Full Text (PDF)

Total Page:16

File Type:pdf, Size:1020Kb

Full Text (PDF) 654 Vol. 1, 654–664, July 2003 Molecular Cancer Research Expression of Cyclin-Dependent Kinase 6, but not Cyclin-Dependent Kinase 4, Alters Morphology of Cultured Mouse Astrocytes Karen K. Ericson, David Krull, Peter Slomiany, and Martha J. Grossel Department of Biology, Connecticut College, New London, CT Abstract mechanism involving the accumulation of the p53 and p130 Disruption of the pRb pathway is a common mechanism growth-suppressing proteins (6), and activation of cdk6 in tumor formation. The D-cyclin-associated kinases, precedes cdk4 activation in T cells (7, 8). Also, differences in cyclin-dependent kinase (cdk) 4 and cdk6, are important subcellular localization of cdk4 and cdk6 and in the timing of regulators of the G1-S phase transition and are elevated nuclear localization have been noted in several cell types by in several types of cancers, including gliomas. To several researchers (9–12). Data from tumor studies also investigate potential functional differences in these suggest differences in these kinases. Cdk4, and not cdk6, is kinases, mouse astrocytes were taken from chimeric specifically targeted in melanoma (13, 14) while cdk6 activity mice and propagated in tissue culture. These multipolar has been found to be elevated in squamous cell carcinomas (15, tissue-culture astrocytes were infected with viruses 16) and neuroblastomas (17) without alteration of cdk4 activity. expressing either cdk4 or cdk6. Interestingly, expression Cumulatively, these data suggest that cdk4 and cdk6 have of cdk6 resulted in a distinct and rapid morphology unique functions that may be cell-type specific, temporally change from multipolar to bipolar. This change was not regulated, or developmentally distinct. observed in control astrocytes or in astroyctes infected The process of differentiation can be considered as coupled with cdk4. Several other differences in cdk4- and cdk6- processes including: (a) exit from the cell cycle and (b) infected cells were noted, including differential binding expression of tissue-specific genes (18). Differences in protein to a subset of cell-cycle inhibitor proteins and a distinct levels and activity of key cell cycle inhibitors have been noted pattern of subcellular localization of these kinases. on differentiation of some cells. For instance, the cdk inhibitor Immunoblot and immunofluorescence analyses revealed protein, p27kip1, has been identified as part of a timing that cdk6-infected astrocytes had an altered expression mechanism that causes oligodendrocyte precursor cells to profile of known markers of glial differentiation. withdraw from the cell cycle and undergo differentiation. In Together, these data indicate several important differ- this system, p27kip1 accumulation correlates with differentiation CIP1/WAF-1 ences between cdk4 and cdk6 that highlight unique of oliodendrocytes (19–21). Expression of p21 , functional roles for these cyclin-dependent kinases. another cyclin kinase inhibitor protein, has been associated with differentiation in a variety of tissues (22–26). In murine Introduction erythroleukemia (MEL) cells, inhibitors that blocked both cdk2 and cdk6 induced differentiation, but inhibition of cdk2 and The G1 cyclins and associated cyclin-dependent kinases are cdk4 did not (24). Thus, G1 phase inhibitors have been shown critical regulators of G1 phase of the cell cycle. Cell cycle progression is dependent on mitogenic signals that activate to be important in the process of differentiation in muscle, cyclin D and its associated kinases. Cyclin-dependent kinase blood, and adipocytes (18). (cdk) 4 and cdk6 partner with cyclin D proteins to We set out to study functional differences of cdk4 and phosphorylate the Retinoblastoma protein (pRb), allowing cdk6 using a previously described tissue-culture astrocyte onset of S-phase (for review, see Ref. 1). Since the initial model system. This system was chosen because these cells characterization of cdk6, the D-cyclin kinases have been widely contain a minimal number of genetic mutations and are thought to act redundantly to regulate the cell cycle through the physiologically relevant, since overexpression of cdk4 and phosphorylation of pRB (2–5). However, recent studies have cdk6 has been found in glioblastomas and astrocytomas (27– uncovered novel functions of cdk6 and several studies highlight 30). These primary astrocytes were isolated from newborn differences between cdk4 and cdk6. For instance, cdk6 plays a transgenic mice that had been engineered to express the Avian role in halting inappropriate cellular proliferation through a Leukosis Virus (ALV) receptor (TVA) downstream of the GFAP promoter (G-tva) (31). Only GFAP-producing cells expressed the viral receptor and thus only these cells could be infected with ALV (31, 32). Starting cultures of these Received 1/13/03; revised 4/22/03; accepted 5/7/03. astrocytes displayed a star-shaped astrocytic morphology with The costs of publication of this article were defrayed in part by the payment of multiple short projections from a central cell body. Impor- page charges. This article must therefore be hereby marked advertisement in tantly, these cultures did not resemble a bipolar, fibroblast-like accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Grant support: NSF under CAREER grant #9984454 to Martha J. Grossel. morphology typical of glial restricted precursors or type 1 Requests for reprints: Martha J. Grossel, Department of Biology, Box 5331, astrocytes [for further descriptions of glial differentiation, see Connecticut College, 270 Mohegan Ave., New London, CT 06320. Phone: (860) 439-5209; Fax: (860) 439-2519. E-mail: [email protected] Lee et al. (33)]. Because cdk4 expression has been well Copyright D 2003 American Association for Cancer Research. characterized in this system (32), these tissue culture Downloaded from mcr.aacrjournals.org on October 1, 2021. © 2003 American Association for Cancer Research. Molecular Cancer Research 655 astrocytes provided an ideal model in which to compare as a control. Astrocytes were infected with viral supernatants known functions, and to investigate novel functions of cdk4 from DF-1 cells producing ALV that expressed the cDNAs of and cdk6. Results presented here demonstrate that infection of AP, cdk4, or cdk6. Protein expression in transfected DF-1 cells cdk6, but not cdk4, is sufficient to cause morphogenesis of was confirmed by immunoblot and virus-containing super- cultured mouse astrocytes. natants from DF-1 cells were filtered onto G-tva mouse astrocytes and infected for 3 days. Protein expression of cdk4 and cdk6 in these astrocytes was confirmed by immunoblot Results (Fig. 1A). Endogenous cdk6 was not expressed to detectable Morphological Differences in Cdk4- and levels in these astrocytes. Cdk6-Infected Astrocytes Within days after infection, it was apparent that cdk6- To study overlapping and discreet functions of cdk4 and infected astrocytes had a strikingly different morphology than cdk6, G-tva mouse astrocytes were infected with either of the those infected with cdk4. The cdk6-infected cells displayed a two D-cyclin-associated kinases or alkaline phosphatase (AP) fibroblast-like, bipolar morphology. Digital images of these FIGURE 1. Morphology of AP-, cdk4-, and cdk6- infected astroyctes. A. Immunoblot of primary mouse astrocytes infected with AP, cdk4, or cdk6, as indicated. One hundred twenty micrograms of protein extract were loaded per lane. The blot is probed with a mixture of cdk4 and cdk6 antibodies to detect both proteins on one blot. B. Phase-contrast micro- scopy of infected astroyctes grown in monolayer. All images are taken at the same magnification. RCAS-cdk4/6 are RCAS- cdk4-infected astrocytes that were super-infected with RCAS-cdk6 virus. C, D. Scanning electron microscopy (SEM) of fixed astrocytes at lower (C) and higher (D) magnifications. Scale bars on images indi- cate size. In panel C, scale bar is 20 AM; in panel D, scale bar is 10 AM. Downloaded from mcr.aacrjournals.org on October 1, 2021. © 2003 American Association for Cancer Research. 656 Expression of Cdk6 Alters Morphology of Astrocytes cells grown in a monolayer are shown in Fig. 1B. Alkaline- Cdk4-Infected and Cdk6-Infected Cells Display Distinct phosphatase and cdk4-infected astrocytes displayed a star- Growth Curves shaped, flat morphology containing several short projections Changes in cell size associated with cdk6 expression might be emanating from a wide central cell body. In sharp contrast, a result of changes in cellular proliferation. To study cell division cdk6-infected mouse astrocytes displayed an elongated spindle- properties of cdk4 and cdk6, growth curves were generated. shaped morphology with bipolar processes. Cdk6-infected Infected astrocytes were grown until confluent and then were cells contain a narrow soma that is collapsed around the split, counted, and replated at a density of 1 Â 105 cells per plate nucleus and greatly extended projections emanating from and followed over a period of up to 120 days. Growth curves of the central cell body. In addition, many of the cells are phase- one of four independent infections are displayed in Fig. 2. The bright in the area surrounding the central cell body, a property growth curves of both cdk4- and cdk6-infected cells were not seen in AP and cdk4 cells. The projections that extended remarkably consistent across all infections; in four separate trials from the cdk6 cells formed connections between adjacent cells of cdk4 and cdk6 infections, these cells escaped
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]
  • INK4 Locus of the Tumor-Resistant Rodent, the Naked Mole Rat, Expresses a Functional P15/P16 Hybrid Isoform
    INK4 locus of the tumor-resistant rodent, the naked mole rat, expresses a functional p15/p16 hybrid isoform Xiao Tiana,1, Jorge Azpuruaa,1,2, Zhonghe Kea, Adeline Augereaub, Zhengdong D. Zhangc, Jan Vijgc, Vadim N. Gladyshevb, Vera Gorbunovaa,3, and Andrei Seluanova,3 aDepartment of Biology, University of Rochester, Rochester, NY 14627; bBrigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115; and cAlbert Einstein College of Medicine, Bronx, NY 10461 Edited* by Eviatar Nevo, Institute of Evolution, Haifa, Israel, and approved December 1, 2014 (received for review September 21, 2014) The naked mole rat (Heterocephalus glaber) is a long-lived and because, in mammals, it encodes three distinct tumor suppressors: tumor-resistant rodent. Tumor resistance in the naked mole rat p16INK4a, p15INK4b, and p19ARF (p14ARF in human) (9). These is mediated by the extracellular matrix component hyaluronan three proteins coordinate a signaling network that depends on of very high molecular weight (HMW-HA). HMW-HA triggers hy- the activities of the retinoblastoma protein (RB) and the p53 persensitivity of naked mole rat cells to contact inhibition, which is tumor suppressor protein. The p16INK4a and p15INK4b pro- associated with induction of the INK4 (inhibitors of cyclin depen- teins are cyclin-dependent kinase inhibitors that directly in- dent kinase 4) locus leading to cell-cycle arrest. The INK4a/b locus is hibit the binding of cyclins to their target cyclin-dependent among the most frequently mutated in human cancer. This locus kinases (10). p16INK4a is involved in establishing replicative encodes three distinct tumor suppressors: p15INK4b, p16INK4a, and senescence, oncogene-induced senescence, and stress-induced INK4b ARF (alternate reading frame).
    [Show full text]
  • 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]
  • 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]
  • Galectin-4 Interaction with CD14 Triggers the Differentiation of Monocytes Into Macrophage-Like Cells Via the MAPK Signaling Pathway
    Immune Netw. 2019 Jun;19(3):e17 https://doi.org/10.4110/in.2019.19.e17 pISSN 1598-2629·eISSN 2092-6685 Original Article Galectin-4 Interaction with CD14 Triggers the Differentiation of Monocytes into Macrophage-like Cells via the MAPK Signaling Pathway So-Hee Hong 1,2,3,4,5, Jun-Seop Shin 1,2,3,5, Hyunwoo Chung 1,2,4,5, Chung-Gyu Park 1,2,3,4,5,* 1Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul 03080, Korea 2Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul 03080, Korea 3Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea 4Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea 5Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea Received: Jan 28, 2019 ABSTRACT Revised: May 13, 2019 Accepted: May 19, 2019 Galectin-4 (Gal-4) is a β-galactoside-binding protein mostly expressed in the gastrointestinal *Correspondence to tract of animals. Although intensive functional studies have been done for other galectin Chung-Gyu Park isoforms, the immunoregulatory function of Gal-4 still remains ambiguous. Here, we Department of Microbiology and Immunology, Seoul National University College of Medicine, demonstrated that Gal-4 could bind to CD14 on monocytes and induce their differentiation 103 Daehak-ro, Jongno-gu, Seoul 03080, into macrophage-like cells through the MAPK signaling pathway. Gal-4 induced the phenotypic Korea. changes on monocytes by altering the expression of various surface molecules, and induced E-mail: [email protected] functional changes such as increased cytokine production and matrix metalloproteinase expression and reduced phagocytic capacity.
    [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]
  • Targeting the Mitogen-Activated Protein Kinase Pathway: Physiological Feedback and Drug Response
    Published OnlineFirst May 14, 2010; DOI: 10.1158/1078-0432.CCR-09-3064 Molecular Pathways Clinical Cancer Research Targeting the Mitogen-Activated Protein Kinase Pathway: Physiological Feedback and Drug Response Christine A. Pratilas1,2 and David B. Solit3,4 Abstract Mitogen-activated protein kinase (MAPK) pathway activation is a frequent event in human cancer and is often the result of activating mutations in the BRAF and RAS oncogenes. Targeted inhibitors of BRAF and its downstream effectors are in various stages of preclinical and clinical development. These agents offer the possibility of greater efficacy and less toxicity than current therapies for tumors driven by oncogenic mutations in the MAPK pathway. Early clinical results with the BRAF-selective in- hibitor PLX4032 suggest that this strategy will prove successful in a select group of patients whose tu- mors are driven by V600E BRAF. Relief of physiologic feedback upon pathway inhibition may, however, attenuate drug response and contribute to the development of acquired resistance. An im- proved understanding of the adaptive response of cancer cells to MAPK pathway inhibition may thus aid in the identification of those patients most likely to respond to targeted pathway inhibitors and provide a rational basis for tailored combination strategies. Clin Cancer Res; 16(13); 3329–34. ©2010 AACR. Background these genetic alterations activate common downstream effectors of transformation. One of the central regulators of growth factor-induced Activating BRAF mutations are found clustered within cell proliferation and survival in both normal and cancer the P-loop (exon 11) and activation segment (exon 15) cells is the RAS protein.
    [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]
  • 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]