Intrinsic Cooperation Between P16 and P21 in the Onset of Cellular

Total Page:16

File Type:pdf, Size:1020Kb

Load more

Published OnlineFirst November 9, 2010; DOI: 10.1158/0008-5472.CAN-10-0801 Published OnlineFirst on November 9, 2010 as 10.1158/0008-5472.CAN-10-0801 Tumor and Stem Cell Biology Cancer Research Intrinsic Cooperation between p16INK4a and p21Waf1/Cip1 in the Onset of Cellular Senescence and Tumor Suppression In vivo Shinji Takeuchi1,4,6, Akiko Takahashi1, Noriko Motoi2, Shin Yoshimoto1, Tomoko Tajima1,3, Kimi Yamakoshi1, Atsushi Hirao5, Shigeru Yanagi3, Kiyoko Fukami3, Yuichi Ishikawa2, Saburo Sone6, Eiji Hara1, and Naoko Ohtani1 Abstract Although the p16INK4a and p21Waf1/Cip1 cyclin-dependent kinase (CDK) inhibitors are known to play key roles in cellular senescence in vitro, their roles in senescence remain rather poorly understood in vivo. This situation is partly due to the possibility of compensatory effect(s) between p16INK4a and p21Waf1/Cip1 or to the upregulation of functionally related CDK inhibitors. To directly address the cooperative roles of p16INK4a and INK4a Waf1/Cip1 p21Waf1/Cip1 in senescence in vivo, we generated a mouse line simply lacking both p16 and p21 genes [double-knockout (DKO)]. Mouse embryonic fibroblasts (MEF) derived from DKO mice displayed no evidence of cellular senescence when cultured serially in vitro. Moreover, DKO MEFs readily escaped Ras- induced senescence and overrode contact inhibition in culture. This was not the case in MEFs lacking either INK4a Waf1/Cip1 p16 or p21 , indicating that p16INK4a and p21Waf1/Cip1 play cooperative roles in cellular senescence and contact inhibition in vitro. Notably, we found the DKO mice to be extremely susceptible to 7,12-dimethylbenz (a)anthracene/12-O-tetradecanoylphorbol-13-acetate–induced skin carcinogenesis that involves oncogenic mutation of the H-ras gene. Mechanistic investigations suggested that the high incidence of cancer in DKO mice likely reflected a cooperative effect of increased benign skin tumor formation caused by p21Waf1/Cip1 loss, with increased malignant conversion of benign skin tumors caused by p16INK4a loss. Our findings establish an intrinsic cooperation between p16INK4a and p21Waf1/Cip1 in the onset of cellular senescence and tumor sup- pression in vivo. Cancer Res; 70(22); 9381–90. ©2010 AACR. Introduction that cellular senescence can be induced by a variety of po- tentially oncogenic stimuli, such as telomere shortening, The p16INK4a and p21Waf1/Cip1 cyclin-dependent kinase DNA damage, oxidative stress, or oncogene expression (CDK) inhibitors are known to play key roles in the onset (11–14), suggesting that cellular senescence is likely to act as of cellular senescence, a state of permanent cell cycle arrest a tumor suppression mechanism in vivo (15, 16). Although in culture (1–6). The simultaneous induction of p21Waf1/Cip1and the roles of p16Ink4a and p21Waf1/Cip1 in cellular senescence p16Ink4a expression cooperatively blocks the activation of both are well documented in various cell culture studies, the in vivo cyclin D kinase (CDK4/6) and cyclin E kinase (CDK2), allowing roles of these CDK inhibitors are poorly understood (17). For INK4a Waf1/Cip1 the accumulation of the dephosphorylated form of the retino- example, mice lacking p16 (18, 19) or p21 (20–22) blastoma tumor suppressor protein (pRb) and thereby causing exhibit only a little predisposition to spontaneous tumor for- permanent cell cycle arrest (7–10). It has become apparent mation. These observations raise the question of whether the results seen in cell culture studies truly reflect the physiologic in vivo Authors' Affiliations: Divisons of 1Cancer Biology and 2Pathology, The roles of these CDK inhibitors . However, it is also possi- Cancer Institute of Japanese Foundation for Cancer Research (JFCR); ble that these weak phenotypes could be due to functional 3School of Life Science, Tokyo University of Pharmacy and Life compensatory effect(s) between p16Ink4a and p21Waf1/Cip1. Sciences, Tokyo, Japan; Divisions of 4Medical Oncology and INK4a 5Molecular Genetics, Cancer Research Institute, Kanazawa University, To directly address the cooperative roles of p16 and Kanazawa,Japan;and6Institute of Health Biosciences, University of p21Waf1/Cip1 in vivo, we generated a compound mouse line INK4a Waf1/Cip1 Tokushima, Tokushima, Japan simply lacking both of the p16 and p21 genes Note: Supplementary data for this article are available at Cancer [double-knockout (DKO) mouse] on a C57BL/6 background, Research Online (http://cancerres.aacrjournals.org/). which is known to be carcinogenesis resistant (23–25), and Corresponding Author: Naoko Ohtani, Division of Cancer Biology, The Cancer Institute of Japanese Foundation for Cancer Research (JFCR), evaluated the roles of these two critical senescence inducers Tokyo 135-8550, Japan. Phone: 81-3-3570-0605; Fax: 81-3-3570- in vitro and in vivo. Intriguingly, DKO mice are significantly 0457; E-mail: [email protected]. more susceptible to chemical carcinogenesis compared with INK4a Waf1/Cip1 doi: 10.1158/0008-5472.CAN-10-0801 mice lacking either p16 or p21 alone. Moreover, ©2010 American Association for Cancer Research. mouse embryonic fibroblasts (MEF) derived from DKO mice www.aacrjournals.org 9381 Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst November 9, 2010; DOI: 10.1158/0008-5472.CAN-10-0801 Takeuchi et al. are resistant to the onset and the maintenance of cellular se- into 6-cm-diameter dishes. Cells were maintained for 15 days INK4a nescence in culture. These results indicate that p16 in a 3% O2/5% CO2 culture environment, and the medium plays a critical role in cooperating with p21Waf1/Cip1 in tumor was changed twice a week until the cells were photographed suppression in vivo. and counted. Senescence-associated β-galactosidase assay Materials and Methods Senescence-associated β-galactosidase assay was done as described previously (28). −/− −/− Generation of p16 ;p21 mice −/− p16 mice (C57BL/6) were kindly provided by N.E. Sharp- Analysis of intracellular reactive oxygen species less (University of North Carolina Lineberger Comprehensive To assess the generation of intracellular levels of reactive −/− Cancer Center, Chapel Hill, NC; ref. 19). p21 mice (C57BL/6) oxygen species (ROS), cells were incubated with 20 μmol/L were kindly provided by P. Leder (Harvard Medical School, 2′,7′-dichlorofluorescein diacetate (Calbiochem) for 20 min- −/− −/− Boston, MA; ref. 20). p16 ;p21 DKO mice were generated utes at 37°C. The peak excitation wavelength for oxidized −/− −/− by crossing p16 mice and p21 mice. All animals were 2′,7′-dichlorofluorescein was 488 nm and the emission wave- cared for by using protocols approved by the Committee for length was 525 nm as described previously (6). the Use and Care of Experimental Animals of the Japanese Foundation for Cancer Research. Chemically induced skin tumor formation The chemically induced skin tumor formation analysis was Cell culture done as described previously (29). Briefly, mice of the wild- MEFs were generated from E13.5 embryos of wild-type, type, p16-KO, p21-KO, or DKO genotypes in the resting phase −/− −/− −/− −/− p16 , p21 ,orp16 ;p21 DKO mice as previously de- of the hair cycle (8 weeks old) were shaved and treated with scribed (26). MEFs were grown in DMEM supplemented with 100 μg of 7,12-dimethylbenz(a)anthracene (DMBA) in 100 μL 10% fetal bovine serum in 3% O2/5% CO2 for 2 days, har- of acetone. One week after DMBA treatment, mice were sub- vested, viably frozen, and labeled as passage 0. Serial 3T3 cul- sequently treated twice a week with 12.5 μg of 12-O-tetrade- tivation was done as described (27). Cells were counted in canoylphorbol-13-acetate (TPA) in 100 μL of acetone for 20 triplicate. The number of cells present on the third day weeks. Tumors developed in mice were analyzed for histolo- (N3) was divided by the initial cell number (N0 = 3 × 105) gy at 30 weeks after DMBA treatment. and plotted as growth rate (N3/N0; Supplementary Fig. S1). The increase in the population doubling level (ΔPDL) was Histologic analysis calculated according to the following formula: ΔPDL = log Tissues were fixed in 10% buffered formalin, progressively (Nf/N0)/log 2, where N0 is the initial number of cells (3 × 105) dehydrated through gradients of alcohol, and embedded in and Nf is the final number of cells. paraffin. Samples were sectioned on a microtome at 4-μm thickness, deparaffinized in xylene, rehydrated, and then Western blot analysis stained with H&E for histologic analysis. Proteins (40 μg) were analyzed by Western blotting, as pre- viously described (6). The primary antibodies used were Results p16INK4a (11104 IBL), p21Waf1/Cip1 (sc-6246, Santa Cruz Bio- technology, Inc.), p15INK4b (sc-613, Santa Cruz Biotechnology), p16INK4a;p21Waf1/Cip1 DKO MEFs proliferate without any p53 (1C12, Cell Signaling), p19Arf [ab80 (Abcam) or sc-32748 induction of cellular senescence by serial passaging INK4a Waf1/Cip1 (Santa Cruz Biotechnology)], H-Ras (sc-29, Santa Cruz Compound mice lacking both p16 and p21 on a Biotechnology), pRb (554136, BD Pharmingen), β-actin C57BL/6 background (DKO mice) are fertile and born normally (AC-74, Sigma-Aldrich), and α-tublin (DM-1A, Sigma-Aldrich). in the expected Mendelian ratio. MEFs derived from wild-type INK4a Waf1/Cip1 Secondary antibodies were detected by enhanced chemilumi- mice, p16 knockout (p16-KO) mice, p21 knockout nescence (Amersham). (p21-KO) mice, and DKO mice were subjected to serial passage using the 3T3 protocol under standard culture condi- Retrovirus
Recommended publications
  • CDK-Independent and PCNA-Dependent Functions of P21 in DNA Replication

    CDK-Independent and PCNA-Dependent Functions of P21 in DNA Replication

    G C A T T A C G G C A T genes Review CDK-Independent and PCNA-Dependent Functions of p21 in DNA Replication Sabrina Florencia Mansilla , María Belén De La Vega y, Nicolás Luis Calzetta y, Sebastián Omar Siri y and Vanesa Gottifredi * Cell Cycle and Genomic Stability Laboratory, Fundación Instituto Leloir, IIBBA-CONICET, Av. Patricias Argentinas 435, Buenos Aires 1405, Argentina; [email protected] (S.F.M.); [email protected] (M.B.D.L.V.); [email protected] (N.L.C.); [email protected] (S.O.S.) * Correspondence: [email protected] These authors contributed equally to this work. y Received: 9 April 2020; Accepted: 15 May 2020; Published: 28 May 2020 Abstract: p21Waf/CIP1 is a small unstructured protein that binds and inactivates cyclin-dependent kinases (CDKs). To this end, p21 levels increase following the activation of the p53 tumor suppressor. CDK inhibition by p21 triggers cell-cycle arrest in the G1 and G2 phases of the cell cycle. In the absence of exogenous insults causing replication stress, only residual p21 levels are prevalent that are insufficient to inhibit CDKs. However, research from different laboratories has demonstrated that these residual p21 levels in the S phase control DNA replication speed and origin firing to preserve genomic stability. Such an S-phase function of p21 depends fully on its ability to displace partners from chromatin-bound proliferating cell nuclear antigen (PCNA). Vice versa, PCNA also regulates p21 by preventing its upregulation in the S phase, even in the context of robust p21 induction by γ irradiation.
  • The P16 (Cdkn2a/Ink4a) Tumor-Suppressor Gene in Head

    The P16 (Cdkn2a/Ink4a) Tumor-Suppressor Gene in Head

    The p16 (CDKN2a/INK4a) Tumor-Suppressor Gene in Head and Neck Squamous Cell Carcinoma: A Promoter Methylation and Protein Expression Study in 100 Cases Lingbao Ai, M.D., Krystal K. Stephenson, Wenhua Ling, M.D., Chunlai Zuo, M.D., Perkins Mukunyadzi, M.D., James Y. Suen, M.D., Ehab Hanna, M.D., Chun-Yang Fan, M.D., Ph.D. Departments of Pathology (LA, KKS, CZ, PM, CYF) and Otolaryngology-Head and Neck Surgery (CYF, JYS, EH), University of Arkansas for Medical Sciences; and School of Public Health (LA, WL), Sun-Yat Sen University, Guangzhou, China apparent loss of p16 protein expression appears to The p16 (CDKN2a/INK4a) gene is an important be an independent prognostic factor, although loss tumor-suppressor gene, involved in the p16/cyclin- of p16 protein may be used to predict overall pa- dependent kinase/retinoblastoma gene pathway of tient survival in early-stage head and neck squa- cell cycle control. The p16 protein is considered to mous cell carcinoma. be a negative regulator of the pathway. The gene encodes an inhibitor of cyclin-dependent kinases 4 KEY WORDS: Gene inactivation, Head and and 6, which regulate the phosphorylation of reti- neck squamous cell carcinoma, p16, Promoter noblastoma gene and the G1 to S phase transition of hypermethylation. the cell cycle. In the present study, p16 gene pro- Mod Pathol 2003;16(9):944–950 moter hypermethylation patterns and p16 protein expression were analyzed in 100 consecutive un- The development of head and neck squamous cell treated cases of primary head and neck squamous carcinoma is believed to be a multistep process, in cell carcinoma by methylation-specific PCR and im- which genetic and epigenetic events accumulate as munohistochemical staining.
  • P14ARF Inhibits Human Glioblastoma–Induced Angiogenesis by Upregulating the Expression of TIMP3

    P14ARF Inhibits Human Glioblastoma–Induced Angiogenesis by Upregulating the Expression of TIMP3

    P14ARF inhibits human glioblastoma–induced angiogenesis by upregulating the expression of TIMP3 Abdessamad Zerrouqi, … , Daniel J. Brat, Erwin G. Van Meir J Clin Invest. 2012;122(4):1283-1295. https://doi.org/10.1172/JCI38596. Research Article Oncology Malignant gliomas are the most common and the most lethal primary brain tumors in adults. Among malignant gliomas, 60%–80% show loss of P14ARF tumor suppressor activity due to somatic alterations of the INK4A/ARF genetic locus. The tumor suppressor activity of P14ARF is in part a result of its ability to prevent the degradation of P53 by binding to and sequestering HDM2. However, the subsequent finding of P14ARF loss in conjunction with TP53 gene loss in some tumors suggests the protein may have other P53-independent tumor suppressor functions. Here, we report what we believe to be a novel tumor suppressor function for P14ARF as an inhibitor of tumor-induced angiogenesis. We found that P14ARF mediates antiangiogenic effects by upregulating expression of tissue inhibitor of metalloproteinase–3 (TIMP3) in a P53-independent fashion. Mechanistically, this regulation occurred at the gene transcription level and was controlled by HDM2-SP1 interplay, where P14ARF relieved a dominant negative interaction of HDM2 with SP1. P14ARF-induced expression of TIMP3 inhibited endothelial cell migration and vessel formation in response to angiogenic stimuli produced by cancer cells. The discovery of this angiogenesis regulatory pathway may provide new insights into P53-independent P14ARF tumor-suppressive mechanisms that have implications for the development of novel therapies directed at tumors and other diseases characterized by vascular pathology. Find the latest version: https://jci.me/38596/pdf Research article P14ARF inhibits human glioblastoma–induced angiogenesis by upregulating the expression of TIMP3 Abdessamad Zerrouqi,1 Beata Pyrzynska,1,2 Maria Febbraio,3 Daniel J.
  • Involvement of the Cyclin-Dependent Kinase Inhibitor P16 (Ink4a) in Replicative Senescence of Normal Human Fibroblasts

    Involvement of the Cyclin-Dependent Kinase Inhibitor P16 (Ink4a) in Replicative Senescence of Normal Human Fibroblasts

    Proc. Natl. Acad. Sci. USA Vol. 93, pp. 13742–13747, November 1996 Biochemistry Involvement of the cyclin-dependent kinase inhibitor p16 (INK4a) in replicative senescence of normal human fibroblasts DAVID A. ALCORTA*†,YUE XIONG‡,DAWN PHELPS‡,GREG HANNON§,DAVID BEACH§, AND J. CARL BARRETT* *Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709; ‡Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599; and §Howard Hughes Medical Institute, Cold Spring Harbor Laboratories, Cold Spring Harbor, NY 11724 Communicated by Raymond L. Erickson, Harvard University, Cambridge, MA, September 19, 1996 (received for review on May 15, 1996) ABSTRACT Human diploid fibroblasts (HDFs) can be viewed in ref. 5). In senescent fibroblasts, CDK2 is catalytically grown in culture for a finite number of population doublings inactive and the protein down-regulated (7). CDK4 is also before they cease proliferation and enter a growth-arrest state reported to be down-regulated in senescent cells (8), while the termed replicative senescence. The retinoblastoma gene prod- status of CDK6 has not been previously addressed. The uct, Rb, expressed in these cells is hypophosphorylated. To activating cyclins for these CDKs, cyclins D1 and E, are present determine a possible mechanism by which senescent human in senescent cells at similar or elevated levels relative to early fibroblasts maintain a hypophosphorylated Rb, we examined passage cells (8). A role of the CDK inhibitors in senescence the expression levels and interaction of the Rb kinases, CDK4 was revealed by the isolation of a cDNA of a highly expressed and CDK6, and the cyclin-dependent kinase inhibitors p21 message in senescent cells that encoded the CDK inhibitor, p21 and p16 in senescent HDFs.
  • Transcriptional Regulation of the P16 Tumor Suppressor Gene

    Transcriptional Regulation of the P16 Tumor Suppressor Gene

    ANTICANCER RESEARCH 35: 4397-4402 (2015) Review Transcriptional Regulation of the p16 Tumor Suppressor Gene YOJIRO KOTAKE, MADOKA NAEMURA, CHIHIRO MURASAKI, YASUTOSHI INOUE and HARUNA OKAMOTO Department of Biological and Environmental Chemistry, Faculty of Humanity-Oriented Science and Engineering, Kinki University, Fukuoka, Japan Abstract. The p16 tumor suppressor gene encodes a specifically bind to and inhibit the activity of cyclin-CDK specific inhibitor of cyclin-dependent kinase (CDK) 4 and 6 complexes, thus preventing G1-to-S progression (4, 5). and is found altered in a wide range of human cancers. p16 Among these CKIs, p16 plays a pivotal role in the regulation plays a pivotal role in tumor suppressor networks through of cellular senescence through inhibition of CDK4/6 activity inducing cellular senescence that acts as a barrier to (6, 7). Cellular senescence acts as a barrier to oncogenic cellular transformation by oncogenic signals. p16 protein is transformation induced by oncogenic signals, such as relatively stable and its expression is primary regulated by activating RAS mutations, and is achieved by accumulation transcriptional control. Polycomb group (PcG) proteins of p16 (Figure 1) (8-10). The loss of p16 function is, associate with the p16 locus in a long non-coding RNA, therefore, thought to lead to carcinogenesis. Indeed, many ANRIL-dependent manner, leading to repression of p16 studies have shown that the p16 gene is frequently mutated transcription. YB1, a transcription factor, also represses the or silenced in various human cancers (11-14). p16 transcription through direct association with its Although many studies have led to a deeper understanding promoter region.
  • Clusterin-Mediated Apoptosis Is Regulated by Adenomatous Polyposis Coli and Is P21 Dependent but P53 Independent

    Clusterin-Mediated Apoptosis Is Regulated by Adenomatous Polyposis Coli and Is P21 Dependent but P53 Independent

    [CANCER RESEARCH 64, 7412–7419, October 15, 2004] Clusterin-Mediated Apoptosis Is Regulated by Adenomatous Polyposis Coli and Is p21 Dependent but p53 Independent Tingan Chen,1 Joel Turner,1 Susan McCarthy,1 Maurizio Scaltriti,2 Saverio Bettuzzi,2 and Timothy J. Yeatman1 1Department of Interdisciplinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida; and 2Dipartimento di Medicina Sperimentale, Sezione di Biochimica, Biochimica Clinica e Biochimica dell’Esercizio Fisico, Parma, Italy ABSTRACT ptosis (15). Additional data suggest that a secreted form of clusterin acts as a molecular chaperone, scavenging denatured proteins outside Clusterin is a widely expressed glycoprotein that has been paradoxi- cells following specific stress-induced injury such as heat shock. cally observed to have both pro- and antiapoptotic functions. Recent Other data, show that overexpression of a specific nuclear form of reports suggest this apparent dichotomy of function may be related to two different isoforms, one secreted and cytoplasmic, the other nuclear. To clusterin acts as a prodeath signal (16). Furthermore, studies of human clarify the functional role of clusterin in regulating apoptosis, we exam- colon cancer suggest a conversion from the nuclear form of clusterin ined its expression in human colon cancer tissues and in human colon to the cytoplasmic form, which may promote tumor progression (17). cancer cell lines. We additionally explored its expression and activity using Recently, a link between the accumulation of the nuclear form of models of adenomatous polyposis coli (APC)- and chemotherapy-induced clusterin and anoikis induction in prostate epithelial cells was shown apoptosis. Clusterin RNA and protein levels were decreased in colon (18).
  • Complete Deletion of Apc Results in Severe Polyposis in Mice

    Complete Deletion of Apc Results in Severe Polyposis in Mice

    Oncogene (2010) 29, 1857–1864 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 $32.00 www.nature.com/onc SHORT COMMUNICATION Complete deletion of Apc results in severe polyposis in mice AF Cheung1, AM Carter1, KK Kostova1, JF Woodruff1, D Crowley1,2, RT Bronson3, KM Haigis4 and T Jacks1,2 1Koch Institute and Department of Biology, MIT, Cambridge, MA, USA; 2Howard Hughes Medical Institute, MIT, Cambridge, MA, USA; 3Department of Pathology, Tufts University School of Medicine and Veterinary Medicine, Boston, MA, USA and 4Masschusetts General Hospital Cancer Center, Harvard Medical School Department of Pathology, Charlestown, MA, USA The adenomatous polyposis coli (APC) gene product is region of APC termed the mutation cluster region and mutated in the vast majority of human colorectal cancers. result in retained expression of an N-terminal fragment APC negatively regulates the WNT pathway by aiding in of the APC protein (Kinzler and Vogelstein, 1996). the degradation of b-catenin, which is the transcription Genotype–phenotype correlations involving germline factor activated downstream of WNT signaling. APC APC mutations suggest that different lengths and levels mutations result in b-catenin stabilization and constitutive of APC expression can influence the number of polyps WNT pathway activation, leading to aberrant cellular in the gut, the distribution of polyps and extra-colonic proliferation. APC mutations associated with colorectal manifestations of the disease (Soravia et al., 1998; cancer commonly fall in a region of the gene termed the Nieuwenhuis and Vasen, 2007). Specifically, patients mutation cluster region and result in expression of an that present clinically with attenuated FAP have N-terminal fragment of the APC protein.
  • The P16 Status of Tumor Cell Lines Identifies Small Molecule Inhibitors Specific for Cyclin-Dependent Kinase 41

    The P16 Status of Tumor Cell Lines Identifies Small Molecule Inhibitors Specific for Cyclin-Dependent Kinase 41

    Vol. 5, 4279–4286, December 1999 Clinical Cancer Research 4279 The p16 Status of Tumor Cell Lines Identifies Small Molecule Inhibitors Specific for Cyclin-dependent Kinase 41 Akihito Kubo,2 Kazuhiko Nakagawa,2, 3 CDK4 kinase inhibitors that may selectively induce growth Ravi K. Varma, Nicholas K. Conrad, inhibition of p16-altered tumors. Jin Quan Cheng, Wen-Ching Lee, INTRODUCTION Joseph R. Testa, Bruce E. Johnson, INK4A 4 The p16 gene (also known as CDKN2A) encodes p16 , Frederic J. Kaye, and Michael J. Kelley which inhibits the CDK45:cyclin D and CDK6:cyclin D com- Medicine Branch [A. K., K. N., N. K. C., F. J. K., B. E. J.] and plexes (1). These complexes mediate phosphorylation of the Rb Developmental Therapeutics Program [R. K. V.], National Cancer Institute, Bethesda, Maryland 20889; Department of Medical protein and allow cell cycle progression beyond the G1-S-phase Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania checkpoint (2). Alterations of p16 have been described in a wide 19111 [J. Q. C., W-C. L., J. R. T.]; and Department of Medicine, variety of histological types of human cancers including astro- Duke University Medical Center, Durham, North Carolina 27710 cytoma, melanoma, leukemia, breast cancer, head and neck [M. J. K.] squamous cell carcinoma, malignant mesothelioma, and lung cancer. Alterations of p16 can occur through homozygous de- ABSTRACT letion, point mutation, and transcriptional suppression associ- ated with hypermethylation in cancer cell lines and primary Loss of p16 functional activity leading to disruption of tumors (reviewed in Refs. 3–5). the p16/cyclin-dependent kinase (CDK) 4:cyclin D/retino- Whereas the Rb gene is inactivated in a narrow range of blastoma pathway is the most common event in human tumor cells, the pattern of mutational inactivation of Rb is tumorigenesis, suggesting that compounds with CDK4 ki- inversely correlated with p16 alterations (6–8), suggesting that nase inhibitory activity may be useful to regulate cancer cell a single defect in the p16/CDK4:cyclin D/Rb pathway is suffi- growth.
  • AP-1 in Cell Proliferation and Survival

    AP-1 in Cell Proliferation and Survival

    Oncogene (2001) 20, 2390 ± 2400 ã 2001 Nature Publishing Group All rights reserved 0950 ± 9232/01 $15.00 www.nature.com/onc AP-1 in cell proliferation and survival Eitan Shaulian1 and Michael Karin*,1 1Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, California, CA 92093-0636, USA A plethora of physiological and pathological stimuli extensively discussed previously (Angel and Karin, induce and activate a group of DNA binding proteins 1991; Karin, 1995). that form AP-1 dimers. These proteins include the Jun, The mammalian AP-1 proteins are homodimers and Fos and ATF subgroups of transcription factors. Recent heterodimers composed of basic region-leucine zipper studies using cells and mice de®cient in individual AP-1 (bZIP) proteins that belong to the Jun (c-Jun, JunB proteins have begun to shed light on their physiological and JunD), Fos (c-Fos, FosB, Fra-1 and Fra-2), Jun functions in the control of cell proliferation, neoplastic dimerization partners (JDP1 and JDP2) and the closely transformation and apoptosis. Above all such studies related activating transcription factors (ATF2, LRF1/ have identi®ed some of the target genes that mediate the ATF3 and B-ATF) subfamilies (reviewed by (Angel eects of AP-1 proteins on cell proliferation and death. and Karin, 1991; Aronheim et al., 1997; Karin et al., There is evidence that AP-1 proteins, mostly those that 1997; Liebermann et al., 1998; Wisdom, 1999). In belong to the Jun group, control cell life and death addition, some of the Maf proteins (v-Maf, c-Maf and through their ability to regulate the expression and Nrl) can heterodimerize with c-Jun or c-Fos (Nishiza- function of cell cycle regulators such as Cyclin D1, p53, wa et al., 1989; Swaroop et al., 1992), whereas other p21cip1/waf1, p19ARF and p16.
  • Loss of P21 Disrupts P14arf-Induced G1 Cell Cycle Arrest but Augments P14arf-Induced Apoptosis in Human Carcinoma Cells

    Loss of P21 Disrupts P14arf-Induced G1 Cell Cycle Arrest but Augments P14arf-Induced Apoptosis in Human Carcinoma Cells

    Oncogene (2005) 24, 4114–4128 & 2005 Nature Publishing Group All rights reserved 0950-9232/05 $30.00 www.nature.com/onc Loss of p21 disrupts p14ARF-induced G1 cell cycle arrest but augments p14ARF-induced apoptosis in human carcinoma cells Philipp G Hemmati1,3, Guillaume Normand1,3, Berlinda Verdoodt1, Clarissa von Haefen1, Anne Hasenja¨ ger1, DilekGu¨ ner1, Jana Wendt1, Bernd Do¨ rken1,2 and Peter T Daniel*,1,2 1Department of Hematology, Oncology and Tumor Immunology, University Medical Center Charite´, Campus Berlin-Buch, Berlin-Buch, Germany; 2Max-Delbru¨ck-Center for Molecular Medicine, Berlin-Buch, Germany The human INK4a locus encodes two structurally p16INK4a and p14ARF (termed p19ARF in the mouse), latter unrelated tumor suppressor proteins, p16INK4a and p14ARF of which is transcribed in an Alternative Reading Frame (p19ARF in the mouse), which are frequently inactivated in from a separate exon 1b (Duro et al., 1995; Mao et al., human cancer. Both the proapoptotic and cell cycle- 1995; Quelle et al., 1995; Stone et al., 1995). P14ARF is regulatory functions of p14ARF were initially proposed to usually expressed at low levels, but rapid upregulation be strictly dependent on a functional p53/mdm-2 tumor of p14ARF is triggered by various stimuli, that is, suppressor pathway. However, a number of recent reports the expression of cellular or viral oncogenes including have implicated p53-independent mechanisms in the E2F-1, E1A, c-myc, ras, and v-abl (de Stanchina et al., regulation of cell cycle arrest and apoptosis induction by 1998; Palmero et al., 1998; Radfar et al., 1998; Zindy p14ARF. Here, we show that the G1 cell cycle arrest et al., 1998).
  • Expression of P16 INK4A and P14 ARF in Hematological Malignancies

    Expression of P16 INK4A and P14 ARF in Hematological Malignancies

    Leukemia (1999) 13, 1760–1769 1999 Stockton Press All rights reserved 0887-6924/99 $15.00 http://www.stockton-press.co.uk/leu Expression of p16INK4A and p14ARF in hematological malignancies T Taniguchi1, N Chikatsu1, S Takahashi2, A Fujita3, K Uchimaru4, S Asano5, T Fujita1 and T Motokura1 1Fourth Department of Internal Medicine, University of Tokyo, School of Medicine; 2Division of Clinical Oncology, Cancer Chemotherapy Center, Cancer Institute Hospital; 3Department of Hematology, Showa General Hospital; 4Third Department of Internal Medicine, Teikyo University, School of Medicine; and 5Department of Hematology/Oncology, Institute of Medical Science, University of Tokyo, Tokyo, Japan The INK4A/ARF locus yields two tumor suppressors, p16INK4A tumor suppressor genes.10,11 In human hematological malig- ARF and p14 , and is frequently deleted in human tumors. We nancies, their inactivation occurs mainly by means of homo- studied their mRNA expressions in 41 hematopoietic cell lines and in 137 patients with hematological malignancies; we used zygous deletion or promoter region hypermethylation a quantitative reverse transcription-PCR assay. Normal periph- (reviewed in Ref. 12). In tumors such as pancreatic adenocar- eral bloods, bone marrow and lymph nodes expressed little or cinomas, esophageal squamous cell carcinomas and familial undetectable p16INK4A and p14ARF mRNAs, which were readily melanomas, p16INK4A is often inactivated by point mutation, detected in 12 and 17 of 41 cell lines, respectively. Patients with 12 INK4A which is not the case in hematological malignancies. On hematological malignancies frequently lacked p16 INK4C INK4D ARF the other hand, genetic aberrations of p18 or p19 expression (60/137) and lost p14 expression less frequently 12 (19/137, 13.9%).
  • Uncommon Association of Germline Mutations of RET Proto-Oncogene

    Uncommon Association of Germline Mutations of RET Proto-Oncogene

    European Journal of Endocrinology (2008) 158 417–422 ISSN 0804-4643 CASE REPORT Uncommon association of germline mutations of RET proto-oncogene and CDKN2A gene L Foppiani1, F Forzano2, I Ceccherini3, W Bruno4, P Ghiorzo4, F Caroli3, P Quilici5, R Bandelloni5, A Arlandini6, G Sartini6, M Cabria7 and P Del Monte1 1Endocrinology 2Genetics Laboratory, Galliera Hospital, 16128 Genova, Italy, 3Laboratory of Molecular Genetics, G Gaslini Institute, 16148 Genova, Italy, 4Department of Oncology, Biology and Genetics/Medical Genetics Service, University of Genova, 1632 Genova, Italy, 5Pathology Unit, 6Division of Surgery and 7Nuclear Medicine, Galliera Hospital, 16128 Genova, Italy (Correspondence should be addressed to L Foppiani who is now at S S D Endocrinologia, Mura delle Cappuccine 14, 16128 Genova, Italy; Email: [email protected]) Abstract Introduction: Calcitonin measurement is advised in the diagnosis of thyroid nodules, as it is an accurate marker of medullary thyroid carcinoma (MTC). C-cell hyperplasia (CCH)-induced hypercalcitoninemia cannot be distinguished from that induced by MTC, unless surgery is performed. Case: We report the clinical and biological features of a patient with a family history of cancer, including melanoma and pancreatic cancer, who had previously undergone surgery for melanoma. He presented the unusual association of papillary thyroid carcinoma (PTC), normocalcemic hyperpara- thyroidism, and hypercalcitoninemia with a pathological response to pentagastrin, which was histologically deemed secondary to CCH. Multiple endocrine neoplasia (MEN) 2A was diagnosed. RET gene analysis showed a p.V804M missense mutation in exon 14, a low- but variably penetrant defect found in both sporadic and MEN2A-associated MTC/CCH, and a p.G691S polymorphism in exon 11.