The Interplay of Proteins Encoded by CDKN1A, CDKN1B, CDKN2A and TP53 in the Prognosis of Breast Cancer

Total Page:16

File Type:pdf, Size:1020Kb

The Interplay of Proteins Encoded by CDKN1A, CDKN1B, CDKN2A and TP53 in the Prognosis of Breast Cancer Research Article Clinics in Oncology Published: 02 Aug, 2019 The Interplay of Proteins Encoded by CDKN1A, CDKN1B, CDKN2A and TP53 in the Prognosis of Breast Cancer Valentina Robila1*, Bryce S Hatfield1, Nitai Mukhopadhyay2 and Michael O Idowu1 1Department of Pathology, VCU School of Medicine, USA 2Department of Biostatistics, VCU School of Medicine, USA Abstract The use of palbociclib has increased the spotlight on cell cycle regulators, especially on the proteins encoded by CDKN2A (p16), CDKN1A (p21), CDKN1B (p27), cyclin D1 and TP53 (p53). Cell cycle regulators and cyclin-dependent kinases show significant promise as new breast cancer biomarkers. Expression patterns of small sets of proteins in breast carcinoma were reported, and their overall role in breast cancer progression was investigated, with mixed results. The goal of our study was to simultaneously evaluate multiple cell cycle regulators in breast carcinoma. The protein expression was assessed on Tissue Microarrays (TMA) and the intensity and percentage of positive tumor cells were quantified. Statistical analysis was performed to establish patterns of protein associations, as well as correlation with prognostic factors, including metastases/recurrence and overall survival. Non-triple negative carcinoma showed significantly increased expression of cyclin D1, marginally increased expression of p21, 27, and decreased expression of p16, p53. This is in contrast to triple negative carcinoma which expressed high levels of p16 and p53. Statistically significant dual proteins associations were demonstrated, including direct correlations of expression of cyclin D1 with p21 or 27, and indirect correlations with p16 and p53. Cyclin D1 (p=0.0001) and p21 (p=0.0049) expression imparted a favorable outcome on development of metastases and/or survival, while p16 (p=0.0048) had a negative impact, particularly on non-triple negative carcinoma. The remainder of the tested proteins showed no prognostic significance. Given these proteins expression patterns, associations and prognostic impact, the results render cyclin D1, p21 and p16 as potential biomarkers for risk OPEN ACCESS stratification in breast carcinoma. *Correspondence: Keywords: Invasive breast carcinoma; Cyclin dependent kinases; p21; p16; Metastases Valentina Robila, Department of Pathology, VCU School of Medicine, Introduction 1200 E Marshall St. Gateway 6, PO Breast carcinoma is the most common cancer accounting for approximately 30% of new cancer Box 980662, Richmond, VA, USA, Tel: diagnoses and 14% of cancer deaths in women according to the 2018 statistics of the American 804-628-2999; Fax: 804-828-8733; Cancer Society [1]. While survival has improved with early diagnosis and advancement in E-mail: [email protected] treatment, breast cancer is still the second most common cause of death in women [1]. The recent Received Date: 03 Jul 2019 FDA approvals of palbociclib, ribociclib, and abemaciclib, selective inhibitors of Cyclin Dependent Accepted Date: 29 Jul 2019 Kinase 4 (CDK4) and CDK6, for treatment of hormone positive advanced breast cancer has brought Published Date: 02 Aug 2019 renewed focus on cell cycle regulators, especially CDK Inhibitors (CDKI). A critical function of Citation: CDKs is phosphorylation of Retinoblastoma protein (Rb) leading to progression from G1 to S phase Robila V, Hatfield BS, Mukhopadhyay of the cell cycle, while CDK inhibitors are believed to prevent this progression [2]. Conversely, N, Idowu MO. The Interplay of Proteins inactivation of CDKI leads to unopposed CDK4/6-Cyclin D1 complex formation and progression Encoded by CDKN1A, CDKN1B, from G1 to S phase of the cell cycle [2,3]. The CDK inhibitor proteins can be divided into 2 families: INK4A INKB CDKN2A and TP53 in the Prognosis 1) the INK4 family and 2) the CIP/KIP family. The INK4 family comprises of p16 , p15 , INKC INKD-4 of Breast Cancer. Clin Oncol. 2019; 4: p18 and p19 these bind to CDK4/6 and inhibit its kinase activity, leading to cell cycle CIP1 KIP1 KIP2 1644. arrest in G1 phase. The CIP/KIP family on the other hand consists of p21 , p27 and p57 ; these bind to a broader spectrum of cyclin/CDK complexed already formed (including cyclin D/ Copyright © 2019 Valentina Robila. CDK4/6 complex) and are therefore able to inhibit cell cycle progression [4]. p21 was shown to This is an open access article allow for repair of damaged DNA, [5] while p27, expressed in high concentrations in quiescent cells, distributed under the Creative decreases during entry into the cell cycle and promotes tumorigenesis [6,7]. Commons Attribution License, which permits unrestricted use, distribution, In Estrogen Receptor (ER) positive breast cancer, the prognostic significance of cyclin D1 and reproduction in any medium, over expression is still unclear. Some studies show cyclin D1 expression is associated with a poor prognosis and anti estrogen resistance, while others claim it is associated with improved outcomes provided the original work is properly [8-12]. Among CDKIs, cyclin-dependent kinase inhibitor 2A (CDKN2A) p16 expression was cited. Remedy Publications LLC., | http://clinicsinoncology.com/ 1 2019 | Volume 4 | Article 1644 Valentina Robila, et al., Clinics in Oncology - Breast Cancer shown to play a significant role in advanced tumor stage, higher Table 1: Patients demographics. grade, increased tumor proliferation, hormone receptor negativity, Characteristic Number patients (%) metastasis, and decreased overall survival [4,13-17]. Age <50 61 (28.1) Similarly, there is strong evidence correlating the reduced or >50 156 (71.8) absent p21 expression and progression of colorectal carcinoma, Ethnicity White 101 (46.5) Hodgkin lymphoma, and breast carcinoma, which tends to be African-American 107 (49.3) expressed more often in HER2 negative than in HER2 positive tumors Other 10 (4.6) [18-22]. A p27 decreased expression has been seen in the transition Body Mass Index (BMI) <35 171 (78.8) from ductal carcinoma in situ to invasive ductal carcinoma [6]. In >=35 46 (21.1) addition, a study of African American women with breast cancer Histologic type showed a significant decrease in p27 expression coupled with c-Myc Ductal 186 (85.7) Lobular 19 (8.7) over expression and decreased overall survival in patients with ER Other 12 (5.5) negative, PR negative, triple negative, and grade 3 breast cancers by Histologic grade Immunohistochemistry (IHC) [23]. I 28 (12.9) II 65 (29.9) The emerging role and early successes of CDK4/6 inhibitors to III 95 (43.7) n/a 29 (13.3) treat advanced breast cancer has highlighted the importance of finding Hormonal status specific biomarkers that could guide patient selection and treatment. Luminal A 135 (62.2) Additionally, these early successes have led to growing interest in Luminal B 10 (4.6) applying these drugs to early stage breast cancer [24]. Given that HER2 13 (5.9) Triple negative 52 (23.9) inhibitors of CDK4/CDK6 affect the progression from G1/S or G2/M Unknown 7 (3.2) phase of the cell cycle, it stands to reason to expect the expressions Pathologic tumor stage of cell cycle protein regulators like p16, p21 and p27 to achieve and T1 83 (38.2) T2 107 (49.3) maintain tumor cells growth arrest. In the present study, we sought a T3 23 (10.5) comprehensive approach and simultaneously evaluated multiple cell T4 4 (1.8) cycle regulator proteins, including p16, p21, p27, cyclin D1 and p53, Pathologic nodal stage N0 83 (38.2) in invasive breast carcinoma. Their immunohistochemical expression N1 61 (28.1) and correlation with clinicopathologic features such as distant N2 31 (14.2) metastasis or survival were investigated. N3 18 (8.2) Unknown 24 (11) Materials and Methods Recurrence None 40 (18.4) Patient demographics Loco-regional 137 (63.1) Distant 61 (28.1) The study analyzed invasive breast carcinoma from patients Unknown 26 (11.9) diagnosed and treated at our institution between 2005 and 2013. Survival Consult cases and cases reviewed for second opinions were excluded. Alive free/ unknown disease 60 (27.6) Alive with disease 87 (40) Only cases with adequate follow-up information were included. Died of disease 70 (32.2) Clinical-pathologic and demographic information including distant metastasis and survival, were collected via patient electronic medical was inferred using single covariate logistic regression. As the records and the Cancer Registry. expressions of several biomarkers are often strongly correlated, models with multiple covariates were avoided. A type I error threshold Immunohistochemistry and scoring of 5% was used to infer statistical significance. As reviewers assigned Tissue microarrays were constructed (1.0 mm cores) using the A score and H score, Cohen’s kappa measure was calculated Beecher ATA27 Tissue microarrayer. Each case has triplicate 1 mm for each marker and in most cases, were statistically significantly cores for the TMA. Immunohistochemistry for Cyclin D1 (clone different from 0. Thus only the average score was used in all analysis. EP12, Dako), p16 (clone INK4a, Roche), p21/WAF1 (clone DCS- Only statistical significant association with both Allred and H score 60.2, Cell Marque,) and p27/KiP1 (clone SX53G8, Cell Marque), and scoring system are considered significant. Significant association p53 (clone DO-7, Dako) were performed. with only one scoring system is not considered medically significant. Two board certified pathologists independently evaluated the Association of time to event outcomes, such as overall survival, was immunohistochemically stained slides, for percentage of positive assessed using a Cox proportional hazards regression model. cells and intensity of staining, using scales of 0% to 100% and 0-3, Results respectively. The percentage and intensity scores were converted to Allred score and H score. Allred scoring system uses the sum total Characteristics of the study population of proportion score and intensity score (range 0-8), while H score All 217 patients included in the study were women, age range 27 uses the multiplication of percentage reactivity with intensity score to 92 years old, diagnosed with invasive breast carcinoma, ductal and (range 0-300) [25,26].
Recommended publications
  • Cyclin D1/Cyclin-Dependent Kinase 4 Interacts with Filamin a and Affects the Migration and Invasion Potential of Breast Cancer Cells
    Published OnlineFirst February 28, 2010; DOI: 10.1158/0008-5472.CAN-08-1108 Tumor and Stem Cell Biology Cancer Research Cyclin D1/Cyclin-Dependent Kinase 4 Interacts with Filamin A and Affects the Migration and Invasion Potential of Breast Cancer Cells Zhijiu Zhong, Wen-Shuz Yeow, Chunhua Zou, Richard Wassell, Chenguang Wang, Richard G. Pestell, Judy N. Quong, and Andrew A. Quong Abstract Cyclin D1 belongs to a family of proteins that regulate progression through the G1-S phase of the cell cycle by binding to cyclin-dependent kinase (cdk)-4 to phosphorylate the retinoblastoma protein and release E2F transcription factors for progression through cell cycle. Several cancers, including breast, colon, and prostate, overexpress the cyclin D1 gene. However, the correlation of cyclin D1 overexpression with E2F target gene regulation or of cdk-dependent cyclin D1 activity with tumor development has not been identified. This suggests that the role of cyclin D1 in oncogenesis may be independent of its function as a cell cycle regulator. One such function is the role of cyclin D1 in cell adhesion and motility. Filamin A (FLNa), a member of the actin-binding filamin protein family, regulates signaling events involved in cell motility and invasion. FLNa has also been associated with a variety of cancers including lung cancer, prostate cancer, melanoma, human bladder cancer, and neuroblastoma. We hypothesized that elevated cyclin D1 facilitates motility in the invasive MDA-MB-231 breast cancer cell line. We show that MDA-MB-231 motility is affected by disturbing cyclin D1 levels or cyclin D1-cdk4/6 kinase activity.
    [Show full text]
  • Cyclin-Dependent Kinase 5 Decreases in Gastric Cancer and Its
    Published OnlineFirst January 21, 2015; DOI: 10.1158/1078-0432.CCR-14-1950 Biology of Human Tumors Clinical Cancer Research Cyclin-Dependent Kinase 5 Decreases in Gastric Cancer and Its Nuclear Accumulation Suppresses Gastric Tumorigenesis Longlong Cao1,2, Jiechao Zhou2, Junrong Zhang1,2, Sijin Wu3, Xintao Yang1,2, Xin Zhao2, Huifang Li2, Ming Luo1, Qian Yu1, Guangtan Lin1, Huizhong Lin1, Jianwei Xie1, Ping Li1, Xiaoqing Hu3, Chaohui Zheng1, Guojun Bu2, Yun-wu Zhang2,4, Huaxi Xu2,4,5, Yongliang Yang3, Changming Huang1, and Jie Zhang2,4 Abstract Purpose: As a cyclin-independent atypical CDK, the role of correlated with the severity of gastric cancer based on tumor CDK5 in regulating cell proliferation in gastric cancer remains and lymph node metastasis and patient 5-year fatality rate. unknown. Nuclear localization of CDK5 was found to be significantly Experimental Design: Expression of CDK5 in gastric tumor decreased in tumor tissues and gastric cancer cell lines, and paired adjacent noncancerous tissues from 437 patients was whereas exogenously expression of nucleus-targeted CDK5 measured by Western blotting, immunohistochemistry, and real- inhibited the proliferation and xenograft implantation of time PCR. The subcellular translocation of CDK5 was monitored gastric cancer cells. Treatment with the small molecule during gastric cancer cell proliferation. The role of nuclear CDK5 NS-0011, which increases CDK5 accumulation in the nucleus, in gastric cancer tumorigenic proliferation and ex vivo xenografts suppressed both cancer cell proliferation and xenograft was explored. Furthermore, by screening for compounds in the tumorigenesis. PubChem database that disrupt CDK5 association with its nu- Conclusions: Our results suggest that low CDK5 expression is clear export facilitator, we identified a small molecular (NS-0011) associated with poor overall survival in patients with gastric that inhibits gastric cancer cell growth.
    [Show full text]
  • Molecular Mechanisms of Colon Cancer Progression and Metastasis: Recent Insights and Advancements
    International Journal of Molecular Sciences Review Molecular Mechanisms of Colon Cancer Progression and Metastasis: Recent Insights and Advancements Ahmed Malki 1,*, Rasha Abu ElRuz 1, Ishita Gupta 2,3,4, Asma Allouch 1 , Semir Vranic 2,3 and Ala-Eddin Al Moustafa 2,3,4,* 1 Department of Biomedical Science, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar; [email protected] (R.A.E.); [email protected] (A.A.) 2 College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; [email protected] (I.G.); [email protected] (S.V.) 3 Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha 2713, Qatar 4 Biomedical Research Center, QU Health, Qatar University, Doha 2713, Qatar * Correspondence: [email protected] (A.M.); [email protected] (A.-E.A.M.); Tel.: +974-4403-6557 (A.M.); +974-3097-7440 (A.-E.A.M.) Abstract: Colorectal cancer (CRC), the third most common type of cancer, is the second leading cause of cancer-related mortality rates worldwide. Although modern research was able to shed light on the pathogenesis of CRC and provide enhanced screening strategies, the prevalence of CRC is still on the rise. Studies showed several cellular signaling pathways dysregulated in CRC, leading to the onset of malignant phenotypes. Therefore, analyzing signaling pathways involved in CRC metastasis is necessary to elucidate the underlying mechanism of CRC progression and pharmacotherapy. This review focused on target genes as well as various cellular signaling pathways including Wnt/β-catenin, p53, TGF-β/SMAD, NF-κB, Notch, VEGF, and JAKs/STAT3, which are associated with CRC progression and metastasis.
    [Show full text]
  • 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.
    [Show full text]
  • 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.
    [Show full text]
  • Role for Cyclin D1 in UVC-Induced and P53-Mediated Apoptosis
    Cell Death and Differentiation (1999) 6, 565 ± 569 ã 1999 Stockton Press All rights reserved 13509047/99 $12.00 http://www.stockton-press.co.uk/cdd Role for cyclin D1 in UVC-induced and p53-mediated apoptosis Hirofumi Hiyama1 and Steven A. Reeves*,1 irradiation of human fibroblasts is mediated by the p53- induced cyclin/cdk inhibitor, p21.3 Cell cycle progression 1 Molecular Neuro-Oncology, Neuroscience Center, Neurosurgical Services, through the G1/S boundary is controlled by G1 cyclins, Massachusetts General Hospital and Harvard Medical School, Boston, including D and E type cyclins and their cyclin-dependent Massachusetts 02129, USA kinases (cdk), whose phosphorylation of the retinoblastoma * corresponding author: Steven A. Reeves, Molecular Neuro-Oncology, gene product (pRB) causes a dissociation of the pRB and Neuroscience Center, Massachusetts General Hospital, 149 13th Street, Charlestown, MA 02129, USA. tel.: (617) 726-5510; fax: (617) 726-5079; E2F-1 interaction, and subsequent activation of E2F- e-mail: [email protected] mediated transcription. As an universal inhibitor of cdks, p21 can inhibit phosphorylation of pRB and allow for G1 arrest.4 Recent studies have shown that overexpression of cyclin Received 13.10.98; revised 19.03.99; accepted 23.03.99 D1 in serum-starved cell types can induce apoptosis.5 Edited by T. Cotter Interestingly, the induction of the cell death program was found to be associated with an increase in cyclin D1- dependent kinase activity.6 Moreover, in cells that contain Abstract wild-type p53, the overexpression of E2F-1 leads to S- 7 DNA damaging agents such as ultraviolet (UV) induce cell phase entry and p53-dependent apoptosis.
    [Show full text]
  • Mitosis Vs. Meiosis
    Mitosis vs. Meiosis In order for organisms to continue growing and/or replace cells that are dead or beyond repair, cells must replicate, or make identical copies of themselves. In order to do this and maintain the proper number of chromosomes, the cells of eukaryotes must undergo mitosis to divide up their DNA. The dividing of the DNA ensures that both the “old” cell (parent cell) and the “new” cells (daughter cells) have the same genetic makeup and both will be diploid, or containing the same number of chromosomes as the parent cell. For reproduction of an organism to occur, the original parent cell will undergo Meiosis to create 4 new daughter cells with a slightly different genetic makeup in order to ensure genetic diversity when fertilization occurs. The four daughter cells will be haploid, or containing half the number of chromosomes as the parent cell. The difference between the two processes is that mitosis occurs in non-reproductive cells, or somatic cells, and meiosis occurs in the cells that participate in sexual reproduction, or germ cells. The Somatic Cell Cycle (Mitosis) The somatic cell cycle consists of 3 phases: interphase, m phase, and cytokinesis. 1. Interphase: Interphase is considered the non-dividing phase of the cell cycle. It is not a part of the actual process of mitosis, but it readies the cell for mitosis. It is made up of 3 sub-phases: • G1 Phase: In G1, the cell is growing. In most organisms, the majority of the cell’s life span is spent in G1. • S Phase: In each human somatic cell, there are 23 pairs of chromosomes; one chromosome comes from the mother and one comes from the father.
    [Show full text]
  • 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.
    [Show full text]
  • 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.
    [Show full text]
  • Cyclin D1 Amplification Is Independent of P16 Inactivation in Head And
    Oncogene (1999) 18, 3541 ± 3545 ã 1999 Stockton Press All rights reserved 0950 ± 9232/99 $12.00 http://www.stockton-press.co.uk/onc Cyclin D1 ampli®cation is independent of p16 inactivation in head and neck squamous cell carcinoma K Okami1,3, AL Reed1, P Cairns1, WM Koch1, WH Westra2, S Wehage1, J Jen1 and D Sidransky*,1 1Department of OtolaryngologyÐHead & Neck Surgery, Division of Head and Neck Cancer Research, Johns Hopkins University School of Medicine, 818 Ross Research Building, 720 Rutland Avenue, Baltimore, Maryland, MD 21205-2196, USA; 2Department of Pathology, 7181 Meyer Building, Johns Hopkins Hospital, 600 North Wolfe Street, Baltimore, Maryland, MD 21287, USA; 3Department of Otolaryngology, Yamaguchi University, Ube 755, Japan Progression through the G1 phase of the cell cycle is kinase (cdk) 4 complexes. This cdk4 phosphorylation mediated by phosphorylation of the retinoblastoma activity is negatively regulated by the tumor suppressor protein (pRb) resulting in the release of essential gene p16 (a CDK inhibitor), and positively by cyclin transcription factors such as E2F-1. The phosphorylation D1. Each individual component of this p16-cyclin D1- of pRb is regulated positively by cyclin D1/CDK4 and Rb pathway is commonly targeted in various malig- negatively by CDK inhibitors, such as p16 (CDKN2/ nancies (Weinberg, 1995; Sherr, 1996). We previously MTS-1/INK4A). The p16 /cyclin D1/Rb pathway plays reported a high frequency (83%) of p16 inactivation in a critical role in tumorigenesis and many tumor types human HNSCC (Reed et al., 1996) and less frequent display a high frequency of inactivation of at least one inactivation (13%) of pRb (Yoo et al., 1994).
    [Show full text]
  • 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.
    [Show full text]
  • 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.
    [Show full text]