Combined Depletion of Cell Cycle and Transcriptional Cyclin- Dependent Kinase Activities Induces Apoptosis in Cancer Cells

Total Page:16

File Type:pdf, Size:1020Kb

Combined Depletion of Cell Cycle and Transcriptional Cyclin- Dependent Kinase Activities Induces Apoptosis in Cancer Cells Research Article Combined Depletion of Cell Cycle and Transcriptional Cyclin- Dependent Kinase Activities Induces Apoptosis in Cancer Cells Dongpo Cai,1,2 Vaughan M. Latham, Jr.,1,2 Xinxin Zhang,1,2 and Geoffrey I. Shapiro1,2 1Department of Medical Oncology, Dana-Farber Cancer Institute; 2Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts Abstract COOH-terminal domain (CTD) of RNA polymerase II and promote Selective cyclin-dependent kinase (cdk) 2 inhibition is readily initiation and elongation of nascent mRNA transcripts, respec- compensated. However, reduced cdk2 activity may have tively. Some of the cdks are involved in both cellular processes. antiproliferative effects in concert with other family members. For example, cyclin H-cdk7 is a component of cdk-activating Here, inducible RNAinterference was used to codeplete cdk2 kinase and contributes to cell cycle progression as well as and cdk1 from NCI-H1299 non–small cell lung cancer and transcription. Cyclin E-cdk2 and cyclin B-cdk1 have been shown U2OS osteosarcoma cells, and effects were compared with to phosphorylate the CTD in vitro (3, 4) and therefore may play a those mediated by depletion of either cdk alone. Depletion of role in transcriptional regulation at specific cell cycle phases. cdk2 slowed G progression of NCI-H1299 cells and depletion of Cdk2 was initially considered a viable antineoplastic drug 1 target, because several approaches inhibiting its activity in cancer cdk1 slowed G2-M progression in both cell lines, with associated endoreduplication in U2OS cells. However, com- cells have induced perturbations during multiple cell cycle phases pared with the incomplete cell cycle blocks produced by as well as apoptosis. For example, the inducible expression of a individual depletion, combined depletion had substantial dominant-negative cdk2 mutant in U2OS osteosarcoma cells led to G2 arrest after low-level induction and arrest during both S and consequences, with G2-M arrest predominating in NCI-H1299 cells and apoptosis the primary outcome in U2OS cells. In G2 phases when higher levels were induced (5). Effects on G1 U2OS cells, combined depletion affected RNApolymerase II progression also occurred but were weak and only unmasked expression and phosphorylation, causing decreased expression when cells were synchronized and released from a nocodazole- of the antiapoptotic proteins Mcl-1 and X-linked inhibitor of induced mitotic block. In exponentially growing transformed cell apoptosis (XIAP), effects usually mediated by inhibition of the lines, S and G2 cell cycle effects have also been described after ectopic expression of p27Kip1 (6) or introduction of peptides that transcriptional cdk9. These events do not occur after indi- vidual depletion of cdk2 and cdk1, suggesting that reduction inhibit cdk2 activity (7). The latter approaches have also resulted of cdk2, cdk1, and RNApolymerase II activities all contribute in significant cytotoxicity as has the targeted disruption of cyclin to apoptosis in U2OS cells. The limited cell death induced by A (8). combined depletion in NCI-H1299 cells was significantly These profound effects on S and G2 cell cycle progression and increased by codepletion of cdk9 or XIAP or by simultaneous apoptosis need to be reconciled with the absence of similar cell treatment with the cdk9 inhibitor flavopiridol. These results cycle effects or cell death after introduction of antisense or small show the potency of concomitant compromise of cell cycle and interfering RNA (siRNA) targeting cdk2 in cancer cells (9). One transcriptional cdk activities and may guide the selection of possibility is that the former approaches are targeting both cdk1 clinical drug candidates. (Cancer Res 2006; 66(18): 9270-80) and cdk2. For example, expression of the dominant-negative cdk2 mutant resulted in reduced cyclin B–associated kinase activity (5). Ectopic p27Kip1 would be expected to inhibit both cdks. The Introduction reported cdk2-inhibitory peptides were capable of cdk1 inhibition The cyclin-dependent kinases (cdk) comprise a family divided at high concentration, and the proteasomal degradation of cyclin into two groups based on their roles in cell cycle progression and A should affect cyclin A-cdk1 activity as well. In fact, small hairpin transcriptional regulation (1, 2). Members of the first group form RNA (shRNA)–mediated silencing of cdk1 produced greater effects À À the core components of the cell cycle machinery, including cyclin on cellular proliferation in cdk2 / mouse embryo fibroblasts D–dependent kinases 4 and 6, cyclin E-cdk2 complexes, cyclin than in wild-type cells (10), suggesting functional redundancy A–dependent kinases 2 and 1, and cyclin B-cdk1 complexes, which between these cdks. However, whether shRNA-mediated codeple- facilitate the transition between cell cycle phases. The second tion of cdk2 and cdk1 can similarly affect transformed cells or group includes cyclin H–dependent kinase 7 and cyclin induce apoptosis has not yet been clarified. Additionally, the T–dependent kinase 9 (p-TEFb), which phosphorylate the definition of the effects of combined depletion in established cancer cells is critical for the design of small-molecule cdk Note: Supplementary data for this article are available at Cancer Research Online inhibitors. Although compounds highly selective for cdk2 may (http://cancerres.aacrjournals.org/). have limited utility, those targeting both cdks with high potency Corrected online December 20, 2019. may have therapeutic value. D. Cai, V.M. Latham, Jr., and X. Zhang contributed equally to this work. Inhibition of the transcriptional cdks has also attracted intense Present address for X. Zhang: Department of Medicine, University of California, San Diego, CA. interest because the most sensitive transcripts are those with short Requests for reprints: Geoffrey I. Shapiro, Department of Medical Oncology, half-lives that encode cell cycle regulators, mitotic regulatory Dana-Farber Cancer Institute, Dana 810A, 44 Binney Street, Boston, MA 02115. Phone: kinases, nuclear factor-nB–responsive gene transcripts, and 617-632-4942; Fax: 617-632-1977; E-mail: [email protected]. I2006 American Association for Cancer Research. apoptosis regulators, such as Mcl-1 and X-linked inhibitor of doi:10.1158/0008-5472.CAN-06-1758 apoptosis (XIAP; refs. 11, 12). Diminution of levels of these Cancer ResDownloaded 2006; 66: (18). from September cancerres.aacrjournals.org 15, 2006 on October9270 1, 2021. © 2006 American Association forwww.aacrjournals.org Cancer Research. Downloaded from cancerres.aacrjournals.org on November 4, 2019. © 2006 American Association for Cancer Research. Dual Cell Cycle and Transcriptional Cdk Inhibition transcripts and their encoded proteins may produce anticancer cdk1 or cdk2 shRNA vectors as well as a vector encoding a hygromycin activity or augment apoptotic responses. Flavopiridol, the most selection marker at a 10:1 ratio; individual clones were selected in 200 Ag/ potent known inhibitor of cdk9, has recently shown striking mL hygromycin with blasticidin and puromycin and tested for knockdown A activity in chronic lymphocytic leukemia, which may be in part of both cdks after exposure to 5 g/mL doxycycline for 6 days. For cell growth experiments, cells were plated at 3.75 Â 105 per 10-cm dish in the attributable to a drug-induced decrease in Mcl-1 expression absence or presence of doxycycline. One quarter of the medium was (13, 14). The potential role of cdk2 and cdk1 in RNA polymerase replaced every 3 days. II CTD phosphorylation suggests that their inhibition may similarly Lentivirus production and infection. The CalPhos Mammalian deplete transcripts encoding proteins that affect cancer cell Transfection kit (Clontech Laboratories) was used to transfect lentiviral survival. shRNA as well as relevant helper plasmids into 293T cells. A NCI-H1299TR To define the effect of depletion of cdk2 and cdk1 activities in cdk2/cdk1 double shRNA clone was plated for 24 to 48 hours in the established cancer cells of diverse origin, we have engineered absence or presence of doxycycline before application of the appropriate NCI-H1299 non–small cell lung cancer (NSCLC) cells and U2OS lentivirus at a multiplicity of infection of f5. Cells were washed in PBS osteosarcoma cells to inducibly express shRNA targeting cdk2, 24 hours after transduction and maintained in medium in the absence or cdk1, or both together. This allowed us to probe the effects of presence of doxycycline for an additional 6 or 7 days for pLKO-cdk9 and pLVTHM-XIAP infection, respectively. One quarter of the medium was relatively acute depletion of these cdk activities, an appropriate replaced every 3 days. model for assessment of these kinases as drug targets. U2OS Cell synchronization. Cells were cultured in medium with or without cells express wild-type p53 and Rb as well as cyclins D1 and D3, doxycycline for 24 hours and subsequently replated at 6 Â 105 per 10-cm INK4A and lack p16 expression because of promoter hyper- dish and grown for an additional 24 hours before synchronization. Cells methylation (15, 16). They undergo S and G2-M arrest and were treated with 1 mmol/L hydroxyurea for 24 hours or 4 Ag/mL apoptosis in response to cdk2 inhibition by dominant-negative nocodazole (both from Sigma-Aldrich Co., St. Louis, MO) for 16 hours to cdk2 mutant expression (5), inhibitory peptides (7), and targeted achieve a G1-S or mitotic block, respectively. Cells were subsequently cyclin A degradation (8) but have not shown cell cycle washed in PBS and maintained in medium in the absence or presence of alterations in response to antisense-mediated specific depletion doxycycline. of cdk2 (9). NCI-H1299 cells are similarly Rb wild-type and Flavopiridol treatment. Flavopiridol was provided by the Drug p16INK4A methylated but carry a p53 deletion (17, 18). These cell Synthesis and Chemistry Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute. lines show that either G -M arrest or apoptosis is the major 2 A 50 mmol/L stock solution in DMSO was maintained at À20jC.
Recommended publications
  • Restoring MLL Reactivates Latent Tumor Suppression-Mediated Vulnerability to Proteasome Inhibitors
    Oncogene (2020) 39:5888–5901 https://doi.org/10.1038/s41388-020-01408-7 ARTICLE Restoring MLL reactivates latent tumor suppression-mediated vulnerability to proteasome inhibitors 1 1 2 1 3 1 4 2 Maolin Ge ● Dan Li ● Zhi Qiao ● Yan Sun ● Ting Kang ● Shouhai Zhu ● Shifen Wang ● Hua Xiao ● 1 5 4 1 Chunjun Zhao ● Shuhong Shen ● Zhenshu Xu ● Han Liu Received: 12 March 2020 / Revised: 16 July 2020 / Accepted: 23 July 2020 / Published online: 30 July 2020 © The Author(s) 2020. This article is published with open access Abstract MLL undergoes multiple distinct chromosomal translocations to yield aggressive leukemia with dismal outcomes. Besides their well-established role in leukemogenesis, MLL fusions also possess latent tumor-suppressive activity, which can be exploited as effective cancer treatment strategies using pharmacological means such as proteasome inhibitors (PIs). Here, using MLL-rearranged xenografts and MLL leukemic cells as models, we show that wild-type MLL is indispensable for the latent tumor-suppressive activity of MLL fusions. MLL dysfunction, shown as loss of the chromatin accumulation and subsequent degradation of MLL, compromises the latent tumor suppression of MLL-AF4 and is instrumental for the 1234567890();,: 1234567890();,: acquired PI resistance. Mechanistically, MLL dysfunction is caused by chronic PI treatment-induced epigenetic reprogramming through the H2Bub-ASH2L-MLL axis and can be specifically restored by histone deacetylase (HDAC) inhibitors, which induce histone acetylation and recruits MLL on chromatin to promote cell cycle gene expression. Our findings not only demonstrate the mechanism underlying the inevitable acquisition of PI resistance in MLL leukemic cells, but also illustrate that preventing the emergence of PI-resistant cells constitutes a novel rationale for combination therapy with PIs and HDAC inhibitors in MLL leukemias.
    [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]
  • 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]
  • Cell Cycle Arrest and DNA Endoreduplication Following P21waf1/Cip1 Expression
    Oncogene (1998) 17, 1691 ± 1703 1998 Stockton Press All rights reserved 0950 ± 9232/98 $12.00 http://www.stockton-press.co.uk/onc Cell cycle arrest and DNA endoreduplication following p21Waf1/Cip1 expression Stewart Bates, Kevin M Ryan, Andrew C Phillips and Karen H Vousden ABL Basic Research Program, NCI-FCRDC, Building 560, Room 22-96, West 7th Street, Frederick, Maryland 21702-1201, USA p21Waf1/Cip1 is a major transcriptional target of p53 and there are an increasing number of regulatory mechan- has been shown to be one of the principal mediators of isms that serve to inhibit cdk activity. Primary amongst the p53 induced G1 cell cycle arrest. We show that in these is the expression of cdk inhibitors (CDKIs) that addition to the G1 block, p21Waf1/Cip1 can also contribute can bind to and inhibit cyclin/cdk complexes (Sherr to a delay in G2 and expression of p21Waf1/Cip1 gives rise and Roberts, 1996). These fall into two classes, the to cell cycle pro®les essentially indistinguishable from INK family which bind to cdk4 and cdk6 and inhibit those obtained following p53 expression. Arrest of cells complex formation with D-cyclins, and the p21Waf1/Cip1 in G2 likely re¯ects an inability to induce cyclin B1/cdc2 family that bind to cyclin/cdk complexes and inhibit kinase activity in the presence of p21Waf1/Cip1, although the kinase activity. inecient association of p21Waf1/Cip1 and cyclin B1 The p21Waf1/Cip1 family of CDKIs include p21Waf1/Cip1, suggests that the mechanism of inhibition is indirect. p27 and p57 and are broad range inhibitors of cyclin/ Cells released from an S-phase block were not retarded cdk complexes (Gu et al., 1993; Harper et al., 1993; in their ability to progress through S-phase by the Xiong et al., 1993; Firpo et al., 1994; Polyak et al., presence of p21Waf1/Cip1, despite ecient inhibition of 1994; Toyoshima and Hunter, 1994; Lee et al., 1995; cyclin E, A and B1 dependent kinase activity, suggesting Matsuoka et al., 1995).
    [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]
  • Caspase-Dependent Activation of Cyclin-Dependent Kinases During Fas-Induced Apoptosis in Jurkat Cells
    Proc. Natl. Acad. Sci. USA Vol. 95, pp. 6785–6790, June 1998 Cell Biology Caspase-dependent activation of cyclin-dependent kinases during Fas-induced apoptosis in Jurkat cells BIN-BING ZHOU,HONGLIN LI,JUNYING YUAN, AND MARC W. KIRSCHNER Department of Cell Biology, Harvard Medical School, Boston, MA 02115 Contributed by Marc W. Kirschner, April 7, 1998 ABSTRACT The activation of cyclin-dependent kinases To study the mechanism of cdc2 and cdk2 activation in (cdks) has been implicated in apoptosis induced by various apoptotic cells, we examined cyclin synthesis, cyclin degrada- stimuli. We find that the Fas-induced activation of cdc2 and tion, and posttranslational modifications of cdc2 and cdk2 cdk2 in Jurkat cells is not dependent on protein synthesis, during Fas-induced apoptosis in Jurkat cells. We find that Fas which is shut down very early during apoptosis before induction activates cdc2 and cdk2, despite a potential loss of caspase-3 activation. Instead, activation of these kinases these proteins caused by the very rapid drop in the capacity for seems to result from both a rapid cleavage of Wee1 (an protein synthesis. Activation of these kinases seems to result inhibitory kinase of cdc2 and cdk2) and inactivation of from the maintenance of cyclin levels by rapid inactivation of anaphase-promoting complex (the specific system for cyclin APC, through a caspase-dependent cleavage of one of its degradation), in which CDC27 homolog is cleaved during subunits, and tyrosine dephosphorylation of cdks caused by a apoptosis. Both Wee1 and CDC27 are shown to be substrates cleavage of the inhibitory kinase Wee1.
    [Show full text]
  • The Expression of P21/WAF-1 and Cyclin B1 Mediate Mitotic Delay in X-Irradiated Fibroblasts
    ANTICANCER RESEARCH 25: 1123-1130 (2005) The Expression of p21/WAF-1 and Cyclin B1 Mediate Mitotic Delay in x-Irradiated Fibroblasts MICKAEL J. CARIVEAU1,2, CHARLES J. KOVACS2, RON R. ALLISON2, ROBERTA M. JOHNKE2, GERHARD W. KALMUS3 and MARK EVANS2 1Department of Physics, East Carolina University, Greenville NC, 27858; 2Department of Radiation Oncology, The Brody School of Medicine, East Carolina University, Greenville NC, 27858; 3Department of Biology, East Carolina University, Greenville NC, 27858, U.S.A. Abstract. Background: To better understand the relationship Initiation and maintenance of cell cycle arrest is regulated between mitotic delay and the disruption of cyclin B1 and p21 in by a group of proteins known as the cyclins which function x-irradiated fibroblasts, studies were carried out to establish by coupling to their corresponding cyclin dependent kinases correlations between the downregulation of cyclin B1 by the cyclin (CDK) (6-9). Of particular interest to cell cycle arrest in kinase inhibitor (CKI) p21 and the induction of mitotic delay in response to DNA damage is the G2/M damage checkpoint the NIH3T3 fibroblast. Materials and Methods: Cell cycle kinetics which is regulated by cyclin B1/CDK1 and initiated by DNA were used to analyze mitotic delay in irradiated NIH3T3 cells and damage activation of the cyclin kinase inhibitor p21(10-14). immunocytochemistry incorporated to assess the expression of The inhibitory potential of p21 is well established; however, cyclin B1 and p21, following 2 or 4Gy x-irradiation. Results and there is still much confusion about the role of p21 in the Discussion: Results indicate a dose dependent increase in mitotic irradiated cell (15, 16).
    [Show full text]
  • Epithelial Cell Death Analysis of Cell Cycle by Flow Cytometry White Paper
    Epithelial Cell Death Analysis of Cell Cycle by Flow Cytometry White Paper Authors: Savithri Balasubramanian, John Tigges, Vasilis Toxavidis, Heidi Mariani. Affiliation: Beth Israel Deaconess Medical Center Harvard Stem Cell Institute a Beckman Coulter Life Sciences: Epithelial Cell Death Analysis of Cell Cycle by Flow Cytometry PRINCIPAL OF TECHNIQUE Background: Cell cycle, or cell-division cycle, is the series of events that takes place in a cell leading to its division and duplication (replication). In cells without a nucleus (prokaryotic), cell cycle occurs via a process termed binary fission. In cells with a nucleus (eukaryotes), cell cycle can be divided in two brief periods: interphase—during which the cell grows, accumulating nutrients needed for mitosis and duplicating its DNA—and the mitosis (M) phase, during which the cell splits itself into two distinct cells, often called «daughter cells». Cell-division cycle is a vital process by which a single-celled fertilized egg develops into a mature organism, as well as the process by which hair, skin, blood cells, and some internal organs are renewed. Cell cycle consists of four distinct phases: G1 phase, S phase (synthesis), G2 phase (collectively known as interphase) and M phase (mitosis). M phase is itself composed of two tightly coupled processes: mitosis, in which the cell’s chromosomes are divided between the two daughter cells, and cytokinesis, in which the cell’s cytoplasm divides in half forming distinct cells. Activation of each phase is dependent on the proper progression and completion of the previous one. Cells that have temporarily or reversibly stopped dividing are said to have entered a state of quiescence called G0 phase.
    [Show full text]
  • Role of Cyclin-Dependent Kinase 1 in Translational Regulation in the M-Phase
    cells Review Role of Cyclin-Dependent Kinase 1 in Translational Regulation in the M-Phase Jaroslav Kalous *, Denisa Jansová and Andrej Šušor Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Rumburska 89, 27721 Libechov, Czech Republic; [email protected] (D.J.); [email protected] (A.Š.) * Correspondence: [email protected] Received: 28 April 2020; Accepted: 24 June 2020; Published: 27 June 2020 Abstract: Cyclin dependent kinase 1 (CDK1) has been primarily identified as a key cell cycle regulator in both mitosis and meiosis. Recently, an extramitotic function of CDK1 emerged when evidence was found that CDK1 is involved in many cellular events that are essential for cell proliferation and survival. In this review we summarize the involvement of CDK1 in the initiation and elongation steps of protein synthesis in the cell. During its activation, CDK1 influences the initiation of protein synthesis, promotes the activity of specific translational initiation factors and affects the functioning of a subset of elongation factors. Our review provides insights into gene expression regulation during the transcriptionally silent M-phase and describes quantitative and qualitative translational changes based on the extramitotic role of the cell cycle master regulator CDK1 to optimize temporal synthesis of proteins to sustain the division-related processes: mitosis and cytokinesis. Keywords: CDK1; 4E-BP1; mTOR; mRNA; translation; M-phase 1. Introduction 1.1. Cyclin Dependent Kinase 1 (CDK1) Is a Subunit of the M Phase-Promoting Factor (MPF) CDK1, a serine/threonine kinase, is a catalytic subunit of the M phase-promoting factor (MPF) complex which is essential for cell cycle control during the G1-S and G2-M phase transitions of eukaryotic cells.
    [Show full text]
  • Targeting Cyclin-Dependent Kinase 9 and Myeloid Cell Leukaemia 1 in MYC-Driven B-Cell Lymphoma
    Targeting cyclin-dependent kinase 9 and myeloid cell leukaemia 1 in MYC-driven B-cell lymphoma Gareth Peter Gregory ORCID ID: 0000-0002-4170-0682 Thesis for Doctor of Philosophy September 2016 Sir Peter MacCallum Department of Oncology The University of Melbourne Doctor of Philosophy Submitted in total fulfilment of the degree of Abstract Aggressive B-cell lymphomas include diffuse large B-cell lymphoma, Burkitt lymphoma and intermediate forms. Despite high response rates to conventional immuno-chemotherapeutic approaches, an unmet need for novel therapeutic by resistance to chemotherapy and radiotherapy. The proto-oncogene MYC is strategies is required in the setting of relapsed and refractory disease, typified frequently dysregulated in the aggressive B-cell lymphomas, however, it has proven an elusive direct therapeutic target. MYC-dysregulated disease maintains a ‘transcriptionally-addicted’ state, whereby perturbation of A significant body of evidence is accumulating to suggest that RNA polymerase II activity may indirectly antagonise MYC activity. Furthermore, very recent studies implicate anti-apoptotic myeloid cell leukaemia 1 (MCL-1) as a critical survival determinant of MYC-driven lymphoma. This thesis utilises pharmacologic and genetic techniques in MYC-driven models of aggressive B-cell lymphoma to demonstrate that cyclin-dependent kinase 9 (CDK9) and MCL-1 are oncogenic dependencies of this subset of disease. The cyclin-dependent kinase inhibitor, dinaciclib, and more selective CDK9 inhibitors downregulation of MCL1 are used
    [Show full text]