Human P14arf: an Exquisite Sensor of Morphological Changes and of Short-Lived Perturbations in Cell Cycle and in Nucleolar Function

Total Page:16

File Type:pdf, Size:1020Kb

Human P14arf: an Exquisite Sensor of Morphological Changes and of Short-Lived Perturbations in Cell Cycle and in Nucleolar Function Oncogene (2002) 21, 6779 – 6790 ª 2002 Nature Publishing Group All rights reserved 0950 – 9232/02 $25.00 www.nature.com/onc Human p14Arf: an exquisite sensor of morphological changes and of short-lived perturbations in cell cycle and in nucleolar function The´ re` se David-Pfeuty*,1 and Yolande Nouvian-Dooghe1 1UMR 146 du CNRS, Institut Curie-Recherche, Baˆtiment 110, Centre Universitaire, 91405 Orsay Ce´dex, France The human Ink4a/Arf tumor suppressor locus encodes integrity of cell cycle checkpoint controls (Hartwell and two distinct products: p16Ink4a which prevents phosphor- Weinert, 1989). Defects in checkpoint controls can ylation and inactivation of the retinoblastoma protein trigger either cell death or survival of cells carrying and, p14Arf, a nucleolar protein which activates the genetic anomalies and prone to develop genomic function of the tumor suppressor p53 protein in the instability which is the hallmark of cancer cells nucleoplasm in response to oncogenic stimulation through (Hartwell, 1992). It has been realized for some time an as yet ill-defined mechanism. Here we show that the that the p53 tumor suppressor gene is a component of level of endogenous p14Arf and its balance between the DNA damage checkpoints in mammalian cells since its nucleolus and the nucleoplasm in HeLa cells are function is required to prevent traverse across the G1/S exquisitely sensitive to changes in cell morphology and and G2/M boundaries in the presence of many types of to short-lived perturbations in cell cycle and in nucleolar genotoxic stress (O’Connor, 1997; Giaccia and Kastan, function such as those induced by the cyclin-dependent 1998) via promoting cell cycle arrest or apoptosis. It kinase inhibitor, roscovitine, and the casein kinase II and has been realized for even longer that p53 is mutated or RNA synthesis inhibitor, DRB. Most remarkably, lost in more than 50% of human cancers (Hollstein et whereas p14Arf predominantly concentrates in the al., 1994) which provides a hint that it would also serve nucleolus of interphase cells and transiently disappears in vivo to bar cell proliferation in inappropriate between metaphase and early G1 under normal growth environmental settings, a conclusion which has been conditions, it massively and reversibly accumulates in the consolidated by all kinds of in vivo and in vitro nucleoplasm of postmitotic and S-phase cells upon short- investigations (Ko and Prives, 1996; Levine, 1997; term treatment with roscovitine and, at a lesser extent, Agarwal et al., 1998). The possibility that the function DRB. In line with the fact that the nuclear level of p53 of p53 in tumor suppression and in DNA damage reaches a peak between mid-G1 and the G1/S border in response might represent two distinct facets of its p53-expressor cells which lack Arf expression, these activity has emerged recently from the discovery that results provide a clue that, in p53+/Arf+ cells, Arf oncogenic stimulation can induce its activation via a proteins might serve both to speed and to amplify p53- mechanism quite different from that generally used by mediated responses in conditions and cell cycle periods in DNA damages. Whereas the latter utilizes protein which the mechanisms involved in p53 stabilization and kinases to phosphorylate p53 at various sites mostly activation are not fully operational. They further suggest within its N-terminal transactivation domain and to that human endogenous p14Arf might activate p53 disrupt its interaction with its target and negative pathways in physiologic situations by acting inside the regulator, Mdm2 (Prives, 1998), the first relies on the nucleoplasm, especially when normal cell cycle progres- expression of the Arf proteins (Quelle et al., 1995; sion and nucleolar function are compromised. Della Valle et al., 1997), encoded by the late-identified Oncogene (2002) 21, 6779 – 6790. doi:10.1038/sj.onc. Arf (alternate reading frame) transcript of the Ink4a 1205871 gene (Quelle et al., 1995; Duro et al., 1995; Stone et al., 1995; Mao et al., 1995). Human p14Arf, like mouse Keywords: p14Arf; cell cycle regulation; tumor suppres- p19Arf, stabilizes p53 owing to its capability to interact sion; cell morphology; cyclin-dependent kinase directly with Mdm2 (Pomerantz et al., 1998; Zhang et al., 1998; Kamijo et al., 1998; Stott et al., 1998) and, thereby, to inhibit the Mdm2-mediated nuclear export Introduction (Tao and Levine, 1999; Zhang and Xiong, 1999), ubiquitination and degradation of p53 (Honda and Faithful cell reproduction depends on the orderly Yasuda, 1999). Arf upregulation in primary cells completion of S and M phase which relies on the exposed to hyperproliferative signals such as myc (Zindy et al., 1998), E1A (de Stanchina et al., 1998), E2F1 (Bates et al., 1998; Dimri et al., 2000), oncogenic ras (Palmero et al., 1998; Lin and Lowe, 2001) and v- *Correspondence: T David-Pfeuty; E-mail: The´ re` [email protected] abl (Radfar et al., 1998; Cong et al., 1999) seems to be Received 20 February 2002; revised 8 July 2002; accepted 15 July directly involved in stabilizing p53, the physiologic 2002 outcome being cell cycle arrest or apoptosis and, p14Arf subnuclear localization and function T David-Pfeuty and Y Nouvian-Dooghe 6780 possibly, after diverting the p53 pathway, cell trans- response because it would permit to accumulate at the formation (Sherr, 1998; Sharpless and DePinho, 1999). same time high levels of Mdm2 ready to eliminate p53 A rationale for these observations is provided by the as soon as the triggering signal is switched off. This fact that Arf is a direct target gene of some hypothesis that Arf might work at extending the transcription factors of the E2F family (Degregori et duration of reversible p53-mediated cell cycle arrest is al., 1997; Bates et al., 1998) whose function is required already consistent with the report that NARF2 cells to promote S phase entry (Harbour and Dean, 2000; arrested by inducible expression of p14Arf resume Nevins, 2000). Altogether, these data have led to the normal cycling upon removal of the inducer after 2 concept that, via Arf, p53 would also participate in days (Stott et al., 1998). Whether this hypothesis is also oncogene checkpoints that guard cells against hyper- relevant to physiologic situations clearly deserves proliferative signals (Sherr, 1998). further investigation. The fact that the co-expression Yet, the mechanism how Arf proteins activate p53 is of p14Arf and p53 does not occur in tumor cells still unclear because the subcellular compartment in suggests, not only, that it is incompatible with in vitro which p53 and Mdm2 localize preferentially is the tumor cell growth, but also, that some important nucleoplasm and for Arf, it is the nucleolus. Based on aspects of the in vivo Arf regulation are preserved in the results of enforced co-expression experiments, one p53-negative tumor cell lines. Identifying parameters model proposes that Arf proteins function owing to involved in controlling the level of expression and their ability to sequester Mdm2 in the nucleolus and, subnuclear targeting of endogenous p14Arf in this type thence, to trap Mdm2-free p53 inside the nucleus (Tao of tumor cells might therefore be a source of and Levine, 1999; Weber et al., 1999, 2000; Sherr and information not to ignore more than those issued from Weber, 2000) or in PML-like nuclear bodies (Zhang the experiments of ectopic overexpression. and Xiong, 1999). It can not be excluded however that Here we report that both the level of expression and Arf proteins could utilize other tricks to activate p53 subnuclear targeting of endogenous p14Arf are highly given that the nucleolar localization of p14Arf is not variable among clonal variants of the HeLa cell line requested for its function in human cell lines (Llanos et which differ in their morphological appearance. Most al., 2001) and that Mdm2 does not relocalize to the remarkably also, we uncovered that endogenous, but nucleolus following p19Arf-dependent, ras-induced cell not ectopically-expressed, p14Arf disappears in meta- cycle arrest in primary mouse keratinocytes (Lin and phase when the nucleoli disintegrate and re- Lowe, 2001). The major caveat of in vitro studies accumulates in the newly-reformed nucleoli of the aiming at deciphering the mechanism of p53 activation daughter cells at the beginning of G1. Intriguingly by Arf proteins in physiologic situations comes from however, it massively and reversibly accumulates in the the fact that they all rely on the artifice of ectopic nucleoplasm of early G1 and S-phase cells upon short- overexpression because Arf is undetectable in untrans- term treatments with roscovitine and, at a lesser extent, formed cells and its co-expression with p53 does not DRB, that disrupt nucleolar organization. The implica- occur in tumor-derived ones (Pomerantz et al., 1998; tions of these results in relation to the possible in vivo Stott et al., 1998; Bates et al., 1998; Lindstro¨ m et al., functions of the Arf proteins are discussed. 2000). That is why a more conceptual approach to the problem should also be considered. In a recent report Results (David-Pfeuty et al., 2001), we have shown that a short-term treatment of human breast carcinoma Clonal variability in levels and subnuclear localization of MCF-7 cells and non-immortalized IMR-90 fibroblasts endogenous p14Arf with the Cdk inhibitor, roscovitine (Meijer et al., 1997), and the CKII and RNA synthesis inhibitor, DRB Human p14Arf has been reported to localize predomi- (Granick, 1975; Haaf and Ward, 1996), induces a nantly to the nucleolus (Pomerantz et al., 1998; Kamijo reversible activation of p53 that is coupled with a et al., 1998; Stott et al., 1998; Zhang and Xiong, 1999), reversible nucleolar fragmentation and blockade of 45S more precisely to its granular component (Lindstro¨ m et pre-rRNA processing and a reversible brake in G1/S al., 2000; Karayan et al., 2001), in a pattern very transit.
Recommended publications
  • DNA Damage Checkpoint Dynamics Drive Cell Cycle Phase Transitions
    bioRxiv preprint doi: https://doi.org/10.1101/137307; this version posted August 4, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. DNA damage checkpoint dynamics drive cell cycle phase transitions Hui Xiao Chao1,2, Cere E. Poovey1, Ashley A. Privette1, Gavin D. Grant3,4, Hui Yan Chao1, Jeanette G. Cook3,4, and Jeremy E. Purvis1,2,4,† 1Department of Genetics 2Curriculum for Bioinformatics and Computational Biology 3Department of Biochemistry and Biophysics 4Lineberger Comprehensive Cancer Center University of North Carolina, Chapel Hill 120 Mason Farm Road Chapel Hill, NC 27599-7264 †Corresponding Author: Jeremy Purvis Genetic Medicine Building 5061, CB#7264 120 Mason Farm Road Chapel Hill, NC 27599-7264 [email protected] ABSTRACT DNA damage checkpoints are cellular mechanisms that protect the integrity of the genome during cell cycle progression. In response to genotoxic stress, these checkpoints halt cell cycle progression until the damage is repaired, allowing cells enough time to recover from damage before resuming normal proliferation. Here, we investigate the temporal dynamics of DNA damage checkpoints in individual proliferating cells by observing cell cycle phase transitions following acute DNA damage. We find that in gap phases (G1 and G2), DNA damage triggers an abrupt halt to cell cycle progression in which the duration of arrest correlates with the severity of damage. However, cells that have already progressed beyond a proposed “commitment point” within a given cell cycle phase readily transition to the next phase, revealing a relaxation of checkpoint stringency during later stages of certain cell cycle phases.
    [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]
  • Cyclin-Dependent Kinases and P53 Pathways Are Activated Independently and Mediate Bax Activation in Neurons After DNA Damage
    The Journal of Neuroscience, July 15, 2001, 21(14):5017–5026 Cyclin-Dependent Kinases and P53 Pathways Are Activated Independently and Mediate Bax Activation in Neurons after DNA Damage Erick J. Morris,1 Elizabeth Keramaris,2 Hardy J. Rideout,3 Ruth S. Slack,2 Nicholas J. Dyson,1 Leonidas Stefanis,3 and David S. Park2 1Massachusetts General Hospital Cancer Center, Laboratory of Molecular Oncology, Charlestown, Massachusetts 02129, 2Neuroscience Research Institute, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada, and 3Columbia University, New York, New York 10032 DNA damage has been implicated as one important initiator of ization, and DNA binding that result from DNA damage are not cell death in neuropathological conditions such as stroke. Ac- affected by the inhibition of CDK activity. Conversely, no de- cordingly, it is important to understand the signaling processes crease in retinoblastoma protein (pRb) phosphorylation was that control neuronal death induced by this stimulus. Previous observed in p53-deficient neurons that were treated with camp- evidence has shown that the death of embryonic cortical neu- tothecin. However, either p53 deficiency or the inhibition of rons treated with the DNA-damaging agent camptothecin is CDK activity alone inhibited Bax translocation, cytochrome c dependent on the tumor suppressor p53 and cyclin-dependent release, and caspase-3-like activation. Taken together, our re- kinase (CDK) activity and that the inhibition of either pathway sults indicate that p53 and CDK are activated independently alone leads to enhanced and prolonged survival. We presently and then act in concert to control Bax-mediated apoptosis. show that p53 and CDKs are activated independently on par- allel pathways.
    [Show full text]
  • The Cryoelectron Microscopy Structure of the Human CDK-Activating Kinase
    The cryoelectron microscopy structure of the human CDK-activating kinase Basil J. Grebera,b,1,2, Juan M. Perez-Bertoldic, Kif Limd, Anthony T. Iavaronee, Daniel B. Tosoa, and Eva Nogalesa,b,d,f,2 aCalifornia Institute for Quantitative Biosciences (QB3), University of California, Berkeley, CA 94720; bMolecular Biophysics and Integrative Bio-Imaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720; cBiophysics Graduate Group, University of California, Berkeley, CA 94720; dDepartment of Molecular and Cell Biology, University of California, Berkeley, CA 94720; eQB3/Chemistry Mass Spectrometry Facility, University of California, Berkeley, CA 94720; and fHoward Hughes Medical Institute, University of California, Berkeley, CA 94720 Edited by Seth A. Darst, Rockefeller University, New York, NY, and approved August 4, 2020 (received for review May 14, 2020) The human CDK-activating kinase (CAK), a complex composed of phosphoryl transfer (11). However, in addition to cyclin binding, cyclin-dependent kinase (CDK) 7, cyclin H, and MAT1, is a critical full activation of cell cycle CDKs requires phosphorylation of the regulator of transcription initiation and the cell cycle. It acts by T-loop (9, 12). In animal cells, these activating phosphorylations phosphorylating the C-terminal heptapeptide repeat domain of are carried out by CDK7 (13, 14), itself a cyclin-dependent ki- the RNA polymerase II (Pol II) subunit RPB1, which is an important nase whose activity depends on cyclin H (14). regulatory event in transcription initiation by Pol II, and it phos- In human and other metazoan cells, regulation of transcription phorylates the regulatory T-loop of CDKs that control cell cycle initiation by phosphorylation of the Pol II-CTD and phosphor- progression.
    [Show full text]
  • Synergistic Tumor Suppression by Combined Inhibition of Telomerase
    Synergistic tumor suppression by combined inhibition PNAS PLUS of telomerase and CDKN1A Romi Guptaa, Yuying Donga, Peter D. Solomona, Hiromi I. Wetterstenb, Christopher J. Chengc,d, JIn-Na Mina,e, Jeremy Hensonf,g, Shaillay Kumar Dograh, Sung H. Hwangi, Bruce D. Hammocki, Lihua J. Zhuj, Roger R. Reddelf,g, W. Mark Saltzmanc, Robert H. Weissb,k, Sandy Changa,e, Michael R. Greenl,1, and Narendra Wajapeyeea,1 Departments of aPathology and eLaboratory Medicine, Yale University School of Medicine, New Haven, CT 06510; iDepartment of Entomology and bDivision of Nephrology, Department of Internal Medicine, University of California, Davis, California 95616; Departments of cBiomedical Engineering and dMolecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511; fSydney Medical School, University of Sydney, NSW 2006, Australia; gCancer Research Unit, Children’s Medical Research Institute, Westmead, NSW 2145, Australia; hSingapore Institute of Clinical Sciences, Agency for Science Technology and Research (A*STAR), Brenner Center for Molecular Medicine, Singapore 117609; lHoward Hughes Medical Institute and jPrograms in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Massachusetts 01605; and kDepartment of Medicine, Mather VA Medical Center, Sacramento, CA 9565 Contributed by Michael R. Green, June 19, 2014 (sent for review June 8, 2014) Tumor suppressor p53 plays an important role in mediating growth dition to its role in cell cycle regulation, p21 has been shown in inhibition upon telomere dysfunction. Here, we show that loss of a variety of studies to repress apoptosis (9–13). the p53 target gene cyclin-dependent kinase inhibitor 1A (CDKN1A, Here,westudytheroleofp21inthe context of telomerase in- also known as p21WAF1/CIP1) increases apoptosis induction following hibition.
    [Show full text]
  • Full Text (PDF)
    ResearchArticle DNA Damage–Dependent Translocation of B23 and p19ARF Is Regulated by the Jun N-Terminal Kinase Pathway Orli Yogev,1 Keren Saadon,1 Shira Anzi,1 Kazushi Inoue,2 and Eitan Shaulian1 1Department of Experimental Medicine and Cancer Research, Hebrew University Medical School, Jerusalem, Israel and 2Departments of Pathology/Cancer Biology, Wake Forest University Health Sciences, Winston-Salem, North Carolina Abstract arrest or apoptosis. However, increased tumor development in The dynamic behavior of the nucleolus plays a role in the triple knockout mice nullizygous for ARF, p53, and Mdm2 showed detection of and response to DNA damage of cells. Two that ARF acts also in a p53-independent manner (11). Some of the nucleolar proteins, p14ARF/p19ARF and B23, were shown to p53-independent activity is attributed to its ability to reduce rRNA translocate out of the nucleolus after exposure of cells to DNA- processing (12, 13) and inhibit oncogene-induced transcription damaging agents. This translocation affects multiple cellular (14, 15). functions, such as DNA repair, proliferation, and survival. In ARF augments p53 activity mainly in response to oncogenic this study, we identify a pathway and scrutinize the mecha- stress. Its expression is up-regulated in response to deregulated nisms leading to the translocation of these proteins after oncogenic activity due to elevated transcription governed by several transcription factors, such as E2F (16), c-myc (17), AP-1 exposure of cells to DNA-damaging agents. We show that redistribution of B23 and p19ARF after the exposure to (18), or by oncogenic Ras (19). A second mechanism regulating ARF activity involves the control of its subcellular localization (3).
    [Show full text]
  • Metabolic Checkpoints in Cancer Cell Cycle
    City University of New York (CUNY) CUNY Academic Works All Dissertations, Theses, and Capstone Projects Dissertations, Theses, and Capstone Projects 2-2014 Metabolic Checkpoints in Cancer Cell Cycle Mahesh Saqcena Graduate Center, City University of New York How does access to this work benefit ou?y Let us know! More information about this work at: https://academicworks.cuny.edu/gc_etds/106 Discover additional works at: https://academicworks.cuny.edu This work is made publicly available by the City University of New York (CUNY). Contact: [email protected] METABOLIC CHECKPOINTS IN CANCER CELL CYCLE By Mahesh Saqcena A dissertation submitted to the Graduate Faculty in Biochemistry in partial fulfillment of the requirements for the degree of Doctor of Philosophy, The City University of New York 2014 i 2014 Mahesh Saqcena All Rights Reserved ii This manuscript has been read and accepted for the Graduate Faculty in Biochemistry in satisfaction of the dissertation requirement for the degree of Doctor of Philosophy. Date Dr. David A. Foster (Chair of Examining Committee) Date Dr. Edward J. Kennelly (Executive Officer) Dr. Mitchell Goldfarb (Hunter College) Dr. Paul Feinstein (Hunter College) Dr. Richard Kolesnick (Sloan Kettering Institute) Dr. Frederick R. Cross (Rockefeller University) (Supervisory Committee) THE CITY UNIVERSITY OF NEW YORK iii Abstract METABOLIC CHECKPOINTS IN CANCER CELL CYCLE by Mahesh Saqcena Advisor: Dr. David A. Foster Growth factors (GFs) as well as nutrient sufficiency regulate cell division in metazoans. The vast majority of mutations that contribute to cancer are in genes that regulate progression through the G1 phase of the cell cycle. A key regulatory site in G1 is the growth factor-dependent Restriction Point (R), where cells get permissive signals to divide.
    [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]
  • The Involvement of Ubiquitination Machinery in Cell Cycle Regulation and Cancer Progression
    International Journal of Molecular Sciences Review The Involvement of Ubiquitination Machinery in Cell Cycle Regulation and Cancer Progression Tingting Zou and Zhenghong Lin * School of Life Sciences, Chongqing University, Chongqing 401331, China; [email protected] * Correspondence: [email protected] Abstract: The cell cycle is a collection of events by which cellular components such as genetic materials and cytoplasmic components are accurately divided into two daughter cells. The cell cycle transition is primarily driven by the activation of cyclin-dependent kinases (CDKs), which activities are regulated by the ubiquitin-mediated proteolysis of key regulators such as cyclins, CDK inhibitors (CKIs), other kinases and phosphatases. Thus, the ubiquitin-proteasome system (UPS) plays a pivotal role in the regulation of the cell cycle progression via recognition, interaction, and ubiquitination or deubiquitination of key proteins. The illegitimate degradation of tumor suppressor or abnormally high accumulation of oncoproteins often results in deregulation of cell proliferation, genomic instability, and cancer occurrence. In this review, we demonstrate the diversity and complexity of the regulation of UPS machinery of the cell cycle. A profound understanding of the ubiquitination machinery will provide new insights into the regulation of the cell cycle transition, cancer treatment, and the development of anti-cancer drugs. Keywords: cell cycle regulation; CDKs; cyclins; CKIs; UPS; E3 ubiquitin ligases; Deubiquitinases (DUBs) Citation: Zou, T.; Lin, Z. The Involvement of Ubiquitination Machinery in Cell Cycle Regulation and Cancer Progression. 1. Introduction Int. J. Mol. Sci. 2021, 22, 5754. https://doi.org/10.3390/ijms22115754 The cell cycle is a ubiquitous, complex, and highly regulated process that is involved in the sequential events during which a cell duplicates its genetic materials, grows, and di- Academic Editors: Kwang-Hyun Bae vides into two daughter cells.
    [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]
  • A Haploid Genetic Screen Identifies the G1/S Regulatory Machinery As a Determinant of Wee1 Inhibitor Sensitivity
    A haploid genetic screen identifies the G1/S regulatory machinery as a determinant of Wee1 inhibitor sensitivity Anne Margriet Heijinka, Vincent A. Blomenb, Xavier Bisteauc, Fabian Degenera, Felipe Yu Matsushitaa, Philipp Kaldisc,d, Floris Foijere, and Marcel A. T. M. van Vugta,1 aDepartment of Medical Oncology, University Medical Center Groningen, University of Groningen, 9723 GZ Groningen, The Netherlands; bDivision of Biochemistry, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; cInstitute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos#3-09, Singapore 138673, Republic of Singapore; dDepartment of Biochemistry, National University of Singapore, Singapore 117597, Republic of Singapore; and eEuropean Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands Edited by Stephen J. Elledge, Harvard Medical School, Boston, MA, and approved October 21, 2015 (received for review March 17, 2015) The Wee1 cell cycle checkpoint kinase prevents premature mitotic Wee1 kinase at tyrosine (Tyr)-15 to prevent unscheduled Cdk1 entry by inhibiting cyclin-dependent kinases. Chemical inhibitors activity (5, 6). Conversely, timely activation of Cdk1 depends on of Wee1 are currently being tested clinically as targeted anticancer Tyr-15 dephosphorylation by one of the Cdc25 phosphatases drugs. Wee1 inhibition is thought to be preferentially cytotoxic in (7–10). When DNA is damaged, the downstream DNA damage p53-defective cancer cells. However, TP53 mutant cancers do not response (DDR) kinases Chk1 and Chk2 inhibit Cdc25 phos- respond consistently to Wee1 inhibitor treatment, indicating the phatases through direct phosphorylation, which blocks Cdk1 existence of genetic determinants of Wee1 inhibitor sensitivity other activation (11–13).
    [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]