mSnoN is therefore the rate-limiting step in inducing premature senescence and explains why it takes six passages for the m/m MEF to enter senescence. Our data so far suggest a model of how SnoN induces premature senescence (Fig 7F). In cells expressing high levels of SnoN, SnoN not only antagonizes TGFβ signaling, but also upregulates the expression of PML and more importantly, interacts directly with PML. This interaction enables SnoN to be recruited to PML nuclear bodies where it stabilizes p53 and/or other proteins, leading to premature senescence. This ability of SnoN to induce senescence suggests an anti-oncogenic function of SnoN.

Page 40: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

27

High levels of SnoN inhibit transformation of MEF by oncogenes

Since the m/m MEF display enhanced senescence, we wondered whether these cells were more resistant to transformation by oncogenes. It is known that in MEF, while overexpression of an individual oncogene induces senescence (Serrano et al., 1997), (Di Micco et al., 2007), co-expression with another oncogene results in transformation. To assess the transforming ability of m/m MEF, we introduced a constitutively active Ras (Q61L) or c-Myc, either alone or together into the WT or m/m MEF by retroviral infection. After a brief drug selection, the infected cells were evaluated for the ability to undergo anchorage independent growth by a soft-agar assay. When Ras(Q61L) was introduced individually into the WT MEF, it triggered senescence (Fig. 8B) but not transformation (Fig. 8A). However, overexpression of both Ras(Q61L) and Myc together resulted in oncogenic transformation (Fig 8A). In m/m MEF that already overexpressed mSnoN, introduction of Ras(Q61L) or c-Myc alone did not result in transformation (Fig. 8A and data not shown). Rather, Ras(Q61L) triggered a slightly more severe senescence (Fig 8B), suggesting that mSnoN did not function as an oncogene. In the presence of both Ras(Q61L) and c-Myc, m/m MEF displayed a much reduced level of transformation (Fig 8A and enlarged figures in Supplemental Fig. 2). Approximately 30% of these cells still displayed senescence while none of the WT MEF expressing Ras(Q61L) and c-Myc were senescent (Fig. 8B). This suggests that mSnoN inhibits oncogene-induced transformation of MEF. To confirm that this inhibition of transformation is due to the high level expression of mSnoN but not to the enhanced Smad activity, shRNA for SnoN or Smad3 was introduced into the MEF together with Ras (Q61L) and c-Myc. Consistent with the role of SnoN in supporting premature senescence, reducing SnoN level in the m/m MEF restored high level of soft-agar colony formation while reducing Smad3 had no effect (Fig 8A). This ability to inhibit oncogenic transformation is not unique to mSnoN but also displayed by WT SnoN. Overexpression of WT SnoN together with Ras (Q61L) and Myc in WT or m/m MEF significantly reduced the colony formation (Fig 8A). Taken together, these results suggest that SnoN displays anti-tumorigenic activity and inhibits oncogene-induced transformation possibly through inducing senescence.

Blocking the SnoN-PML interaction or abolishing p53 eliminates the anti-oncogenic activity of SnoN

Our results predict that the tumor suppressor activity of SnoN is dependent on the SnoN-PML interaction and on p53. Thus disruption of the SnoN-PML interaction or elimination of p53 should abolish the tumor suppressor activity of SnoN and turn it into a bona fide oncogene. To test this, we first examined the activity of SnoN∆322-366 that is defective in the interaction with PML in the soft-agar colony assay. Indeed, while WT SnoN failed to transform WT MEF together with the active Ras oncogene, SnoN∆322-366 and Ras readily induced anchorage-independent growth as effectively as Ras and Myc (Fig. 8C and enlarged panels in Supplemental Fig. 2). Interestingly, co-expression of SnoN∆322-366 and Myc did not result in transformation (Fig. 8C).

We also examined the ability of WT and mSnoN to induce transformation of p53-/- MEF cells. In these cells, expression of the active Ras alone is sufficient for transformation (Fig. 8D and enlarged panels in Supplemental Fig. 2). Interestingly, expression of WT SnoN or SnoN∆322-366 alone was sufficient for transformation of p53-/- MEF (Fig. 8D), confirming that they function as oncogenes. In contrast, mSnoN failed to induce transformation of p53-/- MEF

Page 41: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

28

(Fig. 8D), most likely because it cannot repress Smad signaling and therefore is defective in the pro-oncogenic branch. Consistent with this idea that repressing Smad activity is critical for the transformation of MEF by SnoN, knocking down both Smad2 and Smad3 by siRNA or Smad3 alone by shRNA was sufficient to induce transformation of MEF when p53 is absent (Fig 8D). Although the extent of transformation induced by reducing Smads was not as strong as overexpression of WT SnoN or SnoN∆322-366, most likely due to the incomplete knockdown of the Smads (Fig 4C), it nevertheless supports the notion that antagonizing Smad activity is critical for the transforming activity of SnoN. Taken together, these results indicate that SnoN possesses pro-oncogenic activity through antagonizing the Smad proteins and anti-oncogenic activity through the PML and p53 proteins. Disruption of the PML-p53 pathway is sufficient to inactivate the tumor suppressor activity of SnoN but preserves the oncogenic function of SnoN.

Page 42: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

29

Discussion

Senescence has been recognized as an anti-tumorigenic mechanism in vitro and in vivo to prevent the accumulation of harmful oncogenic mutations. Here we have shown that elevated SnoN triggers premature senescence through binding to and co-localizing with the PML protein in the PML nuclear bodies. This interaction results in stabilization of p53 and subsequent premature senescence. The ability of SnoN to induce senescence is independent of its ability to bind to and antagonize the Smad proteins but is dependent on high levels of SnoN expression. The premature senescence triggered by SnoN does not involve DNA damage responses but is reminiscent of tumor suppressor-induced senescence. Indeed, MEF cells with high levels of SnoN are less susceptible to transformation by oncogenes. More importantly, the knock-in mice are more resistant to carcinogen-induced skin tumorigenesis possibly due to the induction of senescence in the epidermis. Thus, we have elucidated a novel tumor suppressor activity of SnoN through inducing senescence and revealed the first molecular pathway for this activity.

Although the induction of senescence by SnoN only occurs under conditions of SnoN overexpression, it is nevertheless physiologically relevant. In many human cancer cells, SnoN expression is highly elevated due to amplification of the 3q26 amplicon, increased transcription of the snoN gene or inhibition of SnoN degradation to a level similar to that found in m/m MEF. Since SnoN has been classified as an oncogene in the past, one could argue that SnoN-induced senescence is a typical case of oncogene-induced senescence (Serrano et al., 1997), (Di Micco et al., 2007). However, SnoN induced senescence is different from oncogene-induced senescence in several important aspects. First, senescence induced by oncogenes such as hyperactive Ras requires activation of the INK4A/ARF locus (Serrano et al., 1997), (Sharpless, 2005), (Courtois-Cox et al., 2008). However, neither p16INK4A nor p19ARF are critical for SnoN-induced senescence. Secondly, the DNA damage pathway is a critical mediator of oncogene-induced senescence (Bartkova et al., 2006), (Di Micco et al., 2006), (Mallette and Ferbeyre, 2007). Oncogenes such as activated Ras cause unbalanced DNA replication and initiate a classic p53-dependent DNA-damage response pathway. However in SnoN-induced senescence, activation of DNA damage or check point pathways is not detected. Thirdly, oncogenes co-operate with each other to induce transformation of MEF. However, not only did the expression of SnoN fail to induce transformation in the presence of an active Ras or Myc, it even inhibited the transformation induced by Ras and Myc. Finally, data from mouse skin carcinogenesis model confirmed that the mSnoN functions as a tumor suppressor in vivo. Therefore, SnoN can display a tumor suppressor activity through inducing cellular senescence.

The idea that SnoN possesses both pro-oncogenic and anti-oncogenic activities is consistent with earlier observations of SnoN expression patterns in human cancer tissues and cell lines (Villanacci et al., 2008). The pro-oncogenic activity and tumor suppressor activity of SnoN are clearly mediated by two separate pathways (Fig. 7F), with the pro-oncogenic activity depending antagonism of the TGFβ/Smad pathway (Zhu et al., 2006) and the tumor suppressor activity relying on PML and p53. Selective inactivation of the PML/p53 branch allows the oncogenic activity of SnoN to be fully manifested as in the case of SnoN∆322-366 or the transformation of p53-/- MEF, while mutation of the Smad binding activity of SnoN exposes the tumor suppressor activity of SnoN. The co-existence of both pro-oncogenic and anti-oncogenic activities in one protein is not unique to SnoN. The Ndy1 protein, a Jumonji C domain containing histone demethylase, also harbors pro-oncogenic activity through inhibition of

Page 43: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

30

senescence and tumor suppressor activity through maintaining genomic integrity and inhibition of cell proliferation (Pfau et al., 2008), (Suzuki et al., 2006), (Frescas et al., 2007). This emerging group of proteins with both pro-oncogenic and anti-oncogenic activities highlights the complexity of cellular events accompanying malignant progression.

Tumor suppressors are often inactivated or deleted during malignant progression. If SnoN contains anti-tumorigenic activity, why is it upregulated in many human cancer cells? We speculate that at early stages of tumorigenesis, tumor cells may upregulate SnoN expression in an attempt to halt tumor growth through inducing senescence. Thus, SnoN upregulation may initially serve as a barrier for malignant progression. To overcome this barrier, tumor cells may specifically inactivate the SnoN senescence pathway through downregulating p53 or PML, leaving cells with high levels of SnoN but do not undergo senescence. These high levels of SnoN may then promote tumor growth through its pro-oncogenic activity. Thus, high levels of SnoN expression may be the outcome of a complex evolving process during tumorigenesis. This model also implies that it is more advantageous for the cancer cells to maintain a high level of SnoN expression while inactivating its anti-oncogenic pathway at downstream points than to delete it. Indeed, our results that SnoN potently induces oncogenic transformation of p53-/- MEF and that SnoN∆322-366 functions as an oncogene support this model. Future experiments will determine whether human cancer cells with high levels of SnoN expression also harbor mutations in downstream components of the SnoN senescence pathway. This may have important implications in potential targeting of SnoN in human cancer.

Page 44: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

31

Materials and methods

Mice To generate knock-in mice, the targeting vector was introduced into the E14 mouse embryonic stem (ES) cells derived from 129P2/OlaHsd mice and injected into the blastocysts of C57BL/6 background. The resulted chimera were crossbred with a CMV (Cytomegalovirus immediate-early promoter)-cre transgenic mice to generate the knock-in mice and backcrossed to C57BL/6 for additional 5 generations. Knockout SnoN mice in C57BL/6 background were obtained from Dr. S. Pearson-White. Knockout p53 mice were obtained from The Jackson Laboratory. All animal works were approved and confirmed by the Animal Care and Use Committee at UC Berkeley. Cells, antibodies, and constructs 293T cells and Hep3B cells were maintained as described before ((Zhu et al., 2005). Anti-Flag antibody was purchased from Sigma; antibodies for p53 (ab26) and p19ARF (ab80) from Abcam; anti-PML (Mab 3738) from Chemicon; antibodies for phosphorylated ATM, ATR, Chk1, Chk2 and Rb from Cell Signaling; antibodies for p16INK4A, Smad3 (FL245), Smad7 (H79), Chk1 (FL478) and Tubulin from Santa Cruz biotechonology.

Primary MEF were prepared from E13.5 day embryos as described previously (Serrano et al., 1997) and maintained in DMEM medium with 10% FBS. A standard 3T9 protocol was employed to compare population doubling (Marı´a A. Blasco Et, 1997). shRNA in pSuper-Retro-puro were introduced into MEF by retroviral infection as described previously (Zhu et al., 2006). Mixture of siRNA duplex targeting mouse Smad2 and Smad3 were purchased from Dharmacon and transfected into MEF by Nucleofector system (Amaxa). The sequences of various shRNAs are: sh-Smad3: 5’ggccatcaccacgcagaac3’; sh-SnoN: 5’ctccattctgcagaggaag3’; sh-p19ARF: 5’caccggaatcctggaccag3’; sh-p53: 5’gtactctcctcccctcaat3’; sh-PML: 5’cccacttagaagacaggac3’

RT-PCR Total RNA extraction, cDNA preparation and PCR were performed using the standard protocol. The primers used to amplify the different genes by PCR are: p53: 5’actgccatggaggagtcac3’/5’tcagtctgagtcaggcccc3’; PML: 5’agcaggaggcttctcagacagt3’/ 5’cttgatgatcttcctggagcaa3’; p19ARF: 5’gttttcttggtgaagttcgtgc3’/5’tcatcacctggtccaggattc3’

Retroviral infection cDNAs or shRNAs in retroviral vectors were transfected into the Phoenix-E cells to generate high titer of viral supernatants, which were used subsequently to infect the MEF cells at P3. 48 hours later, infected cells were selected by culturing in the presence of 2 μg/ml puromycin. Pools of puromycin-resistant cells were analyzed in various experiments. For two rounds of infection, puromycin resistant cells were infected again with retroviruses that express neomycin-resistant genes. 48 hr after infection, the doubly infected cells were selected by 600ug/ml G418 for 4 days.

Transfection, co-immunoprecipitation and Luciferase assays 293T, Hep3B or MEF cells were transiently transfected using lipofectamine Plus (Invitrogen). Coimmunoprecipitation assay was performed as described previously ((Zhu et al., 2005). Luciferase assay was performed as described ((Zhu et al., 2005).

SA-β-Gal staining MEF cells were fixed and stained for the expression of the β-galactosidase activity at pH 6.0 (senescence-associated β-gal) according to the instruction (Senescence detection kit, Calbiochem). To detect senescence in mouse tissues, frozen tissue sections of 6μm thickness were stained for SA-β-Gal.

Page 45: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

32

BrdU incorporation assay Cells were incubated with medium containing 0.1mg/ml BrdU (Roche) for 6 hours. After fixation in methanol, cells were incubated with anti-BrdU working solution (Roche) for 30 min at 37°C, stained with Alexa Fluor 488-conjugated goat anti-mouse-IgG (Invitrogen) and visualized by fluorescent microscopy.

Immunofluorescence MEF were grown on glass cover-slips, fixed , permeabilized with 0.1% Triton X-100 for 5 min, stained with the appropriate antibodies and visualized by a Zeiss 510 confocal microscope. Cell nuclei were detected by 4,6-diamidino-2-phenylindole (DAPI) staining. Immunofluorescence in frozen tissue sections was performed following the same procedure except that the samples were permeabilized with 0.5% Triton X-100 for 15 min.

Soft-agar assay 3 ml of growth medium containing 0.66% Bacto-agar was added to a 50mm dish for the bottom agar layer. 6x103 MEF were resuspended in 2 ml medium containing 0.4% agar and overlaid on the hardened bottom layer. 2ml of fresh medium containing 0.4% agar was added to the dish once a week for 4 weeks. The colonies were visualized by staining with 0.5 mg/ml 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) (Sigma) for 4 hrs at 37°C.

Skin tumorigenesis The dorsal skins of 8-week-old mice were shaved and treated topically with 7,12-dimethylbenz(a)anthracene (DMBA; 100 μg/200 μl in acetone). One week later, 12-O-tetradecanoyl-phorbol-13-acetate (TPA; 20 nmol/200 μl in acetone) was applied twice weekly to the same area for 20 weeks (Balmain et al., 1988). Mice were euthanized at the end of 30 weeks. Surgically removed tumors were immediately snap-frozen in liquid nitrogen, and stored at -70°C. Alternatively, dissected skin and tumor samples were fixed in 10% neutral-buffered formalin at 4°C overnight, embedded in paraffin, sectioned to 6-µm thickness, and stained with hematoxylin and eosin (H&E).

Page 46: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

33

Figure legends

Figure 1: Generation of SnoN knock-in mice. A. Targeting constructs. The BamH I fragment of mouse snoN genomic DNA containing exon I and the flanking sequences was subcloned into pBluescript. The R-Smad binding site at residues 85-88 and Co-Smad binding site at residues 266-267 in exon I were mutated to alanine (shown as 85-88AAAA and 266,267AA respectively), and a Neomycin resistance gene flanked by two LoxP sequences was inserted into intron I. These mutations also generated new Sph I and Swa I cleavage sites. B. Genomic DNA was prepared from MEF. DNA fragment encompassing exon I was amplified by PCR and digested with Sph I to yield an 1 kb fragment for WT snoN and 0.8 kb fragment for mSnoN. C. Mutant MEF displayed enhanced TGFβ transcription. p3TP-Lux was transfected into the WT or m/m MEF. Luciferase activity was measured 16 hrs after TGFβ treatment. D. Mutant MEF are more sensitive to TGFβ-induced growth arrest. 5x104 WT or m/m MEF cells were cultured in the presence of TGFβ. The growth of cells were measured 4 days later by cell counting and compared to that of unstimulated cells.

Figure 2: The snoN knock-in mice are resistant to chemical carcinogen-induced skin tumorigenesis. A. Schematic representation of the two-step skin carcinogenesis protocol. B. Percentage of mice that developed papilloma within a 30-week window is shown in the graph. Graph in C showed average numbers of papilloma per mouse. D. The size distributions of papillomas from WT or m/m mice. E. H&E staining of sections from papilloma from +/+ or m/m mice (Scale bar: 200μm). F. Lysates from papilloma samples were subjected to western blotting to measure expression of p53, p16INK4a or phospho-ERK. Tubulin was used as a loading control. G. Frozen papilloma sections from +/+ or m/m mice were stained with SA-β-Gal (Scale bar: 100μm). H. Tumor sections were stained with anti-p19ARF (green; Scale bar: 100μm). The boxed areas were amplified to show the nucleolus staining of p19ARF (Scale bar: 30μm). Nuclei were stained with DAPI (blue).

Figure 3: MEF from the snoN knock-in mice displayed premature senescence. A. WT or m/m MEF were passaged according to the 3T9 protocol. Population doubling was measured by cell counting every three days. B. BrdU incorporation. WT or m/m MEF at P6 were incubated with medium containing BrdU followed by staining with anti-BrdU (Scale bar: 40μm). Quantification of the BrdU-positive cells was shown in the graph. C. SA-β-gal staining. WT or m/m MEF at P6 were subjected to staining with SA-β-gal (Scale bar: 100μm). Quantification is shown in the graph.

Figure 4: Elevated SnoN expression is responsible for premature senescence of the m/m MEF. A. SnoN expression is elevated in the m/m MEF. Total cell lysates from WT and m/m MEF were subjected to western blotting with anti-SnoN. Tubulin was used as a loading control. B. snoN-/- MEF do not undergo premature senescence. MEF prepared from snoN-/- or WT mice were passaged according to the 3T9 protocol. SA-β-gal staining was performed on cells at P6 and P13 (Scale bar: 100μm). C. A siRNA mixture targeting both Smad2 and Smad3 was transfected into m/m MEF at P4. Levels of Smad2, Smad3 and Smad7 were assessed by western

Page 47: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

34

blotting 48 hr after the transfection. D. Reducing Smad signaling does not affect premature senescence of the m/m MEF. WT or m/m MEF expressing the siRNA mixture were stained with SA-β-gal at P6 (Scale bar: 100μm). E. Reducing Smad3 expression by shRNA. The expression of Smad3 and Smad7 was measured by Western blotting. F. Senescence of MEF expressing shSmad3 was assessed by SA-β-gal staining at P6 (Scale bar: 100μm). G. shRNA targeting SnoN or HA-SnoN cDNA in retroviral vectors was introduced into the m/m MEF. SnoN expression was evaluated by western blotting, and senescence of these cell evaluated by SA-β-gal stain at P6 (Scale bar: 100μm) (H).

Figure 5: p53 is required for the premature senescence of m/m MEF. A. p53 and p19ARF are upregulated upon premature senescence of m/m MEF. Expression levels of a variety of proteins were compared in WT and m/m MEF at P4, P6, P13 and upon immortalization (Im). This figure panel was assembled from multiple gels with identical samples. The bracket indicated total Rb. Tubulin was used as a loading control. B. Reducing p53 expression by shRNA. p53 levels in MEF expressing sh-p53 were measured by western blotting. C. Senescence of m/m MEF expressing sh-p53 or sh-p19ARF was determined by SA-β-gal staining at P6 (Scale bar: 100μm). D. p19ARF and p53 levels in MEF expressing sh-p19ARF were measured by western blotting.

Figure 6: Interaction between SnoN and PML is required for premature senescence. A. Endogenous SnoN (Red) and PML (Green) were stained with anti-SnoN or anti-PML, respectively, in WT and m/m MEF at different passages (Scale bar: 10μm). Nuclei were stained by DAPI (Blue). B. Flag-SnoN was introduced into the WT MEF and stained with anti-Flag. Endogenous PML was stained with anti-PML (Scale bar: 10μm). C. Reduction of PML expression by shRNA. The levels of PML and p53 were measured by western blotting. D. PML is required for SnoN-induced premature senescence. SA-β-gal staining was performed at P6 in m/m MEF expressing sh-PML (Scale bar: 100μm). E. SnoN physically associates with PML. Interaction of His-PML with Flag-SnoN (WT or mutant) in co-transfected 293T cells was measured by immunoprecipitation with anti-Flag followed by western blotting with anti-PML. PML levels in the lysate were blotted with anti-PML. F. Interaction between endogenous SnoN and PML at P6 in MEF was measured by immunoprecipitation with anti-PML followed by western blotting with anti-SnoN. Total PML and SnoN levels were measured by western blotting. G. Left: schematic drawing of WT and mutant SnoN proteins. Right: Interaction of Flag-SnoN with His-PML was measured as described in E. H. Interaction of SnoN with PML is required for SnoN-induced premature senescence. Flag-tagged WT or mutant SnoN was introduced into the WT MEF at P3 by retroviral infection. At P7, expression of p53, PML and SnoN were evaluated by western blotting and senescence measured by SA-β-gal staining (Scale bar: 100μm) (I). J. SnoN∆322-366 fails to co-localize with PML. Immunofluorescence staining with anti-Flag (SnoN) and anti-PML was performed (Scale bar: 10μm).

Figure 7: SnoN upregulates PML expression to mediate p53 stabilization.

PML expression is upregulated in the m/m MEF as measured by RT-PCR (A) and western blotting at P6 (B). B. SnoN is required for PML upregulation. The levels of PML and SnoN in control- or shSnoN-expressing cells were determined by western blotting. C. SnoN interacts with both PML and p53 in senescence cells. The levels of SnoN, PML and p53 in nuclear extracts prepared from WT or m/m MEF at P1, P6 and P13 were measured by western blotting.

Page 48: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

35

Interaction of SnoN with PML and p53 was examined by immunoprecipitation with anti-SnoN followed by western blotting with anti-PML or anti-p53. D. SnoN and p53 co-localized in senescence cells. Endogenous SnoN (Red) and p53 (Green) were stained with anti-SnoN or anti-p53, respectively, in MEF cells at P13. E. SnoN level is not affected by PML. The levels of SnoN, PML and p53 in MEF expressing sh-PML were determined by western blotting. F. Model for the pro-oncogenic and anti-oncogneic functions of SnoN.

Figure 8: Oncogenic transformation of MEF cells. A. SnoN inhibits transformation of MEF. WT or m/m MEF at P3 were first infected with retroviruses expressing shRNA or HA-SnoN and infected again with retroviruses expressing H-Ras (Q61L) and c-Myc either individually or together and subjected to a soft-agar assay (Scale bar: 40μm). The colonies were visualized by MTT staining and quantified (shown in the graph to the right). B. m/m MEF cells enhanced oncogene-induced senescence. WT or m/m MEF expressing H-Ras (Q61L) either alone or together with c-Myc or SnoNΔ322-366 were subjected to SA-β-gal staining (Scale bar: 100μm). C. Disruption of the SnoN-PML interaction turns SnoN into a bona fide oncogene. WT MEF at P3 were sequentially infected with retroviruses expressing SnoN (WT or SnoNΔ322-366) first and later H-Ras (Q61L) and c-Myc either individually or together and subjected to a soft-agar assay (Scale bar: 40μm). D. Inactivation of p53 abolishes the tumor suppressor activity of SnoN. WT or p53-/- MEF expressing WT or mutant SnoN or various siRNA or shRNA were subjected to a soft-agar assay. The soft-agar colonies were visualized by MTT (Left; Scale bar: 40μm) and quantified. (* Enlarged figures are showed in supplemental Fig. 2).

Sup_fig 1: Inhibition of TGFβ/Smad signaling in m/m MEF does not block premature senescence. m/m MEFs were cultured in the presence or absence of 10μM SB431542 and subjected to SA-β-gal staining at P3 and P6 (Scale bar: 100μM, Top). The bottom panel showed efficient inhibition of Smad3 phosphorylation by SB431542. Tubulin was used as a loading control. Sup_fig 2: Oncogenic transformation of MEF. A: Enlarged Fig. 8A. B: Enlarged Fig. 8C. C: Enlarged Fig. 8D. The scale bar for all panels is 5μM.

Page 49: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

36

Figures

Figure 1. Generation of SnoN knock-in mice

Page 50: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

37

Figure 2. The snoN knock-in mice are resistant to chemical carcinogen-induced skin tumorigenesis.

Page 51: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

38

Figure 3. MEF from the snoN knock-in mice displayed premature senescence.

Page 52: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

39

Figure 4. Elevated SnoN expression is responsible for premature senescence of the m/m MEF

Page 53: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

40

Figure 5. p53 is required for the premature senescence of m/m MEF

Page 54: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

41

Figure 6. Interaction between SnoN and PML is required for premature senescence.

Page 55: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

42

Figure 7. SnoN upregulates PML expression to mediate p53 stabilization

Page 56: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

43

Figure 8. Oncogenic transformation of MEF cells

Page 57: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

44

Supplemental figure 1

Page 58: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

45

Supplemental figure 2

Page 59: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

46

Chapter 4: SnoN, a novel activator of p53

Page 60: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

47

Abstract

Many proteins have the potential for various functions depending on variables such as the cell context or specific stages. SnoN, while originally recognized as an oncogene, has been reported to have both pro-oncogenic and anti-oncogenic functions in tumorigenesis. It has been demonstrated that the pro-oncogenic function of SnoN is mediated through its repression function in Smad activities and TGFβ signaling. However, the mechanism mediating its anti-oncogenic function has not been understood until recent studies revealing an interaction of SnoN with the PML-p53 pathway. Specifically, highly expressed SnoN can bind to PML and is localized in PML NBs (nuclear bodies) in primary cells. The localization of SnoN to PML NBs leads to the stabilization of p53 and the induction of premature senescence, preventing oncogene induced transformation in cells and carcinogen induced tumorigenesis in mice. It has been reported that elevated levels of SnoN are frequently detected in many human cancer cells and tumor samples playing an important role in regulating tumor initiating and progression. A thorough understanding of the mechanisms mediating the different roles of SnoN in tumorigenesis would facilitate the possible development of the new cancer therapy strategy for these types of cancer patients by utilizing the “good” effects while avoiding the “bad”. Unfortunately, the detailed mechanism leading to stabilization and activation of p53 by SnoN in PML NBs is still unclear. We have shown here, that SnoN can directly bind to p53 resulting in stabilization of p53. PML may function as a scaffold to facilitate the physical interaction between SnoN and p53. The binding of SnoN with p53 results in the release of Mdm2 by competing with Mdm2 to bind with p53 and enhances the acetylation of p53 through other factors localized in PML NBs. Additionally, SnoN can be recruited together with p53 to p53 specific binding elements in the promoters of target genes acting as an activator of p53 transactivation activities. More importantly, SnoN is an important cellular activator of p53 and plays an important role in regulating p53 activation during stress responses. Furthermore, activation of p53 through its interaction with SnoN can trigger premature senescence and block oncogene induced transformation in cells. Activation of p53 by SnoN prevents chemical carcinogen induced tumorigenesis in mice. It suggests that SnoN is a positive regulator of p53 which can activate p53 activities both in vivo and in vitro.

Page 61: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

48

Introduction

Activation of oncogenes and inactivation of tumor suppressor genes are critical events in the multistage tumorigenesis. Oncogenes are a group of genes capable of promoting tumor growth when activated by gain of function mutations; while the tumor suppressor genes can promote transformation when inactivated by loss of function mutations (Hanahan and Weinberg, 2000; Weinberg, 1994). The definitions of oncogene and tumor suppressor gene are very distinct. However, distinguishing an oncogene or a tumor suppressor gene is not always easy in practice. It has been well established that some proteins or signaling pathways may function differently in different cell context. Both oncogenic and anti-oncogenic functions can be possessed by a protein or signaling pathway at the different stages determined by the cell context such as TGFβ pathway and its negative regulator, SnoN (Akhurst and Derynck, 2001; Pan et al., 2009; Zhu et al., 2006).

SnoN is a member of the Ski family of proteins that was identified based on sequence homology with v-Ski, the transforming protein of the Sloan Kettering virus (Nomura et al., 1989). SnoN is ubiquitously expressed in adult cells but at a relatively low levels except during specific stages of differentiation (Deheuninck and Luo, 2009; Jahchan and Luo, ; Luo, 2004). SnoN expression is subjected to stringent regulation in normal mammalian cells at the levels of protein stability and transcriptional activation (Deheuninck and Luo, 2009; Luo, 2004). SnoN transcription can be induced by tissue injury and upon stimulation with certain cytokines such as TGFβ (Kroening et al., 2009).TGFβ activates the Smad2/Smad4 complex that binds directly to the Smad binding elements in the snoN promoter to activate its transcription (Zhu et al., 2005). TGFβ also regulates SnoN stability through an ubiquitin-dependent proteasome degradation pathway. Several E3 ubiquitin ligases that interact with the Smad proteins have been shown to be recruited to SnoN for its ubiquitination, including the Smad ubiquitin regulatory factor (Smurf 2), the Anapahase promoting complex (APC/C) and Arkadia (Deheuninck and Luo, 2009). In some human cancer cells, the degradation pathway of SnoN is disrupted, leading to the elevated SnoN levels (Pot and Bonni, 2008).

Overexpression of SnoN can induce transformation of chicken embryonic fibroblast cells (Boyer et al., 1993). It is therefore classified as an oncogene. Consistent with this model, the human snoN gene is located on chromosome 3q26, a known oncogene locus that is frequently amplified in esophageal and ovarian cancer (Akagi et al., 2008; Nanjundan et al., 2008) and elevated SnoN expression has been detected in many cancer cell lines. The pro-oncogenic activity of SnoN has been attributed to its ability to antagonize TGFβ signaling. SnoN interacts with the Smad proteins and represses their transactivation activity by disrupting the functional heteromeric Smad complexes, recruiting transcription co-repressor complexes and blocking the binding of transcriptional co-activators to the Smads (Akiyoshi et al., 1999; Luo, 2004; Stroschein et al., 1999; Wu et al., 2002). Overexpression of SnoN blocks the growth inhibitory responses to TGFβ (Stroschein et al., 1999; Sun et al., 1999b), and this is suggested to be responsible for the pro-oncogenic activity of SnoN (He et al., 2003). Indeed, reducing SnoN in breast and lung cancer cell lines reversed transformation in vitro and tumor growth in vivo, and re-introduction of WT but not the mutant SnoN lacking the Smad binding sites rescued this response (Zhu et al., 2006).

Page 62: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

49

Recently, more and more evidence indicate that SnoN also contains an anti-oncogenic activity (Zhu et al., 2006). Mice lacking one copy of the snoN gene show in a slightly increased tumor incidence and increased susceptibility to chemical carcinogen-induced tumorigenesis (Shinagawa et al., 2000). In Drosophila, overexpressed dSnoN has a negative effect on cell growth (Ramel et al., 2007). In addition, in some human cancer tissues, SnoN was found to be downregulated (Bravou et al., 2009). Our recent study has linked the anti-tumorigenic activity of SnoN to its ability to induce premature senescence by stabilizing p53 in a PML-dependent manner (Pan et al., 2009). We found that high levels of SnoN in both epithelial and fibroblast cells can bind to PML and be recruited to the PML nuclear bodies where it may induce stabilization of p53, leading to premature senescence and increased apoptotic responses. Consistent with this ability of SnoN to activate the tumor suppressor pathway of PML-p53, overexpression of SnoN inhibited oncogene-induced cellular transformation of MEF cells and significantly blocked chemical carcinogen-induced carcinogenesis in vivo, due likely to the accumulation of senescence cells (Pan et al., 2009).

The tumor suppressor p53 is activated by various stress signals, including DNA damage, oxidative stress, oncogene activation, telomere shortening and metabolic stress, to coordinate cell cycle arrest, apoptosis, senescence and DNA repair processes (Harkin and Hall, 2000; Hussain and Harris, 2006; Lane, 1992). The regulation of p53 mainly occurs at post-transcriptional levels, in particular protein stability (Kruse and Gu, 2009). p53 stability is primarily controlled by the E3 ubiqitin ligase, Mdm2, which induces polyubiquitination of p53, leading to its degradation in a proteasome-dependent manner. Mdm2 may further repress p53 transcription by complexing with p53 on the promoter DNA. The stress signals stabilize p53 by blocking the Mdm2-p53 interaction either through post-translational modification of p53 and Mdm2 or physical competition. Phosphorylation and acetylation of p53 can further activate the transcriptional activity of p53 and facilitate recruitment of other transcription co-activators to the p53 responsive promoter DNA (Brooks and Gu, 2006; Kruse and Gu, 2009; Michael and Oren, 2003; Oren et al., 2002). Activation of p53 in response to cellular stress also requires the PML tumor suppressor protein. PML is essential for the formation of the PML nuclear bodies that have been linked to many cellular processes including stress response, transcriptional control, anti-viral response, DNA repair, apoptosis and senescence(Pearson and Pelicci, 2001; Salomoni and Pandolfi, 2002) and for recruitment of diverse proteins to the nuclear domain (Bernardi and Pandolfi, 2007; Dellaire et al., 2006; Seeler and Dejean, 1999; Shen et al., 2006). PML bodies may serve as scaffolds to increase the local concentrations of factors involved in p53 activation by facilitating phosphorylation and acetylation of p53 (Bode and Dong, 2004; Pearson et al., 2000; Pearson and Pelicci, 2001; Takahashi et al., 2004). Loss of PML allows MEF to bypass senescence due to lack of p53 activation (de Stanchina et al., 2004; Ferbeyre et al., 2000; Marcotte and Wang, 2002; Pearson et al., 2000). In addition to tumor suppression, p53-induced senescence is also involved in regulation of ageing. Expression of a constitutively active p53 in mice protects the mice from tumorigenesis, at the same time accelerates ageing (Donehower, 2009; Rodier et al., 2007; Wesierska-Gadek et al., 2005).

In this report, we performed study to uncover the molecular mechanism by which SnoN activates p53 and further determine the physiological functions of this regulation. Our studies revealed that high levels of SnoN can directly bind to p53 and activating it by preventing Mdm2 from binding to p53 and by enhancing acetylation and phosphorylation of p53. We also present data to suggest that activation of p53 by SnoN plays an important role in tumor suppression and ageing.

Page 63: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

50

Results

SnoN binds to p53 directly.

To explore whether SnoN can interact with p53, Flag-tagged SnoN was transfected into 293T cells together with HA-p53 and isolated by immunoprecipitation with anti-Flag, and SnoN-bound p53 was detected by western blotting with anti-HA (Fig 1A). As shown in Fig. 1A, SnoN can bind to p53 readily when overexpressed, and this interaction was not affected by the presence of the transfected PML. Consistent with this, the mutant SnoN (Δ322-366) defective in binding to PML could bind to p53 as well as WT SnoN when overexpressed (Fig 1B). In a GST-pull down assay, recombinant SnoN purified from bacteria could be pulled down together with GST-p53 but not with GST alone (Fig 1C), suggesting that the SnoN-p53 interaction is direct. To determine whether this interaction also occurs at the endogenous level, mouse embryonic fibroblasts (MEF) were treated with H2O2 (to induce p53), and the SnoN-p53 interaction was examined by co-immunoprecipitation assay. As shown in Fig. 1D, endogenous p53 readily interacts with SnoN. In addition, PML could also be detected in the SnoN immune complex, as shown before (Pan et al., 2009) (Fig 1D). This PML is probably required for stabilization of p53 upon treatment with H2O2 because when the PML level was reduced by shRNA against PML in these cells, p53 expression was decreased dramatically (Fig 1D). Blocking the proteosome-mediated degradation with MG132, a proteosome inhibitor stabilized p53 expression in these cells (Fig. 1D), and SnoN was found to complex with p53. This result suggests that PML is mainly required for stabilization of p53 and recruits both SnoN and p53 to the PML bodies, but is not directly involved in the SnoN-p53 interaction. In addition, binding of SnoN to PML is independent of p53. In p53 -/- MEFs, SnoN colocalized with PML as efficiently as in WT cells (Fig 2D), suggesting binding of SnoN and PML is an upstream event.

The interaction between SnoN and p53 is not unique to fibroblasts. In early passage unimmortalized human normal mammary epithelia cells, 184 cells, SnoN could also bind to p53 (Fig. 1E). Again in the absence of PML, when p53 was stabilized with MG132, endogenous p53 bound to SnoN readily (Fig 1E). Thus, SnoN can interact with p53 in both epithelial and fibroblast cells.

Binding of SnoN and p53 is required for SnoN-induced senescence.

To map the binding domain of p53 on SnoN, Flag-SnoN mutants were co-transfected with HA-p53, and their interactions examined in the co-immunoprecipitation assay. p53 bound to the N-terminal portion SnoN (residue 1-366), but not to the C-terminal fragment (residues 367-684) (Sup_Fig1A). A small deletion of residues 255 to 258 in SnoN completely abolished the binding to p53 (Fig 2B), but did not affect the binding of SnoN to Smad or PML (Sup_Fig 1B and Fig 2C) or the ability of SnoN to repress TGFβ induced transcription (Sup_Fig 1B). This suggests that residues required for binding to p53 did not overlap with those for binding to PML and Smads.

The binding of SnoN to PML appears to occur upstream of the SnoN-p53 interaction and senescence. In p53 -/- cells, SnoN was found to co-localize with PML in the PML NBs (Fig 2D), indicating SnoN can bind to PML independent of p53. Also in these p53 -/- cells, introduction of an active Ras led to oncogenic transformation but not senescence. Under this condition, SnoN still co-localized with PML (Fig 2D), indicating that recruitment of SnoN to the PML NBs

Page 64: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

51

through association with PML is not a consequence of cellular senescence, but rather an upstream event.

Binding of SnoN to p53 is required for stabilization of p53.

Previously we have shown that high levels of SnoN resulted in stabilization of p53, and this is critical for SnoN-induced senescence. To determine whether binding of p53 to SnoN causes stabilization of p53, a pulse-chase assay was performed to compare the half-life of SnoN-bound p53 with that of free p53. As reported before, in the absence of stress signals, p53 has a half life of shorter than 30 min (Fig 3A). In contrast, the half-life of SnoN-bound p53 is significantly lengthened to at least 90 min. Overexpression of PML together with p53 and SnoN did not further stabilize p53. In the same cells, the half-life of SnoN was not affected by the expression of p53 or p53 plus PML (Sup-Fig 2A). Consistent with the requirement of SnoN-p53 binding in stabilization of p53, the SnoN Δ255-258 defective in binding to p53 failed to induce stabilization of p53 (Fig 3B).

SnoN competes with Mdm2 for binding to p53.

The half-life of p53 is tightly controlled by the Mdm2 E3 ubiquitin ligase. Dissociation of Mdm2 from p53 is essential for the stabilization and activation of p53 in response to cellular stress. To determine how SnoN binding stabilizes p53, we compared the association of Mdm2 with p53 in the presence or absence of SnoN. When cells were transfected with a fixed amount of Mdm2, p53 and increasing amounts of SnoN, Mdm2 was gradually competed away from p53 by the increased amount of SnoN (Fig 3C). In the reverse experiment with a fixed amount of SnoN and increasing amounts of Mdm2, the SnoN-p53 complex was not affected by Mdm2 (Fig. 3C). This suggests that SnoN has a higher affinity for p53 than Mdm2. This is further confirmed in an in-vitro binding experiment using bacterially expressed recombinant GST-p53, Mdm2 and SnoN. When GST-p53 and rMdm2 were incubated with increased amount of rSnoN, the SnoN-p53 complex increased while the Mdm2-p53 complex was significantly decreased (Fig 3D). Similarly, in the reverse experiment with a fixed amount of rSnoN and increased amounts of rMdm2, rMdm2 had no effect on the SnoN-p53 complex (Sup_Fig 2B).

SnoN binds to the same site as Mdm2 and enhances the modification of p53.

SnoN may compete with Mdm2 for binding to p53 through two possible mechanisms. First, the SnoN binding site in p53 may overlap with that of Mdm2. To test this, a p53 mutant, L14Q/F19S, defective in binding to Mdm2 but still retaining the transcription activity was examined for its ability to bind to SnoN. Interestingly, this p53 mutant also failed to bind to SnoN (Fig 4A), suggesting that SnoN binds to the same site as Mdm2.

SnoN may also regulate the post-translational modification of p53, for example acetylation or phosphorylation, which is known to reduce the affinity of p53 for Mdm2. To test this, SnoN-bound p53 was isolated from cells co-transfected with Flag-SnoN with anti-Flag, and free-p53 isolated from the SnoN depleted cells with anti-p53. The acetylation and phosphorylation (Ser 18) of the SnoN-bound p53 were compared to that of free p53. As shown in Fig. 4B, SnoN-bound p53 clearly showed elevated acetylation and phosphorylation, suggesting that SnoN may also facilitate p53 modification.

Page 65: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

52

Although SnoN-bound p53 is acetylated and phoshporylated, these modifications are not required for binding of p53 to SnoN. The p53 (8R) mutant lacking all known acetylation Lysine residues and p53 (S18A) mutant both bound to SnoN to the same extent as WT p53 (Fig. 4C). Taken together, our data suggest that SnoN may bind to p53 to promote its acetylation and phosphorylation.

SnoN competes with Mdm2 for binding to the p53 responsive promoter DNA.

Mdm2 not only regulates p53 degradation but also represses its transcriptional activity at the promoter DNA (Kruse and Gu, 2009). Since SnoN competes with Mdm2 for binding to p53, it may also release the transcriptional repression by Mdm2 at the p53 responsive promoters. To test this, the recruitment of SnoN to the p53 responsive element in the p21CIP1 promoter was examined by chromatin immunoprecipitation (ChIP) assay. The p21 promoter contains two p53 responsive elements located at –2.9kb and –1.9kb upstream of the transcription initiation site (el-Deiry et al., 1995). It also contains a TGFβ/Smad responsive element (SBE) at the downstream of RE1 (Seoane et al., 2004). We examined binding of SnoN and Mdm2 to the RE1 region as well as to the SBE region as a control. SnoN was found to bind to RE1, but only upon treatment of cells with H2O2 that induces p53 expression (Fig 5A). This suggests that binding of SnoN to RE1 is p53-dependent. In contrast, binding of SnoN to the SBE region was detected both in the presence and absence of p53 (Fig. 5B), confirming that this binding occurs through a different mechanism from that binding to RE1 and is p53 independent. To further confirm the p53-dependent nature of SnoN binding to RE1, the SnoN ∆255-258 defective in binding to p53 was tested for its ability to be recruited to RE1 by the ChIP assay. As shown in Fig. 5C, while WT SnoN was readily detected at the RE1 region, ∆255-258 was not. Moreover, in p53-/- cells, even WT SnoN failed to be detected at the RE1 site (Fig 5C). Taken together, these data suggest that SnoN can be recruited to the p53 responsive element through its association with p53.

To determine whether SnoN could release Mdm2 from the promoter DNA, binding of Flag-Mdm2 to RE1 was examined in the absence or presence of HA-SnoN. In the absence of SnoN, Mdm2 can be found to bind to RE1 (Fig 5D). However, this binding was dramatically decreased when SnoN was present and bound to RE1 (Fig 5D). This indicates that SnoN can displace Mdm2 from the promoter DNA of p53 responsive genes. PML seems not to be recruited together with SnoN and p53 to the promoter. In ChIP assay with anti-PML, PML was not detected to be associated with RE1 site in p21 promoter after treatment of H2O2 in cells (Fig 5E). Moreover, the interaction of PML with p53 mutant in which all acetylation sites are mutated is stronger than WT p53 (Fig 5F). It suggests that PML has a higher affinity with unacetylated p53. PML might be released from p53 and SnoN complex after p53 is acetylated by the enzymes localized to PML NBs.

SnoN and PML can synergistically activate p53 dependent transcription.

We next examined the effects of SnoN on p53-dependent transcription using a p21 promoter driven luciferase reporter assay in p53-/- MEF cells. Expression of p53 significantly induced p21 transcription (Fig 5G). Since p21 promoter is also induced by TGFβ/Smad pathway through the SBE site, and SnoN can potentially repress p21 transcription through antagonizing the Smads, thus complicating the interpretation of our assay, the mSnoN mutant defective in Smad binding was employed for this experiment. Expression of mSnoN alone had no effect on p21 transcription in p53-/- cells. However, p53-dependent transcription was clearly enhanced by

Page 66: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

53

co-expression of mSnoN (Fig 5G). As reported previously, Mdm2 inhibited p53-dependent transcription. Consistent with the ability of SnoN to block the Mdm2/p53 interaction, mSnoN could significantly rescue p53-dependent transcription in presence of Mdm2 (Fig 5G).

Consistent with previous reports, expression of PML enhanced p53-dependent transcription since PML facilitates p53 stabilization and activation (Fig 5H). We found that expression of both SnoN and PML further increased p21 transcription, probably due to enhanced stabilization and activation of p53 (Fig 5H).

The SnoN/p53 interaction is required for premature senescence.

We have shown before that high levels of SnoN induced premature senescence of MEF in a p53-dependent manner (Pan et al., 2009) (Fig 6A and 6B). To determine whether the interaction of SnoN and p53 is required for this premature senescence, SnoN Δ255-258 defective in binding to p53 was introduced into primary MEF cells at P3. When expressed at similar level as WT SnoN, SnoN Δ255-258 failed to stabilize p53 and did not induce senescence (Fig 6A and 6B). This SnoN molecule still binds to PML, indicating that interaction of SnoN with PML is necessary but not sufficient for activation of p53 and induction of senescence.

Activation of p53 is required for the anti-oncogenic activity of SnoN.

We have shown before that SnoN contains both pro-oncogenic activity, through its ability to bind to and antagonize the Smad proteins, and anti-oncogenic activity, through the activation of PML-p53 pathway (Pan et al., 2009). To determine whether the SnoN-p53 interaction is required for the anti-oncogenic function of SnoN, we introduced SnoN ∆255-258 into WT MEF cells together with the active Ras. While WT SnoN could not induce transformation of MEF cells in conjunction with the active Ras, SnoN Δ255-258 readily transformed MEF together with Ras (Fig 6C). Moreover, overexpression of expression of SnoN Δ255-258 alone in p53-/- MEF is sufficient to transform these cells (Fig 6E). The activation of p53 by SnoN was also defect when the interaction between SnoN and either PML or p53 was disrupted (Fig 6D). Thus, the interaction of SnoN with p53 is critical for its anti-oncogenic function in cells.

Activation of p53 by SnoN has an anti-oncogenic function in vivo.

We have previously generated a knock-in mouse strain expressing the mSnoN defective in Smad binding and shown that this SnoNm/m mouse strain is resistant to chemical-induced carcinogenesis, probably due to senescence triggered by high levels of the mSnoN protein. To determine whether p53 is required for the resistance to tumorigenesis, we crossed the SnoNm/m

mice with p53-/- mice to generate SnoNm/m p53+/-mice that expressed reduced p53 protein and subjected the mice to a two-step skin tumorigenesis protocol. Mice were administered with one dose of DMBA followed by twice weekly treatment of TPA for 30 weeks. Development of papilloma was monitored. As shown before, the SnoNm/m mice displayed a greatly reduced ability to form papillomas when compared to SnoN+/+ or p53+/- mice (Fig 7A-7C). Interestingly, loss of one allele of p53 completely reversed the resistance to carcinogenesis displayed by the SnoNm/m mice. The SnoNm/mp53+/- mice started to developed tumors after 8-week treatment, and papilloma development was observed in all SnoNm/mp53+/- mice, similar to what was observed for SnoN+/+ or p53+/- mice (Fig 7A and 7B). More importantly, unlike SnoNm/m mice, the tumors in SnoNm/mp53+/- mice continued to grow to greater than 10 mm in diameter (Fig7C). When

Page 67: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

54

tumors harvested from these mice were examined for the presence of senescence responses, no senescent signals could be detected in tumor sections from the SnoNm/mp53+/- mice, in clear contrast from those from SnoNm/m mice that exhibited significant numbers of senescent cells (Fig 7C and D). Consistent with these, a high level of p53 was only detected in tumors from the SnoNm/m mice but not in those harvested from SnoNm/mp53+/- or SnoN+/+ mice (Fig 7D). These results indicate that p53 is required for the anti-oncogenic activity of SnoN in vivo, most likely due to SnoN-induced, p53-dependent senescence pathway.

The SnoN-p53 pathway also regulates ageing in mice.

p53 is involved in the regulation of both tumorigenesis and ageing (Donehower, 2009; Feng et al., 2008; Rodier et al., 2007). Activation of p53 not only protects against cancer, but may also accelerate ageing. Since the MEFs isolated from SnoNm/m mice have shown enhanced p53 activation in response to various cellular stress signals, we next asked whether SnoN may also play a role in the ageing process (by regulating p53 activation). Therefore, a cohort of 43 SnoNm/m mice and 34 SnoN+/+ mice were carefully monitored for premature ageing-like phenotypes during lifespan.

About 20.9% of the SnoNm/m mice died within the first year as compared with only 8.8% of wild-type mice (Fig8A and table1). The median lifespan of SnoNm/m mice is about 25 weeks shorter than wild-type mice (Fig8A and table 1). Although the overall life span of the SnoNm/m mice is much shorter than wild-type mice, the SnoNm/m mice developed normally and were indistinguishable from their wild-type littermates in the first a couple of months after birth. Visible premature ageing-like symptoms in SnoNm/m mice were apparent after 6-month of age including body weight loss and marked kyphosis (Fig8B-D). Both male and female SnoNm/m mice stopped gaining body mass around 6 to 12 -months old, while wild-type mice at the same age and gender were still growing (Figure 8B). 18-months old SnoNm/m mice were obviously lighter in weight and have marked smaller size than the age-matched wild-type mice (Figure 8B). As a major consequence of accelerated ageing, SnoNm/m mice displayed reduced reproductive ability. The litter size produced from the first time homozygous parents were markedly less than the first time wild-type parents at the same age (Figure 8E). Interestingly, when the metabolic activities including the oxygen consumption, generation of CO2 and heat, and the general mobility were evaluated in 4 pairs of SnoNm/m and wild-type littermates, no obvious difference was observed between these two cohorts (Table 2) suggesting that the premature ageing observed in SnoNm/m mice is not the typical metabolic ageing.

Since a high level of SnoN might affect p53 dependent cellular responses to stress during the ageing process, then we examined whether senescence or apoptosis responses were augmented in tissues of SnoNm/m mice. Tissue sections from the kidney of 18-month old SnoNm/m or wild-type mice were stained with SA-β-gal activity to compare senescence response (Figure 9A) or cleaved Caspase 3 activity for apoptotic response (Figure 9B). Interestingly, kidney sections from SnoNm/m mice displayed an increased level of senescence and increased numbers of apoptotic cells in contrast to wild-type mice. The enhanced responses to stress in SnoNm/m mice are not restricted to the kidney. An increased number of senescent cells were also observed in tissue sections of the skin of 18-month old SnoNm/m mice (Figure 9C). This suggests that SnoNm/m mice may exhibit an increased sensitivity to stresses as compared to age-matched wild-type mice and this enhanced stress response may contribute to the premature ageing in these mice.

Page 68: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

55

Discussion

SnoN can directly bind to p53 and increase the half-life of p53. The stabilization of p53 by SnoN is the direct consequence of this interaction. The binding of SnoN with p53 results in the release of Mdm2 and the enhanced acetylation of p53 by other factors localized in PML NBs. Additionally, SnoN can be recruited to the promoter of p53 target genes in a p53 dependent manner and act as an activator of p53 by inhibiting the interaction of Mdm2 with p53 at the promoter. SnoN plays an important role in p53 dependent stress responses. Activation of p53 through its interaction with SnoN can trigger premature senescence and block oncogene induced transformation in cells. Activation of p53 by SnoN prevents chemical induced carcinogenesis in mice.

SnoN can activate p53 activity by directly binding to it. It seems that PML is not important for this binding when both SnoN and p53 are highly expressed in cells. Overexpression of PML with SnoN and p53 has been shown to have no effect on the affinity of these two proteins. Moreover, mutant SnoN deficient in PML binding is still able to bind to p53. This idea is further confirmed by an in-vitro binding experiment in which bacterial recombinant SnoN can directly bind to recombinant GST-p53 in GST pull-down experiment, suggesting that the interaction between SnoN and p53 does not require any additional factors or modifications of p53. However, PML is important for the stabilization and activation of p53 in response to stress, since PML NBs are considered as a regulatory center for p53 in various stress responses (Dellaire and Bazett-Jones, 2004; Pearson and Pelicci, 2001). It has been shown that p53 cannot be stabilized under stress conditions when the expression of PML is reduced by shRNA in cells. Thus, the interaction of SnoN and p53 at endogenous levels requires PML since p53 is only stabilized when PML is present or when the degradation is inhibited by a protease inhibitor in the absence of PML. It suggests that PML might function as a scaffold in cells to increase the local concentrations of SnoN and p53 in order to facilitate their interaction with each other. Interestingly, our previous studies have shown that SnoN can also bind to PML and is colocalized with both PML and p53 in senescent cells, suggesting that SnoN, p53 and PML are able to form a complex in cells. We have shown here, that the binding of SnoN with PML is independent of p53 since a mutant SnoN deficient in p53 binding is still able to interact with PML and the co-localization of SnoN and PML can occur in p53 null cells. However, the stabilization of p53 necessary for its direct interaction with SnoN requires PML at endogenous levels. This suggests that the interaction of SnoN with PML may be an upstream event leading to the binding of SnoN with p53 in PML NBs. More importantly, PML is required for many post-transcriptional modification events of p53. Many enzymes for p53 modification have been reported to be localize in PML NBs including CBP, Chk2, HIPK2, and Mdm2 (Takahashi et al., 2004). We have shown that p53 does not undergo normal stabilization and acetylation by H2O2 when PML is reduced. Interestingly, total p53 level can be rescued by treating the cells with a protease inhibitor following knocking down PML, but the acetylation level of p53 cannot. It is also true for the acetylation of SnoN bound p53. The acetylation level of SnoN bound p53 is obviously higher than the acetylation of free p53 following the depletion of SnoN in cells. Although reducing PML expression impairs the acetylation of SnoN bound p53, unacetylated p53 can still bind with SnoN. It further supports that PML is not important for the directly binding of SnoN to p53 under high expression conditions but is likely required for further modification of SnoN bound p53 in PML NBs, thus promoting its transactivation activity. The ability of SnoN to bind with unacetylated p53 is consistent with the in-vitro binding experiment in which the modification of p53 does not seem necessary for their binding. This conclusion

Page 69: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

56

could be further confirmed by the interaction of SnoN with p53 mutants in which the acetylation or phosphorylation sites of p53 are mutated

SnoN can be recruited together with p53 to the p53 specific binding element in the promoters of target genes. There is little evidence to show that PML can be directly recruited to the regulatory elements in the promoter regions to regulate transcription, though PML NBs can be localized to the regions juxtaposed to chromosomes, which is important for the assembly of higher order chromosome structures or the regulation of other activities (Dellaire and Bazett-Jones, 2004; Luciani et al., 2006). Our data also suggests that the complex of SnoN and p53 on the promoter is released from PML NBs based on results from a ChIP assay and the observation that PML can interact with unacetylated mutant p53 at a higher affinity. It seems that the SnoN bound p53 is released from PML NBs after acetylation and recruited to the promoters.

Overexpression of SnoN not only stabilizes p53 but also activates p53 transcription activity. The p21 promoter driven luciferase activity is further enhanced by SnoN when SnoN and p53 are both expressed. Activation of p53 requires removal of Mdm2 from p53 preventing it from acting as a repressor of p53 at the promoters, and proper post-transcriptional modifications of p53. When SnoN is recruited together with p53 to the promoters of p53 target genes, Mdm2 has no chance to bind with p53 since SnoN is able to compete with Mdm2 to bind p53. In addition, the modification of SnoN bound p53 is enhanced by the enzymes localized in PML NBs. Modification states of p53 is essential for its stability, DNA binding affinity and transcriptional activity. It is important to note that SnoN and PML can synergistically activate the transcription of p53 target genes. SnoN not only removes the repressor, Mdm2 to stabilize and activate p53 but it also functions to enhances post-transcriptional modification of p53 through PML NB, further regulating its transcriptional activity. For this reason, we noticed a robust activation of p53 and premature senescence in cells with an elevated level of SnoN.

In a previous study, m/m with mutant SnoN deficient in Smad binding displayed resistance to chemical carcinogen induced tumorigenesis. Knocking in this mutant snoN gene not only enhances TGFβ activities but also increases the stability of SnoN in mice. Accumulation of mutant SnoN due to the enhanced stability induces premature senescence in cells by activating the PML-p53 pathway. Although more senescent cells have been observed in tumors growing on m/m mice, it is still unclear exactly how this resistance in m/m mice arises since both enhanced TGFβ activities and SnoN induced senescence might contribute to the suppression of tumor growth. We have observed here, that the increased level of p53 was detected only in the tumor samples from m/m mice which showed an elevated level of SnoN indicating that the elevated level of SnoN in m/m mice due to enhanced stability by mutations indeed could occur in vivo leading to activation of p53 activities. When one allele of p53 is removed from m/m mice, the enhanced senescence response in papilloma samples is impaired. More importantly, the resistance in m/m mice is defective when one allele of p53 is removed. All m/m mice developed tumors continuing to grow to a big size without arrest when one allele of p53 was removed. It supports that the resistance to chemical carcinogen induced tumorigenesis in m/m mice relies on the activation of p53 by SnoN. Activation of p53 by SnoN in mice can prevent tumor growth. Moreover, in m/m mice, enhanced TGFβ activities similar to those observed in m/m mice are expected because both lack the negative regulator of this pathway. However, we have not observed a similar resistance to skin carcinogenesis in SnoN null mice because p53 cannot be activated as in m/m mice after SnoN is removed. It also suggests that the enhanced TGFβ activities due to SnoN mutations are not important for skin carcinogenesis in our experimental conditions.

Page 70: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

57

Knock-in mice with a mutant snoN gene deficient in Smad bindings showed premature aging related phenotypes. The aging related phenotypes might be directly attributed by the interaction of SnoN with p53 rather than to the enhanced TGFβ activities since those phenotypes were not observed in SnoN knock-out mice. However, the aging phenotypes in knock-in mice are not exactly same as the phenotypes in the hyperactive p53 mutant mice. This difference might reflect the slow process of the upregulation of SnoN by oxidative stress at physiological conditions, leading to a gradual acceleration of p53 activation in mice. However, it is not completely rule out that the enhanced TGFβ activities might participate in the aging process in m/m mice as well.

SnoN is a newly identified positive regulator of p53. Elevated SnoN can activate p53 activities and trigger p53 dependent responses in cells and in the mouse model. SnoN might play an important role in the regulation of p53 dependent stress responses in cells. Most of the results describing the activation of p53 by SnoN in vivo and in vitro were based on the elevation of SnoN levels in cells or mice. Interestingly, SnoN expression in normal cells and adult tissues is usually nearly undetectable although elevated SnoN levels are observed in certain conditions such as in many cancer cells and at certain stages of tissue development. How SnoN expression is regulated under stress conditions intracellularly and whether SnoN may have a role in physiological responses to stresses in vivo are still under exploration.

Page 71: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

58

Materials and methods

Mice, cells, antibodies, and constructs The SnoNm/m (knockin) mice expressing a mutant snoN gene have been described previously (Pan et al., 2009). The p53-/- mice were obtained from The Jackson Laboratory. MEFs were prepared from E13.5 embryos following a published protocol (Serrano et al., 1997). 293T and RIE cells were maintained in DMEM with 10% serum. Hep3B cells were maintained in MEM with 10% serum. 184 primary human mammary epithelial cells were obtained from Dr. Martha Stampfer. The anti-SnoN rabbit polyclonal antibody was raised against a C-terminal peptide of SnoN and has been described before (Nadine's paper). Antibodies against Flag and HA were purchased from Sigma, p53 (ab26) from Abcam; mouse PML (Mab 3738) from Chemicon, acetyl-p53 from Upstate, Mdm2 and Tubulin from Santa Cruz biotechonology and cleaved Caspase 3 from abcam.

Transfection and Luciferase 293T and Hep3B cells were transiently transfected using Lipofectamine Plus (Invitrogen) as described previously (Zhu et al., 2005). Luciferase activity was measured 16 hr after stimulation with 50 pm TGFβ1. For p53 dependent transcription, 0.5 μg of p21-Lux and 0.5 μg of Flag-p53 were co-tranfected with or without 1 μg of Flag-SnoN in the presence or absence of 0.5 μg of Flag-Mdm2 or His-PML.

Immunoprecipitation and Western Blotting Flag- and HA-tagged proteins were co-transfected into 293T cells with or without His-PML and isolated by immunoprecipitation with anti-Flag or anti-HA antibody followed by elution with the corresponding peptides. The elution products are analyzed in Western blotting with antibodies as described previously. Endogenous proteins were isolated by immunoprecipitation with 20ug of antibodies and eluted by boiling in SDS-PAGE sample buffer. Pulse Chase Assay Cells plated in 6-well plates were washed with DMEM lacking Met and Cys, pulsed with 0.4 mCi/ml 35S-express (Roche) for 30 min and chased for different periods of time. GST pull-down assay Recombinant p53, SnoN and Mdm2 were expressed in and purified from E. coli as GST fusion proteins, and GST was excised from GST-SnoN and GST-Mdm2 by Thrombin cleavage. For binding experiments, 1.5 μg of GST-p53 immobilized on the glutothione Sepharose were incubated with 2μg of recombinant SnoN for 1 hour at 4°C. For the competition experiments, 1.5 μg of GST-p53 immobilized on the glutothione Sepharose were incubated with indicated amounts of recombinant SnoN or Mdm2. After extensive washing, the proteins were eluted by boiling in SDS-PAGE sample buffer and detected by western blotting or visualized by Coomasie staining. SA-β-gal staining in cells and tissues The senescence detection kit (Calbiochem) was employed to stain the senescent cells following manufactor's instruction. The 6μm-thick frozen tissue sections were stained for SA-β-gal and couterstained with Eosin. Retroviral infection Retroviral vector was transfected into Phoenix cells to generate the high titer of viruses as described previously (Zhu et al., 2005). The viral supernatant was used to infect the MEFs at P3

Page 72: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

59

in the presence of 4μg/ml of polybrene. 48 hours later, infected cells were splitted and selected in medium containing 2 μg/ml puromycin for another 48 hours. Immunofluorescence MEFs were grown on glass cover-slips, fixed with ice-cold methanol for 20 min, permeabilized with 0.1% Triton X-100 for 5mins and stained with the appropriate primary and secondary antibodies. Nuclei were detected by 4,6-diamidino-2-phenylindole (DAPI) staining. Cells were visualized by a Zeiss 510 confocal microscope. Immunofluorescence staining can be carried out in 6μm-thick frozen tissue sections on coated slides following the same protocol except the permeabilization was carried out with 0.5% Triton X-100 for 15 min. Chromatin immunoprecipitation (ChIP) assay Protein associated DNA fragments were isolated by ChIP assay as described previously (Zhu et al., 2005). The primer pairs used to amplify RE1 regions of the p21 promoters are 5’ cttatatctcccttggtcc 3’ and 5’ tattctgctggcaaagtggg 3’; for SBE regions of p21 promoter, the primers are 5’ acaggatgaggcttttgagg 3’ and 5’ cggcatacatctgtaatccc 3’. BrdU incorporation assay Cells were incubated with medium containing 0.1mg/ml BrdU (Roche) for 6 hours. After fixation in ice-cold methanol, cells were incubated with anti-BrdU working solution (Roche) for 30 min at 37°C, stained with Alexa Fluor 488-conjugated goat anti-mouse-IgG (Invitrogen) and visualized by fluorescent microscopy.

Soft-agar assay Soft-agar assay was employed to assess the anchorage-independent growth of transformed cells as described previously. Basically, Ras infected WT or p53-/- MEFs were further infected with viruses expressing WT or mutant SnoN. The doubly infected cells were grown in 0.4% agar for 4 weeks. The colonies were visualized and counted by staining with 0.5mg/ml of (4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) (Sigma) in PBS for 4 hrs at 37°C.

Two-step Skin carcinogenesis assay The two-step skin carcinogenesis with DMBA/TPA was carried out as described previously (Pan et al., 2009). Mice were euthanized at the end of 30 weeks. Surgically removed tumors were

immediately snap-frozen in liquid nitrogen and stored at -70°C. Alternatively, dissected skin and tumor samples were fixed in 10% neutral-buffered formalin at 4°C overnight, embedded in paraffin, sectioned to 6-µm thickness, and stained with hematoxylin and eosin (H&E). Tumor lysates were obtained by boiling a piece of tumor directly in SDS-PAGE sample buffer. .

Page 73: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

60

Figure legends

Figure 1. SnoN directly bind to p53 when they are highly expressed in cells. A. SnoN physically associates with p53 when both are highly expressed. Flag-tagged p53 and HA-tagged SnoN was transfected with or without His-PML into 293T cells. SnoN that associated with p53 was isolated by immunopricipitation with anti-Flag and detected by western blotting with anti-HA. B. The binding of SnoN with p53 is independent of its binding with PML. Flag-tagged WT SnoN or mutant SnoN (Δ322-366) was co-transfected with HA-tagged p53 into 293T cells. p53 that associated with SnoN was isolated by immunoprecipitation with anti-Flag and detected by western blotting with anti-HA. C. Bacterial expressed recomibant GST-p53 or GST alone was incubated with bacterial expressed recombinant SnoN. rSnoN associated with GST-p53 was isolated by GST pull-down and detected by western blotting with anti-SnoN. The amounts of input GST-p53 and GST were compared by Coommosie staining. The input rSnoN was showed by western blotting with anti-SnoN. D. Endogenous interaction of SnoN and p53. MEFs infected with retrovirus based shRNA against PML or vector were treated with 80μM of H2O2 for 24 hr followed by treating with MG132 or DMSO for 4 hrs. Endogenous p53 and PML associated with endogenous SnoN were isolated by immunoprecipitation with endogenous SnoN and detected by western blotting with anti-p53 or anti-PML. E. Immunoprecipitation with endogenous SnoN was implied to detect the associated p53 and PML in human mammary gland epithelial cells following the same strategy of treatments as in D. Figure 2: Mapping the binding site of p53 in SnoN and SnoN can interact with PML independently of p53. A. The schematic graph to represent SnoN and its mutants. B. Mapping the binding domain of p53 in N-terminus of SnoN. Flag-tagged WT SnoN or deletion mutants carrying serial deletions in N terminal portion of SnoN, was co-transfected with HA-tagged p53 into 293T cells, isolated by immunoprecipitation with anti-HA. C. The binding of SnoN and PML is independent of its interaction with p53. Flag-tagged WT or SnoN mutant unable to bind with p53 was co-transfected with His-PML into 293T cells. PML associated with SnoN was isolated by immunoprecipitation with anti-Flag and detected by western blotting with anti-PML. D. WT or p53 null MEFs were infected by retrovirus based constructs expressing H-Ras gene and selected by puromycin for 2 days. Then, those cells were immunostained with SnoN and PML. DAPI was used to visualize the nucleus. Figure 3: The binding of SnoN to p53 leads to the stabilization of p53 by competing with Mdm 2 binding. A. Overexpression of SnoN increases the half-life of p53. 293T cells transfected with Flag-p53 alone or co-tranfected with Flag-p53 and HA-SnoN in presence/absence of His-PML were pulsed with 35S for 20 mins and then chased for various time points. The half-life of p53 was compared by radioautography followed by immunoprecipitation with anti-Flag. B. Flag-tagged p53 was co-transfected with Flag-tagged SnoN or SnoN mutant into 293T cells. The amounts of p53 and SnoN in cell lysate were compared by western blotting with anti-Flag. C. SnoN can compete with Mdm2 to bind with p53. The fixed amounts of HA-p53 and Flag Mdm2 were co-transfected with increasing amounts of Flag-SnoN (0, 1, 2 or 4 μg) into 293T cells, or the fixed amounts of HA-p53 and Flag-SnoN were co-transfected with increasing amounts of Flag-Mdm2 (0, 1, 2 or 4 μg) into 293T cells. Mdm2 and SnoN that associated with p53 were isolated by immunprecipitation with anti-HA and detected by western blotting with anti-Flag. D. The

Page 74: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

61

competition is directly contributed by the interaction of SnoN and p53. Increasing amounts of bacterial expressed SnoN (0, 5, 10 and 20μg) were incubated with the fixed amounts of bacterial expressed GST-p53 and Mdm2. Mdm2 and SnoN that associated with GST-p53 were isolated by GST pull-down and detected by western blotting with anti-SnoN or anti-Mdm2. The input SnoN and Mdm2 were visualized by Commmsie staining. Figure 4: SnoN binds to the same site as Mdm2 in p53 and also enhances the post-trancriptional modification of p53. A. SnoN binds to the same site as Mdm2 in p53. Flag-tagged WT p53 or mutant p53 (L14Q/F19S) was co-transfected with HA-SnoN. SnoN associated with p53 was isolated by immunprecipitation with anti-Flag and detected by anti-HA. B. SnoN enhances the modification of p53 after binding to it. SnoN bound p53 was isolated by immunprecipitation with anti-Flag from MEFs infected with retrovirus based construct expressing Flag-SnoN. Different amounts of SnoN bound p53 were loaded to compare its acetylation and phosphorylation levels by western blotting with anti-acetyl-p53 (Lys 373 and Lys 382) or anti-phosph-p53 (Ser 18) with those in the remaining p53 after immunoprecipitation with anti-Flag. C. The acetylation of p53 is not required for its interaction with SnoN. MEFs were treated by the same strategy as in Figure 1D. The phosphorlation and aceylation of SnoN bound p53 were detected by western blotting in MEFs infected with retrovirus based shRNA against PML or vector alone. D. Mutations on major modification sites of p53 does not affect its interaction with SnoN. Flag-tagged SnoN was transfected with WT p53 or mutants (8R and S18A). p53 associated with SnoN was isolated by immunprecipitation with anti-Flag and detected by western blotting with anti-p53. Figure 5: SnoN can ben recruited to the p53 specific binding elment of p21 gene and activate p53 dependent transcription by antagonizing Mdm2. A. Schematic graph to represent the p53 specific binding element, RE1 and Smad binding site, SBE in p21 promoter. The primers used in ChIP assay were shown. B. The amounts of SnoN associated with RE1 but not SBE site are increased in stressed cells. In MEFs with or without treatment of H2O2, the fragments containing RE1 and SBE site were amplified by pairs of primers spanning RE1 site or SBE site following ChIP with anti-SnoN or anti-p53. The presence of RE1 and SBE containing fragments in lysate was examined by PCR with the corresponding primers. ChiP with anti-IgG was used as a control. C. The recruitment of SnoN to RE1 site is p53 dependent. Flag tagged WT and SnoN Δ322-366 were transfected into WT or p53 -/- MEFs. The amounts of RE1 containing fragments associated with SnoN were analyzed by ChIP assay with anti-Flag. The presence of RE1 containing fragments in lysate was examined by PCR with the corresponding primers. ChIP with anti-IgG was used as a control. D. SnoN can release Mdm2 from promoter of p53 target gene. p53 was transfected with Flag-Mdm2 and HA-SnoN or Flag-Mdm2 alone into MEFs. The amounts of Mdm2 and SnoN associated with RE1 sites were ampliied by primer 1 and 2 following ChIP with anti-Flag or anti-HA. The presence of RE1 containing fragments in lysate was examined by PCR with the same set of primers in total lysate. ChIP with anti-IgG was used as a control. E. PML is not recruited together with p53. The amount of RE1 containing fragments were analyzed by ChIP assay with anti-PML in cells with or without the treatment of H2O2. F. PML has a higher affinity with unacetylated p53. His-PML was co-transfected with WT p53 or mutant p53 into 293T cells. p53 associated with PML was isolated by using Ni-NTA resins to pull down His-PML and detected by western blotting with anti-p53. G. SnoN antagonizes Mdm2 activities to activate p53 dependent transcription. p21 promoter driven luciferase construct was co-transfected with p53 and mutant SnoN , or with p53 and Mdm2 in presence or absence of mutant SnoN, or with p53 alone or vector alone into p53

Page 75: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

62

null MEFs. The luciferase activities were measured after 48 hours of transfection. The expression levels of p53 were detected by western blotting with anti-p53. Tubulin was used as a loading control. H. SnoN and PML synergistically activate p53 dependent transcription. p21 promoter driven luciferase construct was co-transfected with p53 and SnoN in presence or absence of PML, or with p53 and PML, or with p53 lone or vector alone into p53 null MEFs. The luciferase activities were measured after 48 hours of transfection. The levels of p53 and actylated p53 were detected by western blotting with anti-p53 or anti-actyl-p53 (Lys 373 and Lys 382). Figure 6: Activation of p53 by SnoN can trgger p53 dependent senescence and block oncogene induced transformation in cells. A. The interaction between SnoN and p53 or between SnoN and PML is both required for SnoN induced senescence. MEFs infected with retrovirus based constructs expressing Flag-tagged WT SnoN or SnoN mutants (Δ233-258 or Δ322-366) were stained by SA-b-gal staining. B. Stabilization of p53 requires the interaction between SnoN and p53 or between SnoN and PML. The levels of p53 were detected by western blotting with anti-p53 in MEFs infected with retrovirus based constructs expressing Flag-tagged WT SnoN or SnoN mutants (Δ233-258 or Δ322-366). The expression levels of SnoN were detected by western blotting with anti-Flag. C. The anti-oncogenic function of SnoN is mediated by the interaction with both p53 and PML. WT or p53 null MEFs were infected by retrovirus based constructs expressing Ras and WT SnoN or SnoN mutants. Infected cells were grown in semi-solid soft agar for 4 weeks and the colonies were visualized and counted in 4 different areas. D. The levels of SnoN, p53 and Ras were detected by western blotting with anti-SnoN, anti-p53 or anti-Ras in infected cells. E. Mutations disrupting the interaction with PML and p53 have no effect on the pro-oncogenic function of SnoN. WT or p53 null MEFs were infected by retrovirus based constructs expressing WT SnoN or SnoN mutants. Infected cells were grown in semi-solid soft agar for 4 weeks and the colonies were visualized. Figure 7: Activation of p53 by SnoN prevents chemical carcinogen induced tumorigenesis. A. The anti-oncogenic function of SnoN is mediated by activation of p53 in vivo. 2-step carcinogenesis protocol was implied to SnoN m/m; p53+/-, SnoN m/m, SnoN-/-, p53+/-, and WT mice. The incidence (A), average number (B) and size (C) of tumors growing on those mice in 30-week window were graphed. D. SnoN induced senescence in vivo is required activation of p53 as well. Senescence in tumors with a diameter of around 2mm was detected by SA-β-gal(D) or p19Arf staining (E). F. The expression levels of p53 and SnoN was detected by western blotting with anti-p53 or anti-SnoN. Tubulin was used as a loading control. Figure 8: Premature ageing associated phenotypes in knock-in mice with mutant SnoN. A. A shorter lifespan in m/m mice. Indicated amounts of +/+ and m/m mice were maintained at the same condition for 180 weeks. The Kaplan-Meier survival curve was made based on the recorded numbers for dead tumor-free mice. B. m/m show a less body weight at the old ages. Male or female mice were maintained at the same condition. The body weights were measured at the different ages. C. A certain percentage of m/m mice have a small body size. The 18-month old littermates of +/+ and m/m mice were pictured. D. A certain percentage of m/m mice show kyphosis. The 18-month old littermates of +/+ and m/m mice were scarified and pictured after the skin was removed. E. m/m mice have a less reproductive ability. The indicated pairs of +/+ or m/m mice were mated. The pups produced from the first pregnancy of the mating pairs were recored. Figure 9: Senescence or apoptotic cells are accumulated in aged tissues from knock-in mice.

Page 76: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

63

A. More senescence cells were observed in kidney from m/m mice. Cryostat sections were obtained from kidney of 18-month old +/+ and m/m mice. SA-β-gal staining was implied to examine the senescence cells in kidney. B. More apoptotic cells were observed in kidney from m/m mice. Cryostat sections were obtained from kidney of 18-month old +/+ and m/m mice. Immunofluorescence staining for cleaved Caspase 3 was applied to examine the apoptotic cells in kidney. C. More senescence cells were observed in skin from m/m mice. Cryostat sections were obtained from skin of 18-month old +/+ and m/m mice. Immunofluorescence staining for p19Arf was applied to examine the senescence cells in skin. Sup_Fig 1: Mapping the binding site of p53 in SnoN and SnoN can bind to Smad proteins independent of p53. A. The schematic graphs to represent SnoN and its mutants. B. The N-terminal portion of SnoN is important for the binding of SnoN to p53. Flag-tagged WT SnoN or truncated mutants (1-366 or 366-684) was co-transfected with HA-tagged p53 into 293T cells, p53 associated with SnoN was isolated by immunoprecipitation with anti-Flag and detected by western blotting with anti-HA. C. Mutation in SnoN that abolishes interaction with p53 does not affect its interaction with TGFβ signaling pathway. Flag-tagged WT or SnoN mutant unable to bind with p53 was co-transfected with HA-tagged Smad4 into 293T cells. Smad4 associated with SnoN was isolated by immunoprecipitation with anti-Flag and detected by western blotting with anti-HA in upper panel. D. Flag-tagged WT or SnoN mutant was co-transfected with p3TP-Lux into Hep3B cells. Luciferase activity was measured 16 hrs after treatment with TGFβ in lower panel. Sup_Fig 2: The half life of SnoN is unaffected by p53 and Mdm2 can not release SnoN from p53. A. The half-life of SnoN is not affected by overexpression of p53. 293T cells transfected with HA-SnoN alone or co-tranfected with HA-SnoN and Flag-p53 in presence/absence of His-PML were pulsed with 35S for 20 mins and then chased for various time points. The half-life of SnoN was compared by radioautography followed by immunoprecipitation with anti-HA. B. The increasing amounts of bacterial expressed Mdm2 (0, 1, 2 and 4 μg) were incubated with the fixed amounts of bacterial expressed GST-p53 and SnoN in vitro. Mdm2 and SnoN that associated with GST-p53 were isolated by GST pull-down and detected by western blotting with anti-SnoN or anti-Mdm2. The input rSnoN and rMdm2 were visualized by Coommasie staining. Table 1: Shorter lifespan in m/m mice Table 2: Metabolic activities in aged mice (Collaborate with Dr. Andrew Stahl)

Page 77: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

64

Figures

Figure 1. SnoN directly bind to p53 when they are highly expressed in cells

Page 78: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

65

Figure 2. Mapping the binding site of p53 in SnoN and SnoN can interact with PML independently of p53

Page 79: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

66

Figure 3. The binding of SnoN to p53 leads to the stabilization of p53 by competing with Mdm 2 binding

Page 80: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

67

Figure 4. SnoN binds to the same site as Mdm2 in p53 and also enhances the post-trancriptional modification of p53

Page 81: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

68

Figure 5. SnoN can ben recruited to the p53 specific binding elment of p21 gene and activate p53 dependent transcription by antagonizing Mdm2

Page 82: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

69

Figure 6. Activation of p53 by SnoN can trgger p53 dependent senescence and block oncogene induced transformation in cells

Page 83: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

70

Figure 7. Activation of p53 by SnoN prevents chemical carcinogen induced tumorigenesis

Page 84: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

71

Figure 8. Premature aging associated phenotypes in knock-in mice with mutant SnoN.

Page 85: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

72

Figure 9. Senescence or apoptotic cells are accumulated in aged tissues from knock-in mice.

Page 86: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

73

Sup_Fig 1: Mapping the binding site of p53 in SnoN and SnoN can bind to Smad proteins independent of p53.

Page 87: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

74

Sup_Fig 2: The half life of SnoN is unaffected by p53 and Mdm2 can not release SnoN from p53.

Page 88: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

75

Tables

Table 1: Shorter lifespan in m/m mice

+/+ m/m Median lifespan

(weeks) 100.75 75.3

Maximal lifespan (weeks) 165.9 137.1

Lifespan<1 year 8.8% (3/34) 20.9% (9/43)

Page 89: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

76

Table 2: Metabolic activities in aged mice

Average (n=4)

VO2 (ml/kg/hr)

VCO2 (ml/kg/hr)

Heat (kcal/hr)

Xtot (counts)

+/+ 1216 842 0.691 445

m/m 1250 866 0.692 465

Page 90: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

77

Page 91: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

78

Chapter 5: SnoN, a stress transducer mediating p53 dependent stress responses

Page 92: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

79

Abstract

SnoN has both pro-oncogenic and anti-oncogenic functions during tumorigenesis. Its pro-oncogenic function is mediated by its interaction with the TGFβ signaling pathway. In my recent study, high levels of SnoN can interact with the PML-p53 pathway, providing a potential mechanism for its anti-oncogenic functions. The expression level of SnoN is a critical determinant of its molecular functions within cells. However, under normal physiological conditions, the levels of SnoN are relatively low in most adult tissues and normal cell lines. Elevated levels of SnoN are only observed at the certain stages of tissue development or in pathological conditions such as in certain cancer cell lines. How the expression of SnoN is controlled in cells is still unclear. It is reported here, that oxidative stress might directly induce SnoN expression through DNA damage pathways in addition to other stress activated pathways. It has been shown that levels of SnoN are gradually increased by oxidative stress during cultural of primary MEFs. The induction of SnoN is defective when cells are cultured in low oxygen concentration or in the presence of ROS inhibitor, Exposing cells to oxidative stress can directly induce the expression of SnoN at a transcriptional level through promoter activation. Moreover, the induction of SnoN by oxidative stress seems to require the cooperation of multiple stress activated signaling pathways including activation of ATM/ATR kinases in DNA damage pathways, MEK/Erk kinase and PI3K kinase pathways. However, the activation of a single pathways is insufficient to induce SnoN expression by oxidative. More importantly, the upregulation of SnoN could function as a stress sensor to mediate the p53 dependent responses to oxidative stress. In SnoN null cells, the activation process of p53 is delayed, suggesting that SnoN can accelerate p53 activation and increase the sensitivity of cells to stress.

Page 93: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

80

Introduction

SnoN, a negative regulator of TGFβ signals, plays an important role in tumorigenesis (Deheuninck and Luo, 2009; Luo, 2004). When first identified, SnoN was classified as an oncogene largely based on its ability to induce transformation in avian fibroblast cells. However, increasing evidences supports that SnoN has both pro-oncogenic and anti-oncogenic functions (Deheuninck and Luo, 2009; Jahchan and Luo, ; Pan et al., 2009). Amplification and overexpression of snoN genes have been observed in many human cancer cell lines (Akagi et al., 2008; Imoto et al., 2001; Nanjundan et al., 2008). Furthermore, reducing SnoN expression in human breast or lung cancer lines impairs tumor growth both in vivo and in vitro (Zhu et al., 2006). Transgenic expression of a truncated SnoN under the MMTV promoter in mouse mammary glands promotes tumor growth and metastasis in the background of PyVmT tumor mice (Jahchan et al.). The oncogenic function of SnoN is mediated by its interaction with TGFβ signals. Reintroduction of mutant SnoN deficient in Smad binding can not rescue tumor phenotypes in SnoN knock-down tumor cells (Zhu et al., 2006). However, there are reports that also support the anti-oncogenic function of SnoN. Particularly, the studies in mice lacking one copy of snoN gene have showed a slightly increased incidence of spontaneous tumor and increased sensitivity to chemical-carcinogen induced tumorigenesis (Shinagawa et al., 2000). In my previous reports, a novel PML-p53 pathway interacting with SnoN is identified. SnoN can bind to both PML and p53. The interaction of SnoN with p53 can stabilize and activate p53 by releasing Mdm2 from p53 and enhancing the acetylation of p53 through interacting with PML NBs. Overexpression of SnoN induces premature senescence in cells in a p53 dependent manner. The interaction between SnoN and PML-p53 pathway might provide a potential mechanism for the anti-oncogenic functions of SnoN (Pan et al., 2009). In fact, mice with knock-in stable form of mutant SnoN deficient in Smad binding become resistant to chemical carcinogen induced tumorigenesis probably by activating SnoN induced senescence in cells. The resistance of knock-in mice is mediated by the activation of p53. Lacking one copy of p53 gene in knock-in mice impairs the resistance. In cells, elevated SnoN blocks the oncogene induced transformation through the interaction with both PML and p53. Mutations in PML or p53 binding site can abolish the suppressing effects of SnoN in oncogene-induced transformation. Thurs, by interacting with distinct pathways, SnoN can play an opposite functions in tumorigenesis.

The intracellular level of SnoN seems to be critical in determining the functions of SnoN in cells, especially for the determination of SnoN functions in tumorigenesis. SnoN can interact with p53 only when it is highly expressed in cells. Specifically, highly levels of SnoN induce a premature senescence to block oncogene-induced transformation in cells and carcinogen induced tumorigenesis in mice. SnoN is ubiquitously expressed in all embryonic and adult tissues at a relatively low level. The upregulation of SnoN only occurs at certain stages of tissue development such as in the pregnant stage of mammary gland development or in some human cancer cells (Deheuninck and Luo, 2009; Jahchan and Luo). The previous studies by our group and other have shown that the levels of SnoN are tightly controlled in cells at the levels of protein stability, transcriptional activation and post-transcriptional modification. The stability of SnoN is tightly controlled by several E3 ubiquitin ligases including Smurf2 (Smad ubiqutin regulatory factor), APC (Anaphase promoting complex) and Arkadia(Bonni et al., 2001; Levy et al., 2007; Nagano et al., 2007; Stroschein et al., 2001). Upon TGFβ stimulation, R-Smad protein can bind to SnoN and recruit E3 ubiqitin ligases to SnoN leading to the degradation of SnoN

Page 94: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

81

through proteosome dependent pathway. The rapid turn-over of SnoN by TGFβ signals is believed to release Smad proteins from their antagonizors , thus promoting the propagation of TGFβ signals (Deheuninck and Luo, 2009; Jahchan and Luo, ; Luo, 2004). At the later stages of TGFβ treatment, SnoN transcription is rapidly induced in a Smad dependent manner. After 1 hour of treatment by TGFβ, the complex of Smad2 and Smad4 binds to the SBE site in the snoN promoter activating its transcription. This later stage upregulation of SnoN is thought to help turn off TGFβ signals efficiently(Deheuninck and Luo, 2009; Jahchan and Luo, ; Luo, 2004; Zhu et al., 2005).

However, the regulation of SnoN level in cells at a physiological condition is still unclear. In my previous study, knock-in MEFs (m/m) with mutant SnoN unable to bind to Smad display premature senescence at passage 6. Why do those m/m MEFs show premature senescence after certain passages of culture? The premature senescence is closely related with SnoN levels in m/m MEFs. When the levels of SnoN in MEFs were compared in serial culture, a gradual increased expression of SnoN was observed along with the increased cultural passages in both +/+ and m/m MEFs. However, mutant SnoN in m/m MEFs are more stable due to the mutations deficient in Smad binding and Smad dependent degradation mechanisms. Therefore, mutant SnoN reaches a very high level at passage 6 leading to premature senescence while wt SnoN achieves a similar level at a later passage (Pan et al., 2009). The factors causing upregulation of SnoN in culture conditions is still unclear. One potential factor inducing SnoN expression in culture may be oxidative stress, the essential factor causing cellular senescence in MEFs.

Oxidative stress is caused by oxidative damage in cells mediated by ROS (reactive oxygen species) including superoxide (O2-), hydroxyl radical (.OH) and hydrogen peroxide (H2O2). ROS are constantly generated in cellular metabolism or in responses to environmental stimuli. These species may cause DNA, protein or lipid damage, leading to genetic mutations, chromosome instability and/or other defects in cells. Recently, ROS have been indicated as a second messenger to play a central role in many intracellular signal transduction pathways for a variety of cellular processes including cell growth/proliferation, differentiation, inflammatory responses and intracellular metabolism (Benz and Yau, 2008; Liou and Storz, ; Ma). ROS regulate signal transductions such as MEK/ErK kinase cascade and PI3K/Akt pathway mainly through reversible oxidation of target proteins including tyrosin phosphatases, protein tyrosine kinases, receptor tyrosine kinases and transcription factors (Finkel, 2001; Liou and Storz, ; Rhee et al., 2000). In mouse cells, senescence is mainly induced by the effects of oxidative stress due to long telomere and highly activated telomerase. Oxidative stress causes DNA damage and activates other signaling pathways leading to the activation of p53 and induction of cellular senescence (Campisi and d'Adda di Fagagna, 2007; Yaswen and Campisi, 2007). When mouse cells are cultured in low concentration of oxygen condition or in the presence of inhibitors for ROS, the senescence response is impaired as well as silence of p53 (Itahana et al., 2004; Yaswen and Campisi, 2007).

In culture, we observed that senescence in MEFs is closely correlated with the high level of SnoN which is shown to gradually increase along with cultural time (Pan et al., 2009). We hypothesize that oxidative stress might directly induce SnoN expression through DNA damage pathways and/or other pathways as well. SnoN might function as a stress sensor to activate p53 dependent stress responses through its interaction with PML-p53 pathways. Indeed, our data has shown that m/m MEFs are defective in the senescence response when SnoN is unable to increase in cells cultured in low oxygen concentration or in the presence of ROS inhibitor. Exposing cells to oxidative stress can directly induce the expression of SnoN at a transcriptional level through

Page 95: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

82

promoter activation. Furthermore, the upregulation of SnoN by oxidative stress requires activation of multiple signaling pathways including activation of ATM/ATR kinases in DNA damage pathway, MEK/Erk kinase and PI3K kinase. However, the activation of a single pathway seems to be insufficient for the upregulation of SnoN by oxidative stress. More importantly, the upregulation of SnoN could function as a stress sensor to mediate the p53 dependent stress responses in cells. Induction of SnoN can accelerate p53 activation and increase the sensitivity of cells to stress.

Page 96: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

83

Results

Oxidative stress can regulate expression of SnoN

In serial passages of MEFs, SnoN was observed to gradually increase in cells. To determine whether oxidative stress, the major inducer of senescence in mouse cells, might have some effects on the expression of SnoN in cultured MEFs, wt and m/m MEFs were cultured in the presence of NAC (N-Acetyl-Cystine), a scavenger of ROS. SnoN no longer accumulated in m/m MEFs when NAC was present in the culture medium and accordingly, premature senescence was impaired in m/m MEFs (Figure 1A). When m/m MEFs were cultured in a low oxygen condition, 3% oxygen which removed oxidative stress in culture which removes cellular senescence in mouse cells (Parrinello et al., 2003), SnoN level was not increased at P6. As expected, m/m MEFs fail to show premature senescence at P6 (Figure 1B). Thus, the upregulation of SnoN triggering premature senescence in m/m MEFs is likely due to the presence of oxidative stress in culture. To confirm that oxidative stress can directly induce the expression of SnoN, MEFs were directly treated with H2O2 or Rotenone for various times in order to increase intracellular ROS levels in cells. SnoN levels were significantly increased after 24 hour treatment of either H2O2 or Rotenone as detected by western blotting (Figure 1C). This upregulation of SnoN by oxidative stress might occur at a transcription level. The transcription of snoN gene was shown to increase when SnoN mRNA was examined by semi-quantitative RT-PCR after treatment of either H2O2 or Rotenone (Figure 1D). To further confirm that oxidative stress can induce SnoN expression at the transcriptional level, a luciferase construct containing a 3.2 kb fragment upstream to transcription initiation site of snoN gene, was transfected into MEFs. SnoN promoter driven luciferase activity was increased after treatment of H2O2 in these transfected cells (Figure 1E). It suggests that oxidative stress can directly induce expression of SnoN at the transcriptional level through promoter activation. The upreguation of SnoN is not restricted to directly apply oxidative stress on cells. Other stress signals which may potentially increase intracellular ROS levels can induce SnoN expression as well. Treatment of UV and overexpression of oncogenic Ras are documented to elevate introcellular ROS levels. Accordingly, an induction of SnoN expression was observed as well in UV treated or oncogenic Ras expressing cells (Figure 1F and 1G). It is well established that overexpression of oncogenic Ras in primary cells can induce premature senescence but cause transformation of p53 null cells. Interestingly, the upreguation of SnoN by overexpressed oncogenic Ras was observed in wt and p53 null cells. It suggests that the induction of SnoN by stresses is independent of the senescence response in cells. The upregulation of SnoN is an important intermediate event in course of stress responses and plays an important role in regulating the proper responses to cellular stress as I have demonstrated in chapter 2.

Induction of SnoN requires multiple pathways

SnoN can be induced by oxidative stress. To dissect the pathways mediating the induction of SnoN by oxidative stress, MEFs were pretreated with different inhibitors followed by treatment of H2O2. When cells were pretreated with Caffeine or CGK733 which can block both ATM and ATR kinase activities, the expression of SnoN was unaffected by H2O2 (Figure 2A and 2B). To confirm that both ATM and ATR are involved into the induction of SnoN expression by oxidative stress, the regulation of SnoN expression by oxidative stress was

Page 97: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

84

analyzed in ATM/ATR double knock-out MEFs. ATM-/-; ATR flox/- MEFs were infected with a retrovirus vector expressing Cre recombinase to remove the remaining one allele of atr gene followed by treatment with H2O2. In cells infected by a control virus, SnoN was normally induced by H2O2 even with only one allele of ATR present but not in MEFs infected with Cre in which both ATM and ATR were absent (Figure 2C). Interestingly, in ATM+/+; ATR flox/- MEFs infected by retrovirus vector expressing Cre recombinase, SnoN was still induced by H2O2 (Figure 2C). This might indicate that activation of ATM/ATR kinases in DNA damage responses is necessary for the induction of SnoN by oxidative stress. ATM and ATR may have compensatory mechanisms in the induction of SnoN expression since removal of both ATR and ATM is required to block upregulation of SnoN by H2O2.

ATM and ATR kinases are the critical sensors needed to regulate cell responses to genotoxic stress partially through activation of p53 by increasing its phosphorylation (Efeyan and Serrano, 2007). Interestingly, ATM and ATR can also induce the expression of SnoN under stress. SnoN itself, an activator of p53, can activate p53 and regulate p53 dependent responses to oxidative stress through a phosphorylation independent manner. It is very likely that SnoN can also play a role in ATM and ATR dependent p53 activation. To determine whether the activation of p53 by ATM and ATR is influenced by induction of SnoN, we examine the kinetic patterns of activation of ATM, induction of SnoN and stabilization of p53 under stress. In MEFs treated with a low dose of H2O2 for various times, ATM was observed to be phosphorylated after 4 hours of treatment. Interestingly, the level of p53 was slightly increased even without a significant upregulation of SnoN at this time point (Figure 2D). This suggests that ATM/ATR kinases can cause stabilization and activation of p53 through a SnoN independent manner. It is well established that ATM and ATR can sense p53 through phosphorylation dependent stabilization and activation mechanism. However, the upregulation of SnoN expression occurred at a later time point. After 8 hours of treatment, the level of SnoN began to increase (Figure 2D). After 24 hours, SnoN expression peaked. Interestingly, p53 levels further increased in conjunction with the induction of SnoN. The expression of SnoN is tightly correlated with the increased p53 levels in cells (Figure 2D). In SnoN -/- MEFs, the kinetic pattern of p53 was changed. Early activation of p53 started almost at the same time as wt cells when pATM level went up in 4 hours of treatment. However, the further increased level of p53 in wt cells was absent when SnoN was removed (Figure 2D). This suggests that upregulation of SnoN might also be important for ATM and ATR dependent activation of p53. These results also suggest that the induction of SnoN by stress can accelerate p53 activation, thus increasing the sensitivity of cells to stress. To determine whether other pathways other than DNA damage pathways may be involved in the induction of SnoN by oxidative stress as well, MEFs were pretreated with SB431542 (a TGFβ receptor kinase I inhibitor), PP2 (a Src tyrosine kinase inhibitor), U0126 (a MEK kinase inhibtor) and LY294002 (a PI3K inhibitor) followed by the treatment of H2O2. Interestingly, pre-treating cells with U0126 and LY294004 significantly repressed the upregulation of SnoN by H2O2 but SB431542 and PP2 seems to have no effects (Figure 2E). However, it can not be ruled out that the effects of the inhibitors, U0126 and LY294002 on the induction of SnoN might be due to the decrease of intracellular ROS levels since both MEK kinase and PI3K kinase pathways have been reported to regulate the cellular redox balance in response to oxidative stress (Liu et al., 2005). Interestingly, the percentage of inhibitor-pretreated cells receiving oxidative stress after the treatment with H2O2 for 24 hr were unchanged compared to control cells as monitored by the staining of ROS probe, H2DCFDA (data not

Page 98: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

85

shown). This suggests that both pathways can block the upregulation of SnoN independently of their ability to regulate cellular redox homeostasis. It has recently been reported that the expression of SnoN can be affected by PI3K kinase activity (Nanjundan et al., 2008). To confirm that the PI3K kinase pathway is involved in regulation of SnoN expression by oxidative stress, both wt and m/m MEFs were cultured in the presence or absence of LY290042 inhibitor when isolated. Those MEFs grew much slower in the presence of LY290042 and became very unhealthy accompanying with lots of cell death. However, m/m cells did not enter the premature senescence at P6 in the presence of the inhibitor. More importantly, the level of SnoN in m/m at P6 did not increase by oxidative stress when the inhibitor was present (Figure 2F). It confirms that PI3k kinase is involved into regulating SnoN expression under stress. Interestingly, when a constitutively activated Akt construct expressing Myristoylated Akt, the downstream mediator of PI3K kinase pathway, by retrovirus infection was introduced into MEFs at the early passage (P3), the expression levels of SnoN were similar to the control cells in the presence or absence of H2O2 in medium (Figure 2G). This result indicates that multiple pathways may be involved into the upregulation of SnoN by oxidative stress. In short, DNA damage, MEK kinase and PI3K pathways are all necessary but not sufficient for the upregulation of SnoN in response to oxidative stress.

SnoN plays an important role in regulating the early responses of p53 under stress conditions.

The upregulation of SnoN by oxidative stress indicates that SnoN may play a role in regulating p53 dpendent stress responses since SnoN can stabilize and activate p53. p53 is a cellular stress sensor preventing inappropriate propagation of stressed or damaged cells. Various stress types including oxidative stress, DNA damage, oncogenic activation and chromosome abnormalities can activate p53 to trigger specific responses in cells such as cell cycle arrest, DNA damage repair and apoptosis. To determine whether SnoN is required for p53 dependent stress response, MEFs expressing a retrovirus vector based shRNA against either SnoN or control vector were treated with a low dose of H2O2 to induce premature senescence. p53 was shown to be stabilized and activated after 4 days of treatment as detected by increased total protein levels and the increased acetylation levels (Fig 3B). Accordingly, control cells were shown growt arrest and mostly BrdU negative following 4 days of treatment (Fig 3A). Those cells eventually triggered senescence responses and more than 80% cells showed positive in SA-β-gal staining after 7 days of treatment (Fig 3C). In contrast, when SnoN was knocked down by shRNA, p53 could not be normally stabilized and activated in 4 days of treatment as controls (Fig 3B). Instead, around 40% of knock-down cells were BrdU positive indicating that those SnoN knock-down cells were still in a growth phase despite 4 days of treatment (Fig 3A). Only about 5% knock-down cells showed positive in SA-β-gal staining after 7 days of treatment (Fig 3C). The data suggests that p53 is no longer stabilized and activated in cells undertaking oxidative stress when expression of SnoN is reduced. Therefore, those cells fail to trigger a normal senescence response. When comparing the expression levels of p53, p53 was found much lower in knock-down cells compared to parental cells (Fig 3B). Interestingly, when SnoN knock-down cells were exposed to H2O2 for an increaed amount of time, these cells also stopped growing and showed BrdU negative in 9 days of treatment (Fig 3B). Interestingly, these knock-down cells eventually entered senescence as detected by SA-β-gal staining after 14 days of treatment (Fig 3C). This suggests that p53 is eventually activated at later stages of treatment in

Page 99: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

86

SnoN knockdown cells. In addition, it was observed that the total and acetylated levels of p53 in SnoN knock-down cells were eventually upregulated after increasing the time of exposure to oxidative stress (Fig 3B). These results indicate that SnoN certainly plays a role in the regulation of p53 activation under stress conditions, but it is not the only pathway mediating p53 responses to stress. When SnoN is reduced, p53 stabilization and activation does not occur at the early stages. When these cells are exposed to stress for a longer period of time, other events such as p19Arf activation or robust DNA damages may compensate for the reduced levels of SnoN. Cells with reduced SnoN can eventually sense p53 activation and enter premature senescence through SnoN independent pathways.

SnoN is also important for the activation of p53 in response to stress in epithelial cells.

The anti-proliferating function of SnoN is not only seen in fibroblast cells. In epithelial cell lines, RIE and 184 cells, overexpression of SnoN in epithelia cells can inhibit cell growth and cause senescence-like phenotypes, as indicated previously. In epithelial cells, SnoN can interact with p53 when activated by oxidative stress (Fig 3E), suggesting that SnoN might play a role in regulating stress responses in epithelial cells as well. To test this hypothesis, RIE cells infected with retroviral shSnoN or control vector were treated with H2O2 for 8 hours. p53 was activated in control cells but not in SnoN knock-down cells (Fig 3E). Additionally, control cells stopped growing after 24 hours of treatment, while SnoN knock-down cells continued to grow even in the presence of H2O2 (Fig 3D). Thus, SnoN is required for the p53 dependent responses to oxidative stress in epithelial cells as well.

Page 100: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

87

Discussion

SnoN is a stress sensor mediating the activation of the p53 dependent stress responses. Oxidative stress is able to directly induce the expression of SnoN in both fibroblast and epithelial cells. Multiple pathways are involved in the upregulation of SnoN by stress including the DNA damage pathway, MEK/Erk kinase and PI3K kinase pathways. The induction of SnoN by stress may require the coordination of multiple pathways. The induction of SnoN increases the sensitivity of cells to stress by accelerating p53 activation. Upregulation of SnoN by stress could also accelerate the ageing process in mice. Oxidative stress can cause damage to DNA and serve as a second messenger to activate multiple pathways (Finkel, 2001; Liou and Storz, ; Rhee et al., 2000). We reported here, that DNA damage pathway is required for the induction of SnoN by oxidative stress. Exposure to UV light causing DNA damage in cells can induce the expression of SnoN as well. Exactly how UV treatment senses SnoN expression is still unclear. UV treatments could increase intracellular oxygen radicals in cells but other effects might be also important for the induction of SnoN as well. Drug treatments which inhibit ATM and ATR kinase activities have been shown to block the upregulation of SnoN. ATM and ATR might have compensatory mechanisms in SnoN induction. Removal of either ATM or ATR is not enough to block the induction as both must be inactivated to see an effect. This suggests that sensing ATM or ATM kinases by oxidative stress caused DNA damages leads to the induction of SnoN by some unknown mechanisms. In studying kinetic patterns of ATM activation in conjunction with p53 and SnoN expression, we have observed that the activation of ATM and increased expression of p53 first occurred at the earliest time after 4-hour of treatment with H2O2. Upregulation of SnoN was observed after 8-hour treatment and accordingly a further increased p53 levels were found after 24-hour treatment. It is believed that the secondary increase in p53 levels requires the induction of SnoN, an activator of p53. In SnoN null cells, the secondary increase in p53 levels at 24 hours did not occur although p53 levels did increase at 48 hours likely due to some other compensated pathways. It suggests that SnoN is involved into the activation of p53 during DNA damage. Elevating SnoN expression by stress functions as an accelerator to ensure the intensive p53 responses at the earlier time. Thurs, cellular responses to stress is delayed but not completely blocked when SnoN is removed from cells. The induction of SnoN by oxidative stress occurs at the transcription level. The increased SnoN mRNA was detected following treatment with H2O2 or Rotenone. It is also supported by the luciferase assay following the treatment with H2O2. SnoN promoter driven luciferase is increased by treatment of H2O2 although the increased activity is not very dramatic. It suggests that a cooperation of multiple regulations might occur during the treatment. This increment is not allowed us to further analyze the promoter in order to find out the responsive elements. When the specific elements present in the SnoN promoter sequence were examined, multiple sites capable of associating with different transcription factors were found including Sp1, NF-1 and TCF/LEF suggesting that the upregulation of SnoN might be controlled by multiple pathways. In the treatment with inhibitors, we found that the inhibitors, U0126 for Erk kinase and LY294002 for PI3K kinase was able to block the induction of SnoN by oxidative stress. MEK/Erk kinase and PI3K/Akt kinase pathways are the potential pathways activated by oxidative stress. Interestingly, overexpression of constitutively activated Akt in cells does not affect SnoN expression in the presence or absence of oxidative stress. It further supports the idea that multiple pathways are likely required to collaboratively regulate SnoN expression in response to stress since activation

Page 101: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

88

of single pathway is insufficient. It is obvious that further experiments need to be applied to explore the detailed mechanisms.

The induction of SnoN by oxidative stress indicates that SnoN might function as a stress transducer for p53 activation since elevated SnoN has been shown to have the capability of activating p53. Indeed, in cells with reduced SnoN expression, the activation of p53 is impaired. p53 could not be activated as control cells at the early stage of stress responses when SnoN was knocked down. These knock-down cells showed defective p53 mediated growth arrest and senescence response at the early stage of stress stimulation. However, the knock-down cells eventully stopped growing and entered senescence stage when incubated for a longer time. Accordingly, the stabilization and activation of p53 in these knock-down cells, even though were defective at the early stage, were caught up to the same level as control cells when they were inducated for a longer period of stress stimulation. It suggests that SnoN is important for the early stage of p53 activation under stress but the other pathways might have domimnat effects on p53 activation at the later stage. Interestingly, when the kinetic expression levels of SnoN and p53 in treatment with oxidative stress were compared, the immediate increase of p53 at the very early stage was very likely to depend on the activation of DNA damge pathway but not SnoN since the expression of SnoN is undetected at this point. However, when the level of SnoN was induced by oxidative stress, we have also observed a peak of p53 levels which is defective in SnoN null cells. This suggests that the further activation of p53 by stress is mediated by this upregulationed SnoN expression. Consistantly, the activation of p53 at the later stage was similar in WT and SnoN null cell. All these results support that SnoN takes part in the p53 dependent stress responses and is a signal traducer for stress mediated p53 activation. The timing and intensity of p53 activaition can guide the proper cellular responses to stress signals. It would be interesting to investigate whether this elevated SnoN has an instructive role in determining the specific p53 mediated cellular responses under stress.

In summary, the expression of SnoN is tightly controlled in cells. Oxidative stress can induce SnoN expression at the transcriptional level through the cooperation of multiple pathways. Induced SnoN expression can accelerate p53 activation by its direct binding to p53 and antagonizing Mdm2 activities. SnoN is a stress sensor which plays an important role in accelerating p53 dependent stress responses and contributes to the ageing processes in mice. SnoN can also serve as a marker to label the cells undertaking the high oxidative stress in vivo. It is not surprising that upregulation of SnoN was observed at certain stages of tissue development such as the pregnancy and early lactation stages of mammary gland development and also in some cancer samples. In each of these cases, cells are in the process of intensive growth and proliferation generating an excess of ROS due to the increased metabolic activities. These ROS might directly induce the expression of SnoN regulating stress responses and other physiological effects. The loss of precise control in SnoN induction by oxidative stress may contribute to some pathological processes such as cancer or ageing.

Page 102: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

89

Materials and methods

Cells and antibodies Primary MEFs were isolated from E13.5 day’s embryos following a published protocol (Serrano et al., 1997). MEFs were maintained in DMEM with 10% serum. ATM-/-; ATR flox/- and ATR flox/- MEFs were obtained from Dr. Eric Brown. 293T and RIE cells were maintained in DMEM with 10% serum. Hep3B cells were maintained in MEM with 10% serum. An anti-SnoN rabbit polyclonal antibody was raised against SnoN N-terminus fragment. Antibodies for p53 (ab26); p19Arf (ab 80) and cleaved Caspase 3 were purchased from Abcam; antibody for Tubulin from Santa Cruz; antibodies for phospho-ATM and phospho-Akt from Cell Signaling; antibodies for ATM and ATR from Genetex.

Transfection and Luciferase Hep3B cells were transiently transfected using Lipofectamine and Plus (Invitrogen) following manufacturer's recommendations. For the luciferase assay, 0.5 μg of pSnoN 3.2-Lux was co-transfected. Luciferase activity was measured 24 hr after stimulation with 70μM of H202.

Nuclear extraction and endogenous SnoN immunoprecipitation Nuclear extraction was performed in cells as described previously(Zhu et al., 2005). Basically, nuclei were released from RIE cells by passing cell through 18G 5/8 needle for 15 times. Then, nuclei were lysed in a buffer with 0.1% NP40. Endogenous proteins were isolated by immunoprecipitation with SnoN coated protein A beads.

RT-PCR Total RNA extraction, cDNA preparation and PCR were performed using standard protocol. The primers used to amplify SnoN gene by PCR were 5’ CTTAGATGACTATGGAGATGCATC 3’ and 5’ GTCAATATCACACTTCATGATGCTGTT 3’

SA-β-gal staining in cells and tissues MEFs were cultured in a standard medium in the presence or absence of N-acetyl-cysteine (10mM) or SB431542 (10μM) or LY290042 (10μM) to P6. Or cells were infected with retroviral constructs as described previously (Akhurst and Derynck, 2001; Pan et al., 2009; Zhu et al., 2006) . The senescence detection kit (Calbiochem) was applied to stain senescent cells following the instruction.

Cell treatment with ROS inducers and/or kinase inhibitors Primary MEFs (P3) were pretreated with or without Caffeine (5mM) or CGK733 (Indicated concentrations) or SB431542 (10μM), or PP2 (5μM) , or U0126 (3μM), or LY290042 (10μM) for 6 hrs and then treated with H2O2 (10μM) or Rotenone (4μM) for 24 hrs or indicated times.

Page 103: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

90

UV treatment Most medium in growing cells was removed with a residual amount of medium left in the culture plate. Cells were exposed to 20μJ/cm2 of UV. Fresh medium was added to treated cells and incubated for 8 hrs followed by analysis.

Retrovirus replication and infection Retrovirus vector based constructs were transfected into phoenix ecotropic cells to generate the high titer of virus as described previously (Zhu et al., 2006). The virus supernatants are used to incubate with proliferating MEFs in presence of 8μg/ml of polybrene. 48 hours later, infected cells can be used for analyses. BrdU incorporation assay Cells were incubated with medium containing 0.1mg/ml BrdU (Roche) for 6 hours. After fixation in icecold methanol, cells were incubated with anti-BrdU working solution (Roche) for 30 min at 37°C, stained with Alexa Fluor 488-conjugated goat anti-mouse-IgG (Invitrogen) and visualized by fluorescent microscopy.

Page 104: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

91

Figure legend

Figure 1: Oxidative stress can induce the expression of SnoN. A. wt or m/m MEFs were isolated from embryos and maintained in presence or absence of 10μM of NAC. At passage 6, senescence was detected by SA-β-gal staining and the levels of SnoN were examined by western blotting with anti-SnoN. Tubulin was used as a loading control. B. wt or m/m MEFs were isolated from embryos and maintained in presence of 20% or 3% of oxygen. At passage 13, senescence was detected by SA-β-gal staining and the levels of SnoN were examined by western blotting with anti-SnoN. Tubulin was used as a loading control. C. Oxidative stress is an inducer of SnoN expression. MEFs were treated with 60μM of H2O2 or 1μM of Rotenone for various times. The levels of SnoN were detected by western blotting with anti-SnoN. Tubulin was used as a loading control. D. The upregulation of SnoN by oxidative stress occur at transcription level. MEFs were treated with H2O2 for 24 hours. Semi-quantitative RT-PCR was used to detect the transcription of snoN gene. E. Oxidative stress increases SnoN promoter driven transcription. A luciferase construct carrying 3.2kb of region upstream of snoN gene transcription start site was introduced into MEFs. Cells were treated with various doses of H2O2 after 24 hours of transfection and luciferase activities were measured after 48 hours of transfection. F. RIEs were treated with H2O2 for various times. SnoN and its associated p53 were isolated by immunoprecipitation with anti-SnoN and detected by western blotting with anti-p53 or anti-SnoN. Tubulin was used a loading control. G. MEFs were treated with 20μj/cm2 of UV. The levels of SnoN were detected by western blotting with anti-SnoN. Tubulin was used as a loading control. Figure 2: Pathways mediate the upregulation of SnoN by oxidative stress. A. ATM/ATR kinases are involved in the induction of SnoN by oxidative stress. MEFs pretreated with or without 5μM of Caffeine, were treated with H2O2 for 24 hr. The levels of SnoN and p53 were examined by western blotting with anti-SnoN or anti-p53. The untreated cells were used as a control. Tubulin was used as a loading control. B. MEFs pretreated with or without indicated amounts of CGK733, were treated with H2O2 for 24 hr. The levels of SnoN and p53 were examined by western blotting with anti-SnoN or anti-p53. The untreated cells were used as a control. Tubulin was used as a loading control. C. ATM-/-; ATR flox/- or ATR flox/- MEFs infected with retrovirus based construct expressing Cre or vector were treated with H2O2 for 24 hours. The levels of SnoN, p53, ATM and ATR were examined by western blotting with anti-SnoN or anti-p53 or anti-ATM or anti-ATR. The untreated cells were used as a control. Tubulin was used as a loading control. D. SnoN is required for the activation of p53 at the early stage of DNA damage response. wt or SnoN null MEFs were treated with H2O2 for various times. The expression levels of p53, phospho-ATM and SnoN were detected by western blotting with anti-p53, anti-phospho-ATM or anti-SnoN. The untreated cells were used as a control. Tubulin was used as a loading control. E. PI3K kinase is required for the upregulation of SnoN by oxidative stress. MEFs were treated with 70μM of H2O2 for 24 hours following the pre-treatment of SB431542 (10μM) or PP2 (5μM) or U0126 (3μM) or LY290042 (10μM) or DMSO for 6 hours. The levels of SnoN were examined by western blotting with anti-SnoN. The untreated cells were used as a control. Tubulin was used as a loading control. F. wt or m/m MEFs were isolated from embryos and maintained in presence of SB431542 (10μM) or LY290042 (10μM) or DMSO. At passage 6, senescence was detected by SA-β-gal staining and

Page 105: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

92

the levels of SnoN in passage 3 and passage 6 were examined by western blotting with anti-SnoN. Tubulin was used as a loading control. G. Activation of Akt is not sufficient for the upregulation of SnoN by oxidative stress. MEFs infected with retrovirus based construct expressing Myr-Akt or vector were treated with or without H2O2 for 24 hours. The levels of SnoN and phospho-Akt were examined by western blotting with anti-SnoN or anti-phospho-Akt. Tubulin was used as a loading control. Figure 3: SnoN plays an important role in regulation of p53 dependent stress responses. A. SnoN is required for stress induced growth arrest. MEFs infected with retrovirus based construct expressing shSnoN or vector were treated with H2O2 for various times. Proliferating cells were visualized and counted in 4 different areas by BrdU staining. Nuclei were stained with DAPI. B. Cells treated with H2O2 for various time were stained by SA-β-gal staining. Senescent cells were visualized as counted in 4 different areas. C. SnoN is important for activation of p53 by oxidative stress. The expression levels of p53, acetyl-p53, phosph-ATM and SnoN in cells treated with H2O2 for various time were detected by western blotting with anti-p53, anti-acetyl-p53, anti-phosph-ATM and anti-SnoN. Tubulin was used as a loading control. D. SnoN is required for stress induced senescence in RIE cells. RIE cells infected with retrovirus based construct expressing shSnoN or vector were treated with H2O2 for various times. Proliferating cells were visualized and counted in 4 different areas by BrdU staining. Nuclei were stained with DAPI. E. The expression levels of p53 in RIEs with treatment of H2O2 for various time were detected in cell lysates by western blotting with anti-p53. Tubulin was used as a loading control. SnoN was isolated by immunprecipitation and detected by western blotting. Tubulin was used as a loading control.

Page 106: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

93

Figures

Figure 1. Oxidative stress can induce the expression of SnoN

Page 107: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

94

Figure 2. Pathways mediate the upregulation of SnoN by oxidative stress

Page 108: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

95

Figure 3: SnoN plays an important role in regulation of p53 dependent stress responses.

Page 109: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

96

Chapter 6: Discussion and future direction

Page 110: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

97

Discussion and future direction This dissertation has reported the results from three related projects. Firstly in studying

the roles of SnoN and Smad interaction in mice by knocking in mutant SnoN deficient in Smad binding, I have demonstrated that SnoN has an anti-oncogenic function in vivo through a Smad independent manner. In MEFs isolated from knock-in mice, the role of SnoN in senescence has been uncovered. In fact, elevated SnoN can induce premature senescence in primary cells. By studying the mechanisms underlying SnoN induced senescence, I have elucidated a pathway mediating the role of SnoN in senescence. Specifically, elevated SnoN physically interacts with PML and is localized to PML NBs. The localization of SnoN to PML NBs results in the activation of p53 and the subsequent induction of senescence. In term of the detailed mechanism of p53 activation by SnoN, I continue to explore the possibility of a direct connection between SnoN and p53. In fact, I have shown that SnoN can physically bind to p53 in PML NBs when both are highly expressed in cells. The binding of SnoN to p53 leads to the stabilization of p53 by competitively displacing Mdm2 and enhancement of p53 modification in PML NBs. By this way, SnoN can activate the p53 dependent transcription when recruited together with p53 to the promoters of target genes. SnoN plays an important role in p53 dependent stress responses by serving as an accelerator of p53 activation during stress responses, ensuring rapid and intensive p53 activation. Accelerated activation of p53 by elevated SnoN can induce premature senescence leading to the repression of oncogene induced tumorigenesis in cells. Moreover, accelerated p53 activation by elevated SnoN in mice can prevent carcinogen induced tumorigenesis and accelerate stress activated ageing processes. The studies of SnoN levels and p53 activation in stressed cells led me to examine the regulatory mechanisms of SnoN expression under stress conditions. While SnoN expression is relative low in normal unstressed cells, SnoN levels are significantly elevated in cells that are exposed to certain types of stresses including oxidative stress, UV and oncogenic stress. Interestingly, multiple pathways seem to cooperate together to induce SnoN expression in stressed cells. ATM/ATR kinase, MEK/Erk kinase and PI3K kinase pathways activated under stress conditions have been shown to be necessary for upregulation of SnoN by stresses. However, the factors directly contributing to this upregulation of SnoN require further characterization. In some respects, SnoN can serve as a stress sensor whose expression is regulated by stress signals. Elevated SnoN expression in stressed cells accelerates the activation of p53 to ensure the proper responses. Accelerated activation of p53 by mutant SnoN in knock-in mice prevents carcinogen induced tumorigenesis and also accelerates the ageing process. It is shown here, that elevated SnoN induces premature senescence in primary MEFs and some epithelial cells by activating p53. Interestingly, overexpression of SnoN in other epithelial cells such as many cancer cell lines and human fibroblast cells results in robust cell death. Why does the overexpression of SnoN fail to trigger senescence in those cells? One possibility may reflect the selective responses of p53 in different cell types. Since p53 can trigger either senescence or apoptosis to permanently remove the stressed cells depending on stress types, intensity and cell context, the different responses of SnoN in different cell types might be due to the various responses of p53 activation. The other possibility is that senescence may be controlled by different mechanisms in cells. p53 is the major player to regulate senescence responses in primary MEFs. However, in many cells types, activation of p53 and Rb pathways are both required for induction of senescence. We have observed rapid activation of p53 but unaffected Rb activity when SnoN is highly expressed in MEFs. Activation of p53 might be insufficient to induce premature senescence in cells in which the senescence responses are controlled by both p53 and Rb activities. However, we cannot completely rule out the possibility

Page 111: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

98

that the normal senescence controlling mechanisms are impaired in many cell lines under culture conditions. For example, defects in events downstream of p53 activation such as mutations in p21 gene, an important mediator for p53 activated senescence, can result in immortality in some cells. Activation of p53 in such cells might not have any normal senescence responses. It is important to explore the downstream mechanisms controlling SnoN induced senescence in order to better understand the different responses of cells to elevated SnoN. The expression levels of SnoN are relatively low in normal cells and adult tissues. Elevated SnoN is observed in certain stages of embryogenesis or in distinct stages of adult tissue development and in some cancer. In each of these cases, the cells all exhibit a high proliferative capability. How is SnoN induced in these cells? One potential factor is the elevated levels of ROS produced by proliferating cells reflective of the increased metabolism. Examination of SnoN expression in all tissues with high proliferative capability or increased exposure to high levels of ROS will better elucidate the role of ROS in the regulation of SnoN expression. Why these cells can tolerate such high levels of SnoN is still unclear. In the studies by Ari, Nadine and Erwan, it was observed that SnoN is localized in the cytoplasm of normal tissues with high cell density. The mechanisms controlling the cytoplasmic and nuclear localization of SnoN are still not completely understood. The cytoplasmic localization of SnoN might provide an explanation for the lacking of response despite high levels of SnoN in certain cells or tissues. Since the activation of p53 dependent responses by SnoN must be occur at promoters of p53 target genes in nucleus, the cytoplasmic SnoN remains ineffective. Definitely, a better understanding of the mechanisms controlling SnoN localization will help reveal the mechanisms leading to escaping from p53 dependent anti-proliferation effects in cells with high levels of SnoN.

In terms of the levels of SnoN in human cancer samples, the data is very limited and controversial. In colorectal cancer with microsatellite instability, 39% of samples show an increased expression of SnoN while 33% of samples have decreased expression of SnoN suggesting SnoN might have different effects in various cancer samples depending on cancer types and stages of cancer progression. ROS levels are often elevated in cells with high proliferative potential. Upregulation of SnoN elicited by the increased ROS levels might occur in some aberrantly growing cells such as in hyplasia or benign tumors, thus preventing further progression of those aberrant cells. However, elevated SnoN is not always observed in rapidly growing cancer cells. It is possible that some tumor cells can specifically downregulate SnoN expression or have defects in response to stress. Interestingly, elevated SnoN is indeed observed in many cancer cells. How do these cells bypass the negative effects of SnoN on tumor growth? One possibility is that these cells may silence the pathway mediating the anti-oncogenic function of SnoN through various mechanisms, such as causing mutations in p53 in order to promote tumor progress through the reserved the pro-oncogenic capability of SnoN. In many cases, the elevated expression of SnoN is due to amplification of the snoN gene in cancer cells. Chromosome amplification might indicate an existing defect in the p53 responses in these types of cancers, allowing chromosome instability to occur. Understanding the relationship between the levels of SnoN and p53 activities at different stages of tumorigenesis will provide a clear picture about the role of SnoN in human cancer. Knock-in mice with a stable form of mutant SnoN show signs of premature ageing associated phenotypes. The ageing related phenotypes are presumably associated with the activation of p53 by elevated SnoN in m/m mice but not due to the enhanced Smad activities, since ageing related phenotypes are not observed in SnoN knockout mice in which enhanced Smad activities are enhanced to the level similar as m/m mice. It is worth comparing the

Page 112: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

99

expression levels of SnoN between wt and m/m mice at different ages in the tissues such as in kidney and skin where an accumulation of senescent cells have been detected in the old aged m/m mice. The comparison of SnoN expression at the tissue level might also provide the explanation for the premature ageing phenotypes in m/m mice that can not exactly resemble the phenotypes of hyperactive p53 mice. The other interesting observation is the effect of elevated Ski in MEFs. I have also observed that overexpression of Ski in primary MEFs can induce premature senescence accompanied by significantly increased cell death (Data is not shown). In a previous study by Andy in lab, it was observed that overexpressed Ski resulted in senescence-like morphologies in NIH3T3 cells. The N-terminal portions of SnoN and Ski have very high homology and Ski has been reported to bind with PML through its SANDs domain to localize to PML NBs similar to SnoN. It seems that SnoN and Ski might have the similar effects on activating p53 dependent responses. Additionally, like SnoN, Ski has been shown to posses both pro-oncogenic and anti-oncogenic functions as well. Interestingly, SnoN knockout mice behaved similarly as wt mice in carcinogen induced tumorigenesis or ageing process, although SnoN has been shown to play an important role in regulating stress activated p53 activities. It might indicate that Ski have a compensation function for SnoN if they are functionally similar. When SnoN is removed, Ski may compensate the function of SnoN in tumorigenesis and ageing process. On the other hand, compared to SnoN, elevated Ski might also have some unique characteristics since it also induces cell death in primary MEFs. The characterization of MEFs with knock-in mutant Ski deficient in Smad binding will help to understand the connection of Ski with the PML-p53 pathway and eventually help to clarify mechanistically the importance of Ski in tumorigenesis.

Page 113: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

100

Chapter 7: Reference

Page 114: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

101

Reference Abdollah, S., Macias-Silva, M., Tsukazaki, T., Hayashi, H., Attisano, L. and Wrana, J.L. (1997) TbetaRI phosphorylation of Smad2 on Ser465 and Ser467 is required for Smad2-Smad4 complex formation and signaling. J Biol Chem, 272, 27678-27685. Agarwal, C., Hembree, J.R., Rorke, E.A. and Eckert, R.L. (1994) Transforming growth factor beta 1 regulation of metalloproteinase production in cultured human cervical epithelial cells. Cancer Res, 54, 943-949. Akagi, I., Miyashita, M., Makino, H., Nomura, T., Hagiwara, N., Takahashi, K., Cho, K., Mishima, T., Takizawa, T. and Tajiri, T. (2008) SnoN overexpression is predictive of poor survival in patients with esophageal squamous cell carcinoma. Ann Surg Oncol, 15, 2965-2975. Akhurst, R.J. and Derynck, R. (2001) TGF-beta signaling in cancer--a double-edged sword. Trends Cell Biol, 11, S44-51. Akiyoshi, S., Inoue, H., Hanai, J., Kusanagi, K., Nemoto, N., Miyazono, K. and Kawabata, M. (1999) c-Ski acts as a transcriptional co-repressor in transforming growth factor-beta signaling through interaction with smads. J Biol Chem, 274, 35269-35277. Anzano, M.A., Roberts, A.B. and Sporn, M.B. (1986) Anchorage-independent growth of primary rat embryo cells is induced by platelet-derived growth factor and inhibited by type-beta transforming growth factor. J Cell Physiol, 126, 312-318. Attisano, L., Carcamo, J., Ventura, F., Weis, F.M., Massague, J. and Wrana, J.L. (1993) Identification of human activin and TGF beta type I receptors that form heteromeric kinase complexes with type II receptors. Cell, 75, 671-680. Balint, E.E. and Vousden, K.H. (2001) Activation and activities of the p53 tumour suppressor protein. Br J Cancer, 85, 1813-1823. Balmain, A., Brown, K., Akhurst, R.J. and Fee, F.M. (1988) Molecular analysis of chemical carcinogenesis in the skin. Br J Cancer Suppl, 9, 72-75. Barkas, A.E., Brodeur, D. and Stavnezer, E. (1986) Polyproteins containing a domain encoded by the V-SKI oncogene are located in the nuclei of SKV-transformed cells. Virology, 151, 131-138. Bartkova, J., Rezaei, N., Liontos, M., Karakaidos, P., Kletsas, D., Issaeva, N., Vassiliou, L.V., Kolettas, E., Niforou, K., Zoumpourlis, V.C., Takaoka, M., Nakagawa, H., Tort, F., Fugger, K., Johansson, F., Sehested, M., Andersen, C.L., Dyrskjot, L., Orntoft, T., Lukas, J., Kittas, C., Helleday, T., Halazonetis, T.D., Bartek, J. and Gorgoulis, V.G. (2006) Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature, 444, 633-637. Bassing, C.H., Yingling, J.M. and Wang, X.F. (1994) Receptors for the TGF-beta ligand family. Vitam Horm, 48, 111-156. Benz, C.C. and Yau, C. (2008) Ageing, oxidative stress and cancer: paradigms in parallax. Nat Rev Cancer, 8, 875-879. Bernardi, R. and Pandolfi, P.P. (2007) Structure, dynamics and functions of promyelocytic leukaemia nuclear bodies. Nat Rev Mol Cell Biol, 8, 1006-1016. Blobe, G.C., Schiemann, W.P. and Lodish, H.F. (2000) Role of transforming growth factor beta in human disease. N Engl J Med, 342, 1350-1358. Bode, A.M. and Dong, Z. (2004) Post-translational modification of p53 in tumorigenesis. Nat Rev Cancer, 4, 793-805.

Page 115: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

102

Bonni, S., Wang, H.R., Causing, C.G., Kavsak, P., Stroschein, S.L., Luo, K. and Wrana, J.L. (2001) TGF-beta induces assembly of a Smad2-Smurf2 ubiquitin ligase complex that targets SnoN for degradation. Nat Cell Biol, 3, 587-595. Bottinger, E.P., Jakubczak, J.L., Roberts, I.S., Mumy, M., Hemmati, P., Bagnall, K., Merlino, G. and Wakefield, L.M. (1997a) Expression of a dominant-negative mutant TGF-beta type II receptor in transgenic mice reveals essential roles for TGF-beta in regulation of growth and differentiation in the exocrine pancreas. Embo J, 16, 2621-2633. Bottinger, E.P., Letterio, J.J. and Roberts, A.B. (1997b) Biology of TGF-beta in knockout and transgenic mouse models. Kidney Int, 51, 1355-1360. Boyer, P.L., Colmenares, C., Stavnezer, E. and Hughes, S.H. (1993) Sequence and biological activity of chicken snoN cDNA clones. Oncogene, 8, 457-466. Braig, M., Lee, S., Loddenkemper, C., Rudolph, C., Peters, A.H., Schlegelberger, B., Stein, H., Dorken, B., Jenuwein, T. and Schmitt, C.A. (2005) Oncogene-induced senescence as an initial barrier in lymphoma development. Nature, 436, 660-665. Bravou, V., Antonacopoulou, A., Papadaki, H., Floratou, K., Stavropoulos, M., Episkopou, V., Petropoulou, C. and Kalofonos, H. (2009) TGF-beta repressors SnoN and Ski are implicated in human colorectal carcinogenesis. Cell Oncol, 31, 41-51. Brooks, C.L. and Gu, W. (2006) p53 ubiquitination: Mdm2 and beyond. Mol Cell, 21, 307-315. Buess, M., Terracciano, L., Reuter, J., Ballabeni, P., Boulay, J.L., Laffer, U., Metzger, U., Herrmann, R. and Rochlitz, C. (2004) Amplification of SKI is a prognostic marker in early colorectal cancer. Neoplasia, 6, 207-212. Campisi, J. (2001) Cellular senescence as a tumor-suppressor mechanism. Trends Cell Biol, 11, S27-31. Campisi, J. (2005) Senescent cells, tumor suppression, and organismal ageing: good citizens, bad neighbors. Cell, 120, 513-522. Campisi, J. and d'Adda di Fagagna, F. (2007) Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol, 8, 729-740. Chang, H., Huylebroeck, D., Verschueren, K., Guo, Q., Matzuk, M.M. and Zwijsen, A. (1999) Smad5 knockout mice die at mid-gestation due to multiple embryonic and extraembryonic defects. Development, 126, 1631-1642. Chen, T., Carter, D., Garrigue-Antar, L. and Reiss, M. (1998) Transforming growth factor beta type I receptor kinase mutant associated with metastatic breast cancer. Cancer Res, 58, 4805-4810. Chen, W. and Wahl, S.M. (2003) TGF-beta: the missing link in CD4+CD25+ regulatory T cell-mediated immunosuppression. Cytokine Growth Factor Rev, 14, 85-89. Chen, Z., Trotman, L.C., Shaffer, D., Lin, H.K., Dotan, Z.A., Niki, M., Koutcher, J.A., Scher, H.I., Ludwig, T., Gerald, W., Cordon-Cardo, C. and Pandolfi, P.P. (2005) Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature, 436, 725-730. Chia, J.A., Simms, L.A., Cozzi, S.J., Young, J., Jass, J.R., Walsh, M.D., Spring, K.J., Leggett, B.A. and Whitehall, V.L. (2006) SnoN expression is differently regulated in microsatellite unstable compared with microsatellite stable colorectal cancers. BMC Cancer, 6, 252.

Page 116: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

103

Cohen, S.B., Nicol, R. and Stavnezer, E. (1998) A domain necessary for the transforming activity of SnoN is required for specific DNA binding, transcriptional repression and interaction with TAF(II)110. Oncogene, 17, 2505-2513. Collado, M., Blasco, M.A. and Serrano, M. (2007) Cellular senescence in cancer and ageing. Cell, 130, 223-233. Colmenares, C. and Stavnezer, E. (1989) The ski oncogene induces muscle differentiation in quail embryo cells. Cell, 59, 293-303. Colmenares, C., Teumer, J.K. and Stavnezer, E. (1991) Transformation-defective v-ski induces MyoD and myogenin expression but not myotube formation. Mol Cell Biol, 11, 1167-1170. Courtois-Cox, S., Jones, S.L. and Cichowski, K. (2008) Many roads lead to oncogene-induced senescence. Oncogene. de Stanchina, E., Querido, E., Narita, M., Davuluri, R.V., Pandolfi, P.P., Ferbeyre, G. and Lowe, S.W. (2004) PML is a direct p53 target that modulates p53 effector functions. Mol Cell, 13, 523-535. Dean, D.C., Newby, R.F. and Bourgeois, S. (1988) Regulation of fibronectin biosynthesis by dexamethasone, transforming growth factor beta, and cAMP in human cell lines. J Cell Biol, 106, 2159-2170. Deheuninck, J. and Luo, K. (2009) Ski and SnoN, potent negative regulators of TGF-beta signaling. Cell Res, 19, 47-57. Delany, A.M. and Canalis, E. (2001) The metastasis-associated metalloproteinase stromelysin-3 is induced by transforming growth factor-beta in osteoblasts and fibroblasts. Endocrinology, 142, 1561-1566. Dellaire, G., Ching, R.W., Ahmed, K., Jalali, F., Tse, K.C., Bristow, R.G. and Bazett-Jones, D.P. (2006) Promyelocytic leukemia nuclear bodies behave as DNA damage sensors whose response to DNA double-strand breaks is regulated by NBS1 and the kinases ATM, Chk2, and ATR. J Cell Biol, 175, 55-66. Derynck, R., Akhurst, R.J. and Balmain, A. (2001a) TGF-beta signaling in tumor suppression and cancer progression. Nat Genet, 29, 117-129. Derynck, R., Akhurst, R.J. and Balmain, A. (2001b) TGF-beta signaling in tumor suppression and cancer progression. Nat Genet, 29, 117-129. Derynck, R., Goeddel, D.V., Ullrich, A., Gutterman, J.U., Williams, R.D., Bringman, T.S. and Berger, W.H. (1987) Synthesis of messenger RNAs for transforming growth factors alpha and beta and the epidermal growth factor receptor by human tumors. Cancer Res, 47, 707-712. Derynck, R., Jarrett, J.A., Chen, E.Y., Eaton, D.H., Bell, J.R., Assoian, R.K., Roberts, A.B., Sporn, M.B. and Goeddel, D.V. (1985) Human transforming growth factor-beta complementary DNA sequence and expression in normal and transformed cells. Nature, 316, 701-705. Derynck, R., Zhang, Y. and Feng, X.H. (1998) Smads: transcriptional activators of TGF-beta responses. Cell, 95, 737-740. Di Micco, R., Fumagalli, M., Cicalese, A., Piccinin, S., Gasparini, P., Luise, C., Schurra, C., Garre, M., Nuciforo, P.G., Bensimon, A., Maestro, R., Pelicci, P.G. and d'Adda di Fagagna, F. (2006) Oncogene-induced senescence is a DNA damage response triggered by DNA hyper-replication. Nature, 444, 638-642.

Page 117: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

104

Di Micco, R., Fumagalli, M. and di Fagagna, F. (2007) Breaking news: high-speed race ends in arrest--how oncogenes induce senescence. Trends Cell Biol, 17, 529-536. Dickson, R.B., Kasid, A., Huff, K.K., Bates, S.E., Knabbe, C., Bronzert, D., Gelmann, E.P. and Lippman, M.E. (1987) Activation of growth factor secretion in tumorigenic states of breast cancer induced by 17 beta-estradiol or v-Ha-ras oncogene. Proc Natl Acad Sci U S A, 84, 837-841. Donehower, L.A. (2009) Using mice to examine p53 functions in cancer, ageing, and longevity. Cold Spring Harb Perspect Biol, 1, a001081. Edmiston, J.S., Yeudall, W.A., Chung, T.D. and Lebman, D.A. (2005) Inability of transforming growth factor-beta to cause SnoN degradation leads to resistance to transforming growth factor-beta-induced growth arrest in esophageal cancer cells. Cancer Res, 65, 4782-4788. Efeyan, A. and Serrano, M. (2007) p53: guardian of the genome and policeman of the oncogenes. Cell Cycle, 6, 1006-1010. el-Deiry, W.S., Tokino, T., Waldman, T., Oliner, J.D., Velculescu, V.E., Burrell, M., Hill, D.E., Healy, E., Rees, J.L., Hamilton, S.R. and et al. (1995) Topological control of p21WAF1/CIP1 expression in normal and neoplastic tissues. Cancer Res, 55, 2910-2919. Engle, S.J., Hoying, J.B., Boivin, G.P., Ormsby, I., Gartside, P.S. and Doetschman, T. (1999) Transforming growth factor beta1 suppresses nonmetastatic colon cancer at an early stage of tumorigenesis. Cancer Res, 59, 3379-3386. Eppert, K., Scherer, S.W., Ozcelik, H., Pirone, R., Hoodless, P., Kim, H., Tsui, L.C., Bapat, B., Gallinger, S., Andrulis, I.L., Thomsen, G.H., Wrana, J.L. and Attisano, L. (1996) MADR2 maps to 18q21 and encodes a TGFbeta-regulated MAD-related protein that is functionally mutated in colorectal carcinoma. Cell, 86, 543-552. Fantini, M.C., Becker, C., Monteleone, G., Pallone, F., Galle, P.R. and Neurath, M.F. (2004) Cutting edge: TGF-beta induces a regulatory phenotype in CD4+CD25- T cells through Foxp3 induction and down-regulation of Smad7. J Immunol, 172, 5149-5153. Feng, X.H. and Derynck, R. (2005) Specificity and versatility in tgf-beta signaling through Smads. Annu Rev Cell Dev Biol, 21, 659-693. Feng, Z., Hu, W., Rajagopal, G. and Levine, A.J. (2008) The tumor suppressor p53: cancer and ageing. Cell Cycle, 7, 842-847. Ferbeyre, G., de Stanchina, E., Querido, E., Baptiste, N., Prives, C. and Lowe, S.W. (2000) PML is induced by oncogenic ras and promotes premature senescence. Genes Dev, 14, 2015-2027. Finkel, T. (2001) Reactive oxygen species and signal transduction. IUBMB Life, 52, 3-6. Frescas, D., Guardavaccaro, D., Bassermann, F., Koyama-Nasu, R. and Pagano, M. (2007) JHDM1B/FBXL10 is a nucleolar protein that represses transcription of ribosomal RNA genes. Nature, 450, 309-313. Frolik, C.A., Dart, L.L., Meyers, C.A., Smith, D.M. and Sporn, M.B. (1983) Purification and initial characterization of a type beta transforming growth factor from human placenta. Proc Natl Acad Sci U S A, 80, 3676-3680. Goddard, A.D., Yuan, J.Q., Fairbairn, L., Dexter, M., Borrow, J., Kozak, C. and Solomon, E. (1995) Cloning of the murine homolog of the leukemia-associated PML gene. Mamm Genome, 6, 732-737.

Page 118: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

105

Goggins, M., Shekher, M., Turnacioglu, K., Yeo, C.J., Hruban, R.H. and Kern, S.E. (1998) Genetic alterations of the transforming growth factor beta receptor genes in pancreatic and biliary adenocarcinomas. Cancer Res, 58, 5329-5332. Gorelik, L. and Flavell, R.A. (2000) Abrogation of TGFbeta signaling in T cells leads to spontaneous T cell differentiation and autoimmune disease. Immunity, 12, 171-181. Gorelik, L. and Flavell, R.A. (2002) Transforming growth factor-beta in T-cell biology. Nat Rev Immunol, 2, 46-53. Gorsch, S.M., Memoli, V.A., Stukel, T.A., Gold, L.I. and Arrick, B.A. (1992) Immunohistochemical staining for transforming growth factor beta 1 associates with disease progression in human breast cancer. Cancer Res, 52, 6949-6952. Gorska, A.E., Joseph, H., Derynck, R., Moses, H.L. and Serra, R. (1998) Dominant-negative interference of the transforming growth factor beta type II receptor in mammary gland epithelium results in alveolar hyperplasia and differentiation in virgin mice. Cell Growth Differ, 9, 229-238. Grunert, S., Jechlinger, M. and Beug, H. (2003) Diverse cellular and molecular mechanisms contribute to epithelial plasticity and metastasis. Nat Rev Mol Cell Biol, 4, 657-665. Guzman-Ayala, M., Lee, K.L., Mavrakis, K.J., Goggolidou, P., Norris, D.P. and Episkopou, V. (2009) Graded Smad2/3 activation is converted directly into levels of target gene expression in embryonic stem cells. PLoS ONE, 4, e4268. Hahn, S.A., Schutte, M., Hoque, A.T., Moskaluk, C.A., da Costa, L.T., Rozenblum, E., Weinstein, C.L., Fischer, A., Yeo, C.J., Hruban, R.H. and Kern, S.E. (1996) DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. [see comments]. Science, 271, 350-353. Hanahan, D. and Weinberg, R.A. (2000) The hallmarks of cancer. Cell, 100, 57-70. Harkin, D.P. and Hall, P.A. (2000) Measuring a cell's response to stress: the p53 pathway. Genome Biol, 1, REVIEWS105. Hayashi, I. and Carr, B.I. (1985) DNA synthesis in rat hepatocytes: inhibition by a platelet factor and stimulation by an endogenous factor. J Cell Physiol, 125, 82-90. Hayflick, L. (1965) The Limited in Vitro Lifetime of Human Diploid Cell Strains. Exp Cell Res, 37, 614-636. He, J., Tegen, S.B., Krawitz, A.R., Martin, G.S. and Luo, K. (2003) The transforming activity of Ski and SnoN is dependent on their ability to repress the activity of Smad proteins. J Biol Chem, 278, 30540-30547. Huber, S., Schramm, C., Lehr, H.A., Mann, A., Schmitt, S., Becker, C., Protschka, M., Galle, P.R., Neurath, M.F. and Blessing, M. (2004) Cutting edge: TGF-beta signaling is required for the in vivo expansion and immunosuppressive capacity of regulatory CD4+CD25+ T cells. J Immunol, 173, 6526-6531. Hussain, S.P. and Harris, C.C. (2006) p53 biological network: at the crossroads of the cellular-stress response pathway and molecular carcinogenesis. J Nippon Med Sch, 73, 54-64. Ignotz, R.A. and Massague, J. (1986) Transforming growth factor-beta stimulates the expression of fibronectin and collagen and their incorporation into the extracellular matrix. J Biol Chem, 261, 4337-4345.

Page 119: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

106

Ikeuchi, Y., Stegmuller, J., Netherton, S., Huynh, M.A., Masu, M., Frank, D., Bonni, S. and Bonni, A. (2009) A SnoN-Ccd1 pathway promotes axonal morphogenesis in the mammalian brain. J Neurosci, 29, 4312-4321. Imoto, I., Pimkhaokham, A., Fukuda, Y., Yang, Z.Q., Shimada, Y., Nomura, N., Hirai, H., Imamura, M. and Inazawa, J. (2001) SNO is a probable target for gene amplification at 3q26 in squamous-cell carcinomas of the esophagus. Biochem Biophys Res Commun, 286, 559-565. Inagaki, M., Togawa, K., Carr, B.I., Ghosh, K. and Cohen, J.S. (1992) Antisense oligonucleotides: inhibition of liver cell proliferation and in vivo disposition. Transplant Proc, 24, 2971-2972. Itahana, K., Campisi, J. and Dimri, G.P. (2004) Mechanisms of cellular senescence in human and mouse cells. Biogerontology, 5, 1-10. Itoh, S., Itoh, F., Goumans, M.J. and Ten Dijke, P. (2000) Signaling of transforming growth factor-beta family members through Smad proteins. Eur J Biochem, 267, 6954-6967. Jahchan, N.S. and Luo, K. SnoN in mammalian development, function and diseases. Curr Opin Pharmacol. Jahchan, N.S., You, Y.H., Muller, W.J. and Luo, K. Transforming growth factor-beta regulator SnoN modulates mammary gland branching morphogenesis, postlactational involution, and mammary tumorigenesis. Cancer Res, 70, 4204-4213. Kakonen, S.M., Selander, K.S., Chirgwin, J.M., Yin, J.J., Burns, S., Rankin, W.A., Grubbs, B.G., Dallas, M., Cui, Y. and Guise, T.A. (2002) Transforming growth factor-beta stimulates parathyroid hormone-related protein and osteolytic metastases via Smad and mitogen-activated protein kinase signaling pathways. J Biol Chem, 277, 24571-24578. Kehrl, J.H., Wakefield, L.M., Roberts, A.B., Jakowlew, S., Alvarez-Mon, M., Derynck, R., Sporn, M.B. and Fauci, A.S. (1986) Production of transforming growth factor beta by human T lymphocytes and its potential role in the regulation of T cell growth. J Exp Med, 163, 1037-1050. Krakowski, A.R., Laboureau, J., Mauviel, A., Bissell, M.J. and Luo, K. (2005) Cytoplasmic SnoN in normal tissues and nonmalignant cells antagonizes TGF-beta signaling by sequestration of the Smad proteins. Proc Natl Acad Sci U S A, 102, 12437-12442. Kroening, S., Solomovitch, S., Sachs, M., Wullich, B. and Goppelt-Struebe, M. (2009) Regulation of connective tissue growth factor (CTGF) by hepatocyte growth factor in human tubular epithelial cells. Nephrol Dial Transplant, 24, 755-762. Kruse, J.P. and Gu, W. (2009) Modes of p53 regulation. Cell, 137, 609-622. Kurisaki, A., Kose, S., Yoneda, Y., Heldin, C.H. and Moustakas, A. (2001) Transforming Growth Factor-beta Induces Nuclear Import of Smad3 in an Importin-beta1 and Ran-dependent Manner. Mol Biol Cell, 12, 1079-1091. Lane, D.P. (1992) Cancer. p53, guardian of the genome. Nature, 358, 15-16. Levy, L., Howell, M., Das, D., Harkin, S., Episkopou, V. and Hill, C.S. (2007) Arkadia activates Smad3/Smad4-dependent transcription by triggering signal-induced SnoN degradation. Mol Cell Biol, 27, 6068-6083. Li, Y., Turck, C.M., Teumer, J.K. and Stavnezer, E. (1986) Unique sequence, ski, in Sloan-Kettering avian retroviruses with properties of a new cell-derived oncogene. J Virol, 57, 1065-1072. Liou, G.Y. and Storz, P. Reactive oxygen species in cancer. Free Radic Res, 44, 479-496.

Page 120: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

107

Liu, H., Colavitti, R., Rovira, II and Finkel, T. (2005) Redox-dependent transcriptional regulation. Circ Res, 97, 967-974. Ljungman, M. (2007) The transcription stress response. Cell Cycle, 6, 2252-2257. Luo, K. (2004) Ski and SnoN: negative regulators of TGF-beta signaling. Curr Opin Genet Dev, 14, 65-70. Luo, K. and Lodish, H.F. (1996) Signaling by chimeric erythropoietin-TGF-beta receptors: homodimerization of the cytoplasmic domain of the type I TGF-beta receptor and heterodimerization with the type II receptor are both required for intracellular signal transduction. Embo J, 15, 4485-4496. Ma, Q. Transcriptional responses to oxidative stress: pathological and toxicological implications. Pharmacol Ther, 125, 376-393. Macias-Silva, M., Abdollah, S., Hoodless, P.A., Pirone, R., Attisano, L. and Wrana, J.L. (1996) MADR2 is a substrate of the TGFbeta receptor and its phosphorylation is required for nuclear accumulation and signaling. Cell, 87, 1215-1224. Mallette, F.A. and Ferbeyre, G. (2007) The DNA damage signaling pathway connects oncogenic stress to cellular senescence. Cell Cycle, 6, 1831-1836. Marcotte, R. and Wang, E. (2002) Replicative senescence revisited. J Gerontol A Biol Sci Med Sci, 57, B257-269. Markowitz, S., Wang, J., Myeroff, L., Parsons, R., Sun, L., Lutterbaugh, J., Fan, R.S., Zborowska, E., Kinzler, K.W., Vogelstein, B., Brattain, M. and Willson, J.K.V. (1995) Inactivation of the type II TGF-β receptor in colon cancer cells with microsatellite instability. Science, 268, 1336-1338. Massague, J. (1990) The transforming growth factor-beta family. Annu Rev Cell Biol, 6, 597-641. Massague, J. (1998) TGF-beta signal transduction. Annu Rev Biochem, 67, 753-791. Massague, J. (2000) How cells read TGF-beta signals. Nat Rev Mol Cell Biol, 1, 169-178. Massague, J., Andres, J., Attisano, L., Cheifetz, S., Lopez-Casillas, F., Ohtsuki, M. and Wrana, J.L. (1992) TGF-beta receptors. Mol Reprod Dev, 32, 99-104. Massague, J. and Wotton, D. (2000) Transcriptional control by the TGF-beta/Smad signaling system. Embo J, 19, 1745-1754. Michael, D. and Oren, M. (2003) The p53-Mdm2 module and the ubiquitin system. Semin Cancer Biol, 13, 49-58. Michaloglou, C., Vredeveld, L.C., Soengas, M.S., Denoyelle, C., Kuilman, T., van der Horst, C.M., Majoor, D.M., Shay, J.W., Mooi, W.J. and Peeper, D.S. (2005) BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature, 436, 720-724. Moustakas, A., Souchelnytskyi, S. and Heldin, C.H. (2001) Smad regulation in TGF-beta signal transduction. J Cell Sci, 114, 4359-4369. Muraoka-Cook, R.S., Kurokawa, H., Koh, Y., Forbes, J.T., Roebuck, L.R., Barcellos-Hoff, M.H., Moody, S.E., Chodosh, L.A. and Arteaga, C.L. (2004) Conditional overexpression of active transforming growth factor beta1 in vivo accelerates metastases of transgenic mammary tumors. Cancer Res, 64, 9002-9011. Myeroff, L.L., Parsons, R., Kim, S.J., Hedrick, L., Cho, K.R., Orth, K., Mathis, M., Kinzler, K.W., Lutterbaugh, J., Park, K. and et, a. (1995) A transforming growth factor beta receptor type II gene mutation common in colon and gastric but rare in endometrial cancers with microsatellite instability. Cancer Res, 55, 5545-5547.

Page 121: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

108

Nagano, Y., Mavrakis, K.J., Lee, K.L., Fujii, T., Koinuma, D., Sase, H., Yuki, K., Isogaya, K., Saitoh, M., Imamura, T., Episkopou, V., Miyazono, K. and Miyazawa, K. (2007) Arkadia induces degradation of SnoN and c-Ski to enhance transforming growth factor-beta signaling. J Biol Chem, 282, 20492-20501. Nanjundan, M., Cheng, K.W., Zhang, F., Lahad, J., Kuo, W.L., Schmandt, R., Smith-McCune, K., Fishman, D., Gray, J.W. and Mills, G.B. (2008) Overexpression of SnoN/SkiL, amplified at the 3q26.2 locus, in ovarian cancers: a role in ovarian pathogenesis. Mol Oncol, 2, 164-181. Nomura, N., Sasamoto, S., Ishii, S., Date, T., Matsui, M. and Ishizaki, R. (1989) Isolation of human cDNA clones of ski and the ski-related gene, sno. Nucleic Acids Res, 17, 5489-5500. Nomura, T., Khan, M.M., Kaul, S.C., Dona, H.-D., Wadhwa, R., Colmenares, C., Kohno, I. and Ishii, S. (1999) Ski is a component of the histone deacetylase complex required for transcriptional repression by Mad and thyroid horomone receptor. Genes Dev, 13, 412-423. Oh, S.P., Seki, T., Goss, K.A., Imamura, T., Yi, Y., Donahoe, P.K., Li, L., Miyazono, K., ten Dijke, P., Kim, S. and Li, E. (2000) Activin receptor-like kinase 1 modulates transforming growth factor-beta 1 signaling in the regulation of angiogenesis. Proc Natl Acad Sci U S A, 97, 2626-2631. Ohtaki, N., Yamaguchi, A., Goi, T., Fukaya, T., Takeuchi, K., Katayama, K., Hirose, K. and Urano, T. (2001) Somatic alterations of the DPC4 and Madr2 genes in colorectal cancers and relationship to metastasis. Int J Oncol, 18, 265-270. Ohue, M., Tomita, N., Monden, T., Miyoshi, Y., Ohnishi, T., Izawa, H., Kawabata, Y., Sasaki, M., Sekimoto, M., Nishisho, I., Shiozaki, H. and Monden, M. (1996) Mutations of the transforming growth factor beta type II receptor gene and microsatellite instability in gastric cancer. Int J Cancer, 68, 203-206. Oren, M., Damalas, A., Gottlieb, T., Michael, D., Taplick, J., Leal, J.F., Maya, R., Moas, M., Seger, R., Taya, Y. and Ben-Ze'Ev, A. (2002) Regulation of p53: intricate loops and delicate balances. Ann N Y Acad Sci, 973, 374-383. Oshima, M., Oshima, H. and Taketo, M.M. (1996) TGF-beta receptor type II deficiency results in defects of yolk sac hematopoiesis and vasculogenesis. Dev Biol, 179, 297-302. Pan, D., Zhu, Q. and Luo, K. (2009) SnoN functions as a tumour suppressor by inducing premature senescence. Embo J, 28, 3500-3513. Park, K., Kim, S.J., Bang, Y.J., Park, J.G., Kim, N.K., Roberts, A.B. and Sporn, M.B. (1994) Genetic changes in the transforming growth factor beta (TGF-beta) type II receptor gene in human gastric cancer cells: correlation with sensitivity to growth inhibition by TGF-beta. Proc Natl Acad Sci U S A, 91, 8772-8776. Parrinello, S., Samper, E., Krtolica, A., Goldstein, J., Melov, S. and Campisi, J. (2003) Oxygen sensitivity severely limits the replicative lifespan of murine fibroblasts. Nat Cell Biol, 5, 741-747. Pearson, M., Carbone, R., Sebastiani, C., Cioce, M., Fagioli, M., Saito, S., Higashimoto, Y., Appella, E., Minucci, S., Pandolfi, P.P. and Pelicci, P.G. (2000) PML regulates p53 acetylation and premature senescence induced by oncogenic Ras. Nature, 406, 207-210. Pearson, M. and Pelicci, P.G. (2001) PML interaction with p53 and its role in apoptosis and replicative senescence. Oncogene, 20, 7250-7256. Pearson-White, S. (1993) SnoI, a novel alternatively spliced isoform of the ski protooncogene homolog, sno. Nucleic Acids Res, 21, 4632-4638.

Page 122: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

109

Pearson-White, S. and Crittenden, R. (1997) Proto-oncogene Sno expression, alternative isoforms and immediate early serum response. Nucleic Acids Res, 25, 2930-2937. Pearson-White, S. and McDuffie, M. (2003) Defective T-cell activation is associated with augmented transforming growth factor Beta sensitivity in mice with mutations in the Sno gene. Mol Cell Biol, 23, 5446-5459. Pelzer, T., Lyons, G.E., Kim, S. and Moreadith, R.W. (1996) Cloning and characterization of the murine homolog of the sno proto-oncogene reveals a novel splice variant. Dev Dyn, 205, 114-125. Pfau, R., Tzatsos, A., Kampranis, S.C., Serebrennikova, O.B., Bear, S.E. and Tsichlis, P.N. (2008) Members of a family of JmjC domain-containing oncoproteins immortalize embryonic fibroblasts via a JmjC domain-dependent process. Proc Natl Acad Sci U S A, 105, 1907-1912. Pot, I. and Bonni, S. (2008) SnoN in TGF-beta signaling and cancer biology. Curr Mol Med, 8, 319-328. Ramel, M.C., Emery, C.S., Foulger, R., Goberdhan, D.C., van den Heuvel, M. and Wilson, C. (2007) Drosophila SnoN modulates growth and patterning by antagonizing TGF-beta signalling. Mech Dev, 124, 304-317. Rao, P.E., Petrone, A.L. and Ponath, P.D. (2005) Differentiation and expansion of T cells with regulatory function from human peripheral lymphocytes by stimulation in the presence of TGF-{beta}. J Immunol, 174, 1446-1455. Rhee, S.G., Bae, Y.S., Lee, S.R. and Kwon, J. (2000) Hydrogen peroxide: a key messenger that modulates protein phosphorylation through cysteine oxidation. Sci STKE, 2000, pe1. Roberts, A.B., Anzano, M.A., Lamb, L.C., Smith, J.M., Frolik, C.A., Marquardt, H., Todaro, G.J. and Sporn, M.B. (1982) Isolation from murine sarcoma cells of novel transforming growth factors potentiated by EGF. Nature, 295, 417-419. Roberts, A.B., Anzano, M.A., Wakefield, L.M., Roche, N.S., Stern, D.F. and Sporn, M.B. (1985) Type beta transforming growth factor: a bifunctional regulator of cellular growth. Proc Natl Acad Sci U S A, 82, 119-123. Roberts, A.B., Thompson, N.L., Heine, U., Flanders, C. and Sporn, M.B. (1988) Transforming growth factor-beta: possible roles in carcinogenesis. Br J Cancer, 57, 594-600. Rodier, F., Campisi, J. and Bhaumik, D. (2007) Two faces of p53: ageing and tumor suppression. Nucleic Acids Res, 35, 7475-7484. Salomoni, P. and Pandolfi, P.P. (2002) The role of PML in tumor suppression. Cell, 108, 165-170. Schiemann, W.P., Pfeifer, W.M., Levi, E., Kadin, M.E. and Lodish, H.F. (1999) A deletion in the gene for transforming growth factor beta type I receptor abolishes growth regulation by transforming growth factor beta in a cutaneous T-cell lymphoma. Blood, 94, 2854-2861. Schutte, M., Hruban, R.H., Hedrick, L., Cho, K.R., Nadasdy, G.M., Weinstein, C.L., Bova, G.S., Isaacs, W.B., Cairns, P., Nawroz, H., Sidransky, D., Casero, R., Jr., Meltzer, P.S., Hahn, S.A. and Kern, S.E. (1996) DPC4 gene in various tumor types. Cancer Res, 56, 2527-2530. Seeler, J.S. and Dejean, A. (1999) The PML nuclear bodies: actors or extras? Curr Opin Genet Dev, 9, 362-367. Sehgal, I. and Thompson, T.C. (1999) Novel regulation of type IV collagenase (matrix metalloproteinase-9 and -2) activities by transforming growth factor-beta1 in human prostate cancer cell lines. Mol Biol Cell, 10, 407-416.

Page 123: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

110

Seoane, J., Le, H.V., Shen, L., Anderson, S.A. and Massague, J. (2004) Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell, 117, 211-223. 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. Sharpless, N.E. (2005) INK4a/ARF: a multifunctional tumor suppressor locus. Mutat Res, 576, 22-38. Sharpless, N.E., Bardeesy, N., Lee, K.H., Carrasco, D., Castrillon, D.H., Aguirre, A.J., Wu, E.A., Horner, J.W. and DePinho, R.A. (2001) Loss of p16Ink4a with retention of p19Arf predisposes mice to tumorigenesis. Nature, 413, 86-91. Shen, T.H., Lin, H.K., Scaglioni, P.P., Yung, T.M. and Pandolfi, P.P. (2006) The mechanisms of PML-nuclear body formation. Mol Cell, 24, 331-339. Sherr, C.J. and DePinho, R.A. (2000) Cellular senescence: mitotic clock or culture shock? Cell, 102, 407-410. Shi, Y. and Massague, J. (2003) Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell, 113, 685-700. Shimizu, S., Nishikawa, Y., Kuroda, K., Takagi, S., Kozaki, K., Hyuga, S., Saga, S. and Matsuyama, M. (1996) Involvement of transforming growth factor beta1 in autocrine enhancement of gelatinase B secretion by murine metastatic colon carcinoma cells. Cancer Res, 56, 3366-3370. Shinagawa, T., Dong, H.-D., Xu, M., Maekawa, T. and Ishii, S. (2000) The sno gene, which encodes a component of the histone deacetylation complex, acts as a tumor suppressor in mice. EMBO J., 19, 2280-2291. Shipley, G.D., Pittelkow, M.R., Wille, J.J., Jr., Scott, R.E. and Moses, H.L. (1986) Reversible inhibition of normal human prokeratinocyte proliferation by type beta transforming growth factor-growth inhibitor in serum-free medium. Cancer Res, 46, 2068-2071. Siegel, P.M. and Massague, J. (2003) Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer. Nat Rev Cancer, 3, 807-821. Siegel, P.M., Shu, W., Cardiff, R.D., Muller, W.J. and Massague, J. (2003) Transforming growth factor beta signaling impairs Neu-induced mammary tumorigenesis while promoting pulmonary metastasis. Proc Natl Acad Sci U S A, 100, 8430-8435. Souchelnytskyi, S., Tamaki, K., Engstrom, U., Wernstedt, C., ten Dijke, P. and Heldin, C.H. (1997) Phosphorylation of Ser465 and Ser467 in the C terminus of Smad2 mediates interaction with Smad4 and is required for transforming growth factor-beta signaling. J Biol Chem, 272, 28107-28115. Stavnezer, E., Barkas, A.E., Brennan, L.A., Brodeur, D. and Li, Y. (1986) Transforming Sloan-Kettering viruses generated from the cloned v-ski oncogene by in vitro and in vivo recombinations. J Virol, 57, 1073-1083. Stavnezer, E., Brodeur, D. and Brennan, L.A. (1989) The v-ski oncogene encodes a truncated set of c-ski coding exons with limited sequence and structural relatedness to v-myc. Mol Cell Biol, 9, 4038-4045. Stearns, M.E., Garcia, F.U., Fudge, K., Rhim, J. and Wang, M. (1999) Role of interleukin 10 and transforming growth factor beta1 in the angiogenesis and metastasis of human

Page 124: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

111

prostate primary tumor lines from orthotopic implants in severe combined immunodeficiency mice. Clin Cancer Res, 5, 711-720. Stegmuller, J., Konishi, Y., Huynh, M.A., Yuan, Z., Dibacco, S. and Bonni, A. (2006) Cell-intrinsic regulation of axonal morphogenesis by the Cdh1-APC target SnoN. Neuron, 50, 389-400. Stroschein, S.L., Bonni, S., Wrana, J.L. and Luo, K. (2001) Smad3 recruits the anaphase-promoting complex for ubiquitination and degradation of SnoN. Genes Dev, 15, 2822-2836. Stroschein, S.L., Wang, W., Zhou, S., Zhou, Q. and Luo, K. (1999) Negative feedback regulation of TGF-beta signaling by the SnoN oncoprotein. Science, 286, 771-774. Sun, Y., Liu, X., Eaton, E.N., Lane, W.S., Lodish, H.F. and Weinberg, R.A. (1999a) Interaction of the Ski oncoprotein with Smad3 regulates TGF-beta signaling. Mol Cell, 4, 499-509. Sun, Y., Liu, X., Ng-Eaton, E., Lodish, H.F. and Weinberg, R.A. (1999b) SnoN and Ski protooncoproteins are rapidly degraded in response to transforming growth factor beta signaling. Proc Natl Acad Sci U S A, 96, 12442-12447. Sutrave, P., Copeland, T.D., Showalter, S.D. and Hughes, S.H. (1990) Characterization of chicken c-ski oncogene products expressed by retrovirus vectors. Mol Cell Biol, 10, 3137-3144. Sutrave, P. and Hughes, S.H. (1989) Isolation and characterization of three distinct cDNAs for the chicken c-ski gene. Mol Cell Biol, 9, 4046-4051. Suzuki, T., Minehata, K., Akagi, K., Jenkins, N.A. and Copeland, N.G. (2006) Tumor suppressor gene identification using retroviral insertional mutagenesis in Blm-deficient mice. Embo J, 25, 3422-3431. Takahashi, Y., Lallemand-Breitenbach, V., Zhu, J. and de The, H. (2004) PML nuclear bodies and apoptosis. Oncogene, 23, 2819-2824. Tang, B., Bottinger, E.P., Jakowlew, S.B., Bagnall, K.M., Mariano, J., Anver, M.R., Letterio, J.J. and Wakefield, L.M. (1998) Transforming growth factor-beta1 is a new form of tumor suppressor with true haploid insufficiency. Nat Med, 4, 802-807. Thiery, J.P. (2002) Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer, 2, 442-454. Todaro, G.J. and Green, H. (1963) Quantitative studies of the growth of mouse embryo cells in culture and their development into established lines. J Cell Biol, 17, 299-313. Tsukazaki, T., Chiang, T.A., Davison, A.F., Attisano, L. and Wrana, J.L. (1998) SARA, a FYVE domain protein that recruits Smad2 to the TGFbeta receptor. Cell, 95, 779-791. Tucker, R.F., Shipley, G.D., Moses, H.L. and Holley, R.W. (1984) Growth inhibitor from BSC-1 cells closely related to platelet type beta transforming growth factor. Science, 226, 705-707. Ueki, N., Nakazato, M., Ohkawa, T., Ikeda, T., Amuro, Y., Hada, T. and Higashino, K. (1992) Excessive production of transforming growth-factor beta 1 can play an important role in the development of tumorigenesis by its action for angiogenesis: validity of neutralizing antibodies to block tumor growth. Biochim Biophys Acta, 1137, 189-196. Villanacci, V., Bellone, G., Battaglia, E., Rossi, E., Carbone, A., Prati, A., Verna, C., Niola, P., Morelli, A., Grassini, M. and Bassotti, G. (2008) Ski/SnoN expression in the sequence metaplasia-dysplasia-adenocarcinoma of Barrett's esophagus. Hum Pathol, 39, 403-409. Vousden, K.H. (2002) Activation of the p53 tumor suppressor protein. Biochim Biophys Acta, 1602, 47-59.

Page 125: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

112

Vousden, K.H. and Lu, X. (2002) Live or let die: the cell's response to p53. Nat Rev Cancer, 2, 594-604. Wadhwa, R., Sugihara, T., Taira, K. and Kaul, S.C. (2004) The ARF-p53 senescence pathway in mouse and human cells. Histol Histopathol, 19, 311-316. Wakefield, L.M. and Roberts, A.B. (2002) TGF-beta signaling: positive and negative effects on tumorigenesis. Curr Opin Genet Dev, 12, 22-29. Wan, Y., Liu, X. and Kirschner, M.W. (2001) The anaphase-promoting complex mediates TGF-beta signaling by targeting SnoN for destruction. Mol Cell, 8, 1027-1039. Watanabe, M., Masuyama, N., Fukuda, M. and Nishida, E. (2000) Regulation of intracellular dynamics of Smad4 by its leucine-rich nuclear export signal. EMBO Rep, 1, 176-182. Weinberg, R.A. (1994) Oncogenes and tumor suppressor genes. CA Cancer J Clin, 44, 160-170. Wesierska-Gadek, J., Wojciechowski, J., Ranftler, C. and Schmid, G. (2005) Role of p53 tumor suppressor in ageing: regulation of transient cell cycle arrest and terminal senescence. J Physiol Pharmacol, 56, 15-28. Wilkinson, D.S., Ogden, S.K., Stratton, S.A., Piechan, J.L., Nguyen, T.T., Smulian, G.A. and Barton, M.C. (2005) A direct intersection between p53 and transforming growth factor beta pathways targets chromatin modification and transcription repression of the alpha-fetoprotein gene. Mol Cell Biol, 25, 1200-1212. Wilkinson, D.S., Tsai, W.W., Schumacher, M.A. and Barton, M.C. (2008) Chromatin-bound p53 anchors activated Smads and the mSin3A corepressor to confer transforming growth factor beta-mediated transcription repression. Mol Cell Biol, 28, 1988-1998. Wrana, J.L., Attisano, L., Carcamo, J., Zentella, A., Doody, J., Laiho, M., Wang, X.F. and Massague, J. (1992) TGF beta signals through a heteromeric protein kinase receptor complex. Cell, 71, 1003-1014. Wrana, J.L., Attisano, L., Wieser, R., Ventura, F. and Massague, J. (1994) Mechanism of activation of the TGF-beta receptor. Nature, 370, 341-347. Wu, G., Chen, Y.G., Ozdamar, B., Gyuricza, C.A., Chong, P.A., Wrana, J.L., Massague, J. and Shi, Y. (2000) Structural basis of Smad2 recognition by the Smad anchor for receptor activation. Science, 287, 92-97. Wu, J.W., Krawitz, A.R., Chai, J., Li, W., Zhang, F., Luo, K. and Shi, Y. (2002) Structural mechanism of Smad4 recognition by the nuclear oncoprotein Ski: insights on Ski-mediated repression of TGF-beta signaling. Cell, 111, 357-367. Xiao, Z., Liu, X., Henis, Y.I. and Lodish, H.F. (2000a) A distinct nuclear localization signal in the N terminus of Smad 3 determines its ligand-induced nuclear translocation. Proc Natl Acad Sci U S A, 97, 7853-7858. Xiao, Z., Liu, X. and Lodish, H.F. (2000b) Importin beta mediates nuclear translocation of Smad 3. J Biol Chem, 275, 23425-23428. Xu, X., Brodie, S.G., Yang, X., Im, Y.H., Parks, W.T., Chen, L., Zhou, Y.X., Weinstein, M., Kim, S.J. and Deng, C.X. (2000) Haploid loss of the tumor suppressor Smad4/Dpc4 initiates gastric polyposis and cancer in mice. Oncogene, 19, 1868-1874. 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.

Page 126: SnoN is a stress transducer that is able to function as a tumor … · 2018-10-10 · SnoN is a stress transducer that is able to function as a tumor suppressor in vivo by interacting

113

Yamagiwa, S., Gray, J.D., Hashimoto, S. and Horwitz, D.A. (2001) A role for TGF-beta in the generation and expansion of CD4+CD25+ regulatory T cells from human peripheral blood. J Immunol, 166, 7282-7289. Yaswen, P. and Campisi, J. (2007) Oncogene-induced senescence pathways weave an intricate tapestry. Cell, 128, 233-234. Yin, J.J., Selander, K., Chirgwin, J.M., Dallas, M., Grubbs, B.G., Wieser, R., Massague, J., Mundy, G.R. and Guise, T.A. (1999) TGF-beta signaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases development. J Clin Invest, 103, 197-206. Zhang, F., Lundin, M., Ristimaki, A., Heikkila, P., Lundin, J., Isola, J., Joensuu, H. and Laiho, M. (2003) Ski-related novel protein N (SnoN), a negative controller of transforming growth factor-beta signaling, is a prognostic marker in estrogen receptor-positive breast carcinomas. Cancer Res, 63, 5005-5010. Zhang, X., Egawa, K., Xie, Y. and Ihn, H. (2009) The expression of SnoN in normal human skin and cutaneous keratinous neoplasms. Int J Dermatol, 48, 579-583. Zhang, Y., Feng, X., We, R. and Derynck, R. (1996) Receptor-associated Mad homologues synergize as effectors of the TGF-beta response. Nature, 383, 168-172. Zheng, G., Teumer, J., Colmenares, C., Richmond, C. and Stavnezer, E. (1997) Identification of a core functional and structural domain of the v-Ski oncoprotein responsible for both transformation and myogenesis. Oncogene, 15, 459-471. Zhu, Q., Krakowski, A.R., Dunham, E.E., Wang, L., Bandyopadhyay, A., Berdeaux, R., Martin, G.S., Sun, L. and Luo, K. (2006) Dual Role of SnoN in mammalian tumorigenesis. Mol Cell Biol. Zhu, Q., Pearson-White, S. and Luo, K. (2005) Requirement for the SnoN oncoprotein in transforming growth factor beta-induced oncogenic transformation of fibroblast cells. Mol Cell Biol, 25, 10731-10744.