CDK2 Transcriptional Repression Is an Essential Effector in P53

Total Page:16

File Type:pdf, Size:1020Kb

CDK2 Transcriptional Repression Is an Essential Effector in P53 Published OnlineFirst August 22, 2014; DOI: 10.1158/1541-7786.MCR-14-0163 Cell Cycle and Senescence Molecular Cancer Research CDK2 Transcriptional Repression Is an Essential Effector in p53-Dependent Cellular Senescence— Implications for Therapeutic Intervention Hasan Zalzali1, Bilal Nasr1, Mohamad Harajly1, Hussein Basma1, Farah Ghamloush1, Sandra Ghayad1,Noel€ Ghanem2, Gerard I Evan3, and Raya Saab1 Abstract Cellular senescence, a form of cell-cycle arrest, is a tumor- cence. Pharmacologic inhibition of CDK2 in an in vivo model of suppressor mechanism triggered by multiple tumor-promoting pineal tumor decreased proliferation and promoted early senes- insults, including oncogenic stress and DNA damage. The role of cence, and it also decreased tumor penetrance and prolonged time cyclin-dependent kinase 2 (CDK2) regulation has been evaluated to tumor formation in animals lacking p53. In conclusion, for in models of replicative senescence, but little is known regarding both oncogene- and DNA damage–induced cellular senescence, its role in other senescence settings. Using in vitro and in vivo CDK2 transcript and protein are decreased in a p53- and RB- models of DNA damage–and oncogene-induced cellular senes- dependent manner, and this repression is necessary for cell-cycle cence, it was determined that activation of the tumor-suppressor exit during senescence. protein p53 (TP53) resulted in repression of the CDK2 transcript that was dependent on intact RB. Ectopic CDK2 expression was Implications: These data show that CDK2 inhibition may be sufficient to bypass p53-dependent senescence, and CDK2-spe- useful for cancer prevention in premalignant hyperproliferative cific inhibition, either pharmacologically (CVT313) or by use of a lesions, as well as established tumors. Mol Cancer Res; 13(1); 29–40. dominant-negative CDK2, was sufficient to induce early senes- Ó2014 AACR. Ink4b settings, other CDK inhibitors, such as CDKN2B (p15 ) and Introduction Ink4c CDKN2C (p18 ), have also been shown to play roles in this Cellular senescence is a well-documented tumor-suppressor process (11, 12). response, which in most instances is dependent on functional In contrast with the well-documented roles of the CDK inhi- TP53 (p53), and results in irreversible cell-cycle exit, so preventing Ink4a Cip1 Ink4b Ink4c bitors (p16 , p21 , p15 , and p18 ) in cellular senes- further tumor progression under the influence of oncogenic or cence, the roles of the respective CDKs are not well defined. genotoxic insults (reviewed in refs. 1, 2). Replicative senescence The role of CDK2 has been primarily studied in the setting of occurs due to telomere shortening, but senescence can also be replicative senescence, in which several investigators have shown induced by oncogene activation, loss of tumor suppressors, or that activity of CDK2 decreases in senescent fibroblasts, likely direct DNA damage (reviewed in ref. 3). Cell-cycle regulators are Cip1 through inhibition of Cyclin A–CDK2 complexes by p21 , critically involved in the execution of the senescence response. In whereas CDK2 protein levels remain unchanged (13–15); where- human fibroblasts, the cyclin-dependent kinase (CDK) inhibi- Ink4a Cip1 as in replicative senescence of cultured human umbilical vein tors, CDKN2a (p16 ) and CDKN1A (p21 ), are activated in endothelial cells (HUVEC), reduction of both CDK2 activity and response to oncogenic signals and are essential for cell-cycle exit protein levels were noted (16). Transduction of replicatively during cellular senescence (4–8). In senescence caused by geno- senescent cells with Cyclin E–CDK2 complexes results in cell- toxic insults, such as irradiation or topoisomerase inhibition, p53 Cip1 cycle reentry in a subset of cells, indicating that reduction of CDK2 is activated and this results in p21 induction, which is essential activity is indeed essential for replicative senescence (17). for cell-cycle exit and senescence (5, 9, 10). In yet other in vivo In oncogene-induced senescence, limited data are available, but suggest a physiologic role for CDK2 in opposing senescence. V12 For instance, in RAS -induced senescence, endogenous CDK2 1Department of Pediatric and Adolescent Medicine, American Univer- activity is reported to be decreased (6). MYC-induced senescence sity of Beirut, Beirut, Lebanon. 2Department of Biology, American occurs in a Cdk2-null background (and upon pharmacologic 3 University of Beirut, Beirut, Lebanon. Department of Biochemistry, CDK2 inhibition in fibroblasts), but not in Cdk4-null or Cdk6- University of Cambridge, Cambridge, United Kingdom. null backgrounds (18). We previously showed that in a mouse B. Nasr and M. Harajly contributed equally to this article. model of CCND1 (Cyclin D1)-driven senescence, CDK2 protein Corresponding Author: Hasan Zalzali or Raya Saab, American University of levels are also markedly decreased (19). All the above data Beirut, Riad El Solh Street, Beirut 1107 2020, Lebanon. Phone: 961-1-350000, ext. together prompted us to investigate the role of CDK2 in onco- 4780. Fax: 961-1-377384; E-mail: [email protected]; [email protected] gene-induced and DNA damage–induced senescence. In this doi: 10.1158/1541-7786.MCR-14-0163 report, we uncover a central physiologic role for p53-mediated Ó2014 American Association for Cancer Research. repression of CDK2 during oncogene-induced senescence, and www.aacrjournals.org 29 Downloaded from mcr.aacrjournals.org on September 26, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst August 22, 2014; DOI: 10.1158/1541-7786.MCR-14-0163 Zalzali et al. highlight CDK2 as a possible target to limit premalignant lesions, staining, cells were fixed and stained overnight at 37C, and as well as inhibit progression of established cancer, via induction counterstained with eosin, as in our previous studies (12, 19). of senescence. For BrdUrd incorporation assays, cells were treated with BrdUrd at a concentration of 33 mmol/L for 2 hours, fixed with 50% Materials and Methods methanol/50% acetone solution for 2 minutes, then treated with 2N HCL for 10 minutes, and neutralized by Borate buffer for 12 Mouse studies À À minutes. After blocking, slides were probed with anti-BrdUrd Irbp-Cyclin D1 transgenic mice (20) were bred with p53 / mice antibody (Santa Cruz Biotechnology), Cy3-conjugated secondary (The Jackson Laboratory) or p53ERTAM ki/ki mice (21) and antibody (Jackson Immunoresearch) or Alexa 488–conjugated maintained in a mixed C57BL/6  129/Sv genetic background. antibody (Invitrogen) was used for detection. Stained cells were PCR for targeted alleles was used to verify mouse genotypes as covered with aqueous mounting medium containing DAPI (Vec- described previously (20–22). Animals were euthanized at defined tor Laboratories) and visualized by immunofluorescence micros- time points or when obviously ill in accordance with the American copy. The number of BrdUrd-positive cells was manually counted University of Beirut Institutional Animal Care and Use Committee from at least five representative fields, at Â20 magnification, and guidelines; all animal studies were approved by this committee. normalized to total cell number (counted as DAPI-positive For all in vivo studies, CVT313 was administered by once daily i. nuclei). Digital photomicrographs were obtained using a Zeiss p. injection of 1.25 mg/kg. For bromodeoxyuridine (BrdUrd) 510 NLO multiphoton/confocal laser scanning microscope. incorporation assays, mice were given an i.p. injection of 50 Composite images were constructed using Photoshop CS6 soft- mg/kg of BrdUrd (Sigma-Aldrich), every 2 hours  5 and sacri- ware (Adobe Systems). ficed 2 hours later. Cell accumulation assay Plasmid constructs, virus production, and transduction Cells were plated in 6-well plates at a density of 200,000 cells CMV–CDK2–HA and CMV–CDK2DN–HA plasmid constructs per well. For senescence induction, cells were exposed to 20 Gy were obtained from Addgene [Addgene plasmids 1884 and 1885 irradiation at 1 Gy/min, treated with 10 mmol/L etoposide or (ref. 23); www.addgene.org], and subcloning of the constructs transduced with RasV12-GFP. The cells were collected at 7 days was performed into a lentivirus backbone expressing GFP sepa- V12 (after Ras transduction) or 14 days (after DNA damage), fixed rated by an IRES site. The RasV12-GFP plasmid was purchased with methanol: acetic acid 3:1 for 5 minutes, then stained in 0.5% from Clonetech. HEK 293T cells were used for lentivirus produc- Crystal Violet in methanol for 15 minutes. A dissecting micro- tion (CDK2 constructs) and retrovirus production (RasV12 con- scope was used to view the cells under a magnified field. structs), using calcium phosphate transfection with the appropri- ate respective packaging plasmids. Cell explantation and ex vivo culture Cell culture Mice were euthanized according to the IACUC-approved pro- Human BJ foreskin fibroblasts (ATCC) were cultured at 37C tocol. Pineal cells were explanted at postnatal day 10 (P10), mechanically dispersed, and cells were plated onto 8-well perma- and 5% CO2 in a cell incubator, in DMEM containing 10% FBS, 1% glutamine, and 1% pen/strep. Mouse embryonic fibroblasts nox chamber slides (Nunc), and cultured in DMEM with 10% (MEF) were isolated from E13.5 embryos from wild-type (WT), FBS, 1% glutamine, and 1% pen/strep. Explanted cells were À À ki/ki fl/fl p53 / , p53ERTAM ,andRb mice (in which exon 19 of the treated with CVT313 (Santa Cruz Biotechnology) or DMSO fi Rb gene is flanked by two LoxP sites, allowing induction of vehicle; media were renewed every 3 days, and cells were xed Cre-mediated recombination; ref. 24). MEFs were cultured in after 7 days. BrdUrd and SABG staining and cell counting were DMEM containing 10% FBS, 1% nonessential amino acids, 1% performed as detailed above. Digital photomicrographs were sodium pyruvate, 1% glutamine, and 1% pen/strep. For MEFs analyzed using Adobe Photoshop CS4 software. ki/ki derived from p53ERTAM mice, p53 was reversibly switched on and off by addition or withdrawal of 100 nmol/L 4-OH Histologic studies and immunostaining fl/fl tamoxifen, as specified.
Recommended publications
  • Expression Profiling of KLF4
    Expression Profiling of KLF4 AJCR0000006 Supplemental Data Figure S1. Snapshot of enriched gene sets identified by GSEA in Klf4-null MEFs. Figure S2. Snapshot of enriched gene sets identified by GSEA in wild type MEFs. 98 Am J Cancer Res 2011;1(1):85-97 Table S1: Functional Annotation Clustering of Genes Up-Regulated in Klf4 -Null MEFs ILLUMINA_ID Gene Symbol Gene Name (Description) P -value Fold-Change Cell Cycle 8.00E-03 ILMN_1217331 Mcm6 MINICHROMOSOME MAINTENANCE DEFICIENT 6 40.36 ILMN_2723931 E2f6 E2F TRANSCRIPTION FACTOR 6 26.8 ILMN_2724570 Mapk12 MITOGEN-ACTIVATED PROTEIN KINASE 12 22.19 ILMN_1218470 Cdk2 CYCLIN-DEPENDENT KINASE 2 9.32 ILMN_1234909 Tipin TIMELESS INTERACTING PROTEIN 5.3 ILMN_1212692 Mapk13 SAPK/ERK/KINASE 4 4.96 ILMN_2666690 Cul7 CULLIN 7 2.23 ILMN_2681776 Mapk6 MITOGEN ACTIVATED PROTEIN KINASE 4 2.11 ILMN_2652909 Ddit3 DNA-DAMAGE INDUCIBLE TRANSCRIPT 3 2.07 ILMN_2742152 Gadd45a GROWTH ARREST AND DNA-DAMAGE-INDUCIBLE 45 ALPHA 1.92 ILMN_1212787 Pttg1 PITUITARY TUMOR-TRANSFORMING 1 1.8 ILMN_1216721 Cdk5 CYCLIN-DEPENDENT KINASE 5 1.78 ILMN_1227009 Gas2l1 GROWTH ARREST-SPECIFIC 2 LIKE 1 1.74 ILMN_2663009 Rassf5 RAS ASSOCIATION (RALGDS/AF-6) DOMAIN FAMILY 5 1.64 ILMN_1220454 Anapc13 ANAPHASE PROMOTING COMPLEX SUBUNIT 13 1.61 ILMN_1216213 Incenp INNER CENTROMERE PROTEIN 1.56 ILMN_1256301 Rcc2 REGULATOR OF CHROMOSOME CONDENSATION 2 1.53 Extracellular Matrix 5.80E-06 ILMN_2735184 Col18a1 PROCOLLAGEN, TYPE XVIII, ALPHA 1 51.5 ILMN_1223997 Crtap CARTILAGE ASSOCIATED PROTEIN 32.74 ILMN_2753809 Mmp3 MATRIX METALLOPEPTIDASE
    [Show full text]
  • Charles M. Perou, Phd Associate Professor Departments of Genetics
    Charles M. Perou, PhD Associate Professor Departments of Genetics and Pathology Carolina Center for Genome Sciences Lineberger Comprehensive Cancer Center University of North Carolina at Chapel Hill Identification of GBM Subtypes using Gene Expression Profiling Agilent Custom Affymetrix Human Affymetrix HT-HG- 244k Exon 1.0 Array U133A Array Microarray Identify Samples and Genes represented on all 3 platforms Factor Analysis Single unified gene expression measurement for each gene 202 patients 11,681 genes Identification of GBM Subtypes Census Clustering of the 202 samples X 1740 Unsupervised clustering of 1740 genes suggests that 4 subtypes of GBM exist variably expressed genes selected using a unified gene expression measure across 3 expression platforms Core TCGA Samples (173) Gene Ontology/Pathway: with Subtype-defining genes • ProNeural: 1. nervous system development ProNeural Normal-like EGFR Mesenchymal 2. neuron differentiation (SOXs) 3. cell cycle = proliferation 4. cell adhesion molecules 5. ErbB signaling pathway FBXO3 GABRB2 • Normal‐like: SNCG NTSR2 1. nucleotide metabolic process MBP 2. neurological system process DLL3 3. axon NKX2-2, NRXN1, NRXN2 TOP2B, CDC7, MYB 4. neuron projection SOX2, SOX4, SOX10,SOX11 5. synaptic transmission ERBB3, PAK3, PAK7 NCAM1 OLIG2 • EGFR: 1. regulation of transcription 2. cell migration 3. nervous system development FGFR3 4. cell proliferation PDGFRA 5. metal ion binding EGFR AKT2 GLI2 TGFB3 • Mesenchymal: CASP8, CASP5, CASP4, CASP1 1. immune response COL8A2, COL5A1, COL1A1,COL1A2 2. receptor activity ILR4 CHI3L1 3. wound healing TRADD TLR2, TLR4, 4. cytokine and chemokine IGFBI mediated signaling pathway RELB 5. NF‐B Signaling Pathway Correlations between gene expression subtypes and clinical parameters Survival Analysis of Subtypes n=196 Subtypes are correlated with: 1.
    [Show full text]
  • Supplemental Table S1 (A): Microarray Datasets Characteristics
    Supplemental table S1 (A): Microarray datasets characteristics Title Summary Samples Literature ref. GEO ref. Acquisition of granule Gene expression profiling of 27 (1) GSE 11859 neuron precursor identity cerebellar tumors generated and Hedgehog‐induced from various early and late medulloblastoma in mice. stage CNS progenitor cells Medulloblastomas derived Study of mouse 5 (2) GSE 7212 from Cxcr6 mutant mice medulloblastoma in response respond to treatment with to inhibitor of Smoothened a Smoothened inhibitor Expression profiles of Identification of distinct classes 10 (3) GSE 9299 mouse medulloblastoma of up‐regulated or down‐ 339 & 340 regulated genes during Hh dependent tumorigenesis Genetic alterations in Identification of differently 10 (4) GSE 6463 mouse medulloblastomas expressed genes among CGNPs 339 & and generation of tumors and CGNPs transfected with 340 from cerebellar granule retroviruses that express nmyc neuron precursors or cyclin‐d1 Patched heterozygous Analysis of granule cell 14 (5) GSE 2426 model of medulloblastoma precursors, pre‐neoplastic cells, GDS1110 and tumor cells 1. Schuller U, Heine VM, Mao J, Kho AT, Dillon AK, Han YG, et al. Acquisition of granule neuron precursor identity is a critical determinant of progenitor cell competence to form Shh‐induced medulloblastoma. Cancer Cell 2008;14:123‐134. 2. Sasai K, Romer JT, Kimura H, Eberhart DE, Rice DS, Curran T. Medulloblastomas derived from Cxcr6 mutant mice respond to treatment with a smoothened inhibitor. Cancer Res 2007;67:3871‐3877. 3. Mao J, Ligon KL, Rakhlin EY, Thayer SP, Bronson RT, Rowitch D, et al. A novel somatic mouse model to survey tumorigenic potential applied to the Hedgehog pathway. Cancer Res 2006;66:10171‐10178.
    [Show full text]
  • Tagging Single Nucleotide Polymorphisms in Cell Cycle Control Genes and Susceptibility to Invasive Epithelial Ovarian Cancer
    Research Article Tagging Single Nucleotide Polymorphisms in Cell Cycle Control Genes and Susceptibility to Invasive Epithelial Ovarian Cancer Simon A. Gayther,1 Honglin Song,2 Susan J. Ramus,1 Susan Kru¨ger Kjaer,4 Alice S. Whittemore,5 Lydia Quaye,1 Jonathan Tyrer,2 Danielle Shadforth,2 Estrid Hogdall,4 Claus Hogdall,6 Jan Blaeker,7 Richard DiCioccio,8 Valerie McGuire,5 Penelope M. Webb,9 Jonathan Beesley,9 Adele C. Green,9 David C. Whiteman,9 The Australian Ovarian Cancer Study Group,9 The Australian Cancer Study (Ovarian Cancer),20 Marc T. Goodman,10 Galina Lurie,10 Michael E. Carney,10 Francesmary Modugno,10 Roberta B. Ness,11 Robert P. Edwards,12 Kirsten B. Moysich,7 Ellen L. Goode,13 Fergus J. Couch,13 Julie M. Cunningham,13 Thomas A. Sellers,14 Anna H. Wu,15 Malcolm C. Pike,15 Edwin S. Iversen,16 Jeffrey R. Marks,16 Montserrat Garcia-Closas,17 Louise Brinton,17 Jolanta Lissowska,18 Beata Peplonska,19 Douglas F. Easton,3 Ian Jacobs,1 Bruce A.J. Ponder,2 Joellen Schildkraut,16 C. Leigh Pearce,15 Georgia Chenevix-Trench,9 Andrew Berchuck,16 Paul D.P. Pharoah,2 and on behalf of the Ovarian Cancer Association Consortium 1Translational Research Laboratories, University College London, London, United Kingdom; 2Cancer Research United Kingdom Department of Oncology and 3Cancer Research United Kingdom Genetic Epidemiology Unit, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom; 4Danish Cancer Society, Copenhagen, Denmark; 5Stanford University School of Medicine, Stanford, California; 6University of Copenhagen,
    [Show full text]
  • CDKN2C-Null Leiomyosarcoma: a Novel
    original reports CDKN2C-Null Leiomyosarcoma: A Novel, Genomically Distinct Class of TP53/ RB1–Wild-Type Tumor With Frequent CIC Genomic Alterations and 1p/19q-Codeletion Erik A. Williams, MD1; Radwa Sharaf, PhD1; Brennan Decker, MD, PhD2; Adrienne J. Werth, MD3; Helen Toma, MD3; Meagan Montesion, PhD1; Ethan S. Sokol, PhD1; Dean C. Pavlick, BS1; Nikunj Shah, BS1; Kevin Jon Williams, MD4; Jeffrey M. Venstrom, MD1; Brian M. Alexander, MD, MPH1; Jeffrey S. Ross, MD1,5; Lee A. Albacker, PhD1; Douglas I. Lin, MD, PhD1; Shakti H. Ramkissoon, MD, PhD1,6; and Julia A. Elvin, MD, PhD1 abstract PURPOSE Leiomyosarcoma (LMS) harbors frequent mutations in TP53 and RB1 but few actionable genomic alterations. Here, we searched for recurrent actionable genomic alterations in LMS that occur in the absence of common untreatable oncogenic drivers. METHODS Tissues from 276,645 unique advanced cancers, including 2,570 uterine and soft tissue LMS, were sequenced by hybrid-capture–based next-generation DNA and RNA sequencing/comprehensive genomic profiling of up to 406 genes. We characterized clinicopathologic features of relevant patient cases. RESULTS Overall, 77 LMS exhibited homozygous copy loss of CDKN2C at chromosome 1p32.3 (3.0% of LMS). Genomic alterations (GAs) in TP53, RB1, and ATRX were rare compared with the remainder of the LMS cohort (11.7% v 73.4%, 0% v 54.5%, 2.6% v 24.5%, respectively; all P , .0001). CDKN2C-null LMS patient cases were significantly enriched for GAs in CIC (40.3% v 1.4%) at 19q13.2, CDKN2A (46.8% v 7.0%), and RAD51B (16.9% v 1.7%; all P , .0001).
    [Show full text]
  • Co-Deletion of P18 and P16 /P14 /P15 in Glioblastoma Multiforme
    Review Conspirators in a Capital Crime: Co-deletion of p18INK4c and p16INK4a/p14ARF/p15INK4b in Glioblastoma Multiforme David A. Solomon,1,2 Jung-Sik Kim,2 Walter Jean,3 and Todd Waldman2 1Tumor Biology Training Program, 2Department of Oncology, Lombardi Comprehensive Cancer Center, and 3Department of Neurosurgery, Georgetown University School of Medicine, Washington, District of Columbia Abstract and showing that p18INK4c-deficient mice are tumor prone, these INK4c Glioblastoma multiforme (GBM) is one of the most dreaded recent studies have suggested that inactivation of p18 may cancer diagnoses due to its poor prognosis and the limited play a perhaps underappreciated role in human cancer patho- INK4a genesis. As such, this review will provide a brief history of the treatment options. Homozygous deletion of the p16 / INK4c p14ARF/p15INK4b locus is among the most common genetic discovery of p18 , summarize the studies that link it to cancers in mice and humans, and provide a forward-looking alterations in GBM. Two recent studies have shown that INK4c deletion and mutation of another INK4 family member, assessment of the role of p18 inactivation in the pathogenesis p18INK4c, also drives the pathogenesis of GBM. This minireview of GBM (Fig. 1). INK4c will discuss the known roles for p18 in the initiation and INK4c progression of cancer and suggest opportunities for future Discovery of p18 studies. [Cancer Res 2008;68(21):8657–60] The human p18INK4c gene was initially discovered in 1994 by Guan and colleagues (8) in a yeast two-hybrid screen to identify Introduction proteins that interacted with human cdk6. The mouse homologue was reported the following year by Hirai and colleagues (9).
    [Show full text]
  • Deletions of CDKN2C in Multiple Myeloma: Biological and Clinical Implications Paola E
    Human Cancer Biology Deletions of CDKN2C in Multiple Myeloma: Biological and Clinical Implications Paola E. Leone,1Brian A. Walker,1Matthew W. Jenner,1Laura Chiecchio,2 GianPaolo Dagrada,2 Rebecca K.M. Protheroe,2 David C. Johnson,1Nicholas J. Dickens,1Jose Luis Brito,1Monica Else,1 David Gonzalez,1Fiona M. Ross,2 Selina Chen-Kiang,3 Faith E. Davies,1and Gareth J. Morgan1 Abstract Purpose: Deletions of chromosome 1have been described in 7% to 40% of cases of myeloma with inconsistent clinical consequences. CDKN2C at 1p32.3 has been identified in myeloma cell lines as the potential target of the deletion.We tested the clinical impact of 1p deletion and used high-resolution techniques to define the role of CDKN2C in primary patient material. Experimental Design: We analyzed 515 cases of monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), and newly diagnosed multiple mye- loma using fluorescence in situ hybridization (FISH) for deletions of CDKN2C.In78myeloma cases, we carried out Affymetrix single nucleotide polymorphism mapping and U133 Plus 2.0 ex- pression arrays. In addition, we did mutation, methylation, andWestern blotting analysis. Results: By FISH we identified deletion of 1p32.3 (CDKN2C)in3of66MGUS(4.5%),4of39 SMM (10.3%), and 55 of 369 multiple myeloma cases (15%).We examined the impact of copy number change at CDKN2C on overall survival (OS), and found that the cases with either hemi- zygous or homozygous deletion of CDKN2C had a worse OS compared with cases that were intact at this region (22 months versus 38 months; P = 0.003). Using gene mapping we identified three homozygous deletions at 1p32.3, containing CDKN2C,allofwhichlackedexpressionof CDKN2C.
    [Show full text]
  • Autoimmunity in a Mouse Model of Lupus Deficiency Promotes B1a
    Cyclin-Dependent Kinase Inhibitor Cdkn2c Deficiency Promotes B1a Cell Expansion and Autoimmunity in a Mouse Model of Lupus This information is current as Hari-Hara S. K. Potula, Zhiwei Xu, Leilani Zeumer, Allison of September 25, 2021. Sang, Byron P. Croker and Laurence Morel J Immunol 2012; 189:2931-2940; Prepublished online 15 August 2012; doi: 10.4049/jimmunol.1200556 http://www.jimmunol.org/content/189/6/2931 Downloaded from Supplementary http://www.jimmunol.org/content/suppl/2012/08/15/jimmunol.120055 Material 6.DC1 http://www.jimmunol.org/ References This article cites 42 articles, 14 of which you can access for free at: http://www.jimmunol.org/content/189/6/2931.full#ref-list-1 Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision by guest on September 25, 2021 • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2012 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Cyclin-Dependent Kinase Inhibitor Cdkn2c Deficiency Promotes B1a Cell Expansion and Autoimmunity in a Mouse Model of Lupus Hari-Hara S.
    [Show full text]
  • Cdkn2a, the Cyclin-Dependent Kinase Inhibitor Encoding P16ink4a and P19arf, Is a Candidate for the Plasmacytoma Susceptibility Locus, Pctr1
    Proc. Natl. Acad. Sci. USA Vol. 95, pp. 2429–2434, March 1998 Genetics Cdkn2a, the cyclin-dependent kinase inhibitor encoding p16INK4a and p19ARF, is a candidate for the plasmacytoma susceptibility locus, Pctr1 SHULING ZHANG,EDWARD S. RAMSAY, AND BEVERLY A. MOCK* Laboratory of Genetics, Division of Basic Sciences, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4255 Communicated by Michael Potter, National Institutes of Health, Bethesda, MD, December 24, 1997 (received for review November 17, 1997) ABSTRACT Plasma cell tumor induction in mice by cytoma susceptibilityyresistance locus resides. The protein pristane is under multigenic control. BALByc mice are sus- products of these genes bind to the CDKs CDK4 and CDK6 (6, ceptible to tumor development; whereas DBAy2 mice are 7), which, in turn, form complexes with the D-type G1 cyclins resistant. Restriction fragment length polymorphisms be- that phosphorylate pRb to control both cell cycle and tumor tween BALByc and DBAy2 for Cdkn2a(p16) and Cdkn2b(p15), suppression (8, 9). We compared the sequences of p16 and p18 and between BALByc and Mus spretus for Cdkn2c(p18INK4c) between susceptible (BALByc) and resistant (DBAy2N) were used to position these loci with respect to the Pctr1 locus. strains of mice, and found sequence variants in p16 located in These cyclin-dependent kinase (CDK) inhibitors mapped to a two different ankyrin repeat regions of the gene. DBAy2 (wild 6 cM interval of chromosome 4 between Ifna and Tal1. type) and BALByc A134C and G232A variants of the p16 C.D2-Chr 4 congenic strains harboring DBAy2 alleles asso- genes were fused to the glutathione S-transferase (GST) gene, ciated with the Pctr1 locus contained DBAy2 ‘‘resistant’’ and purified GST fusion proteins were tested for their ability alleles of the CDK4yCDK6 inhibitors p16 and p15.
    [Show full text]
  • Inhibition of CDK4/6 As Therapeutic Approach for Ovarian Cancer Patients: Current Evidences and Future Perspectives
    cancers Review Inhibition of CDK4/6 as Therapeutic Approach for Ovarian Cancer Patients: Current Evidences and Future Perspectives Alessandra Dall’Acqua 1,†, Michele Bartoletti 2,3,† , Nastaran Masoudi-Khoram 1,‡, Roberto Sorio 2, Fabio Puglisi 2,3 , Barbara Belletti 1 and Gustavo Baldassarre 1,* 1 Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, 33081 Aviano, Italy; [email protected] (A.D.); [email protected] (N.M.-K.); [email protected] (B.B.) 2 Medical Oncology and Cancer Prevention Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, 33081 Aviano, Italy; [email protected] (M.B.); [email protected] (R.S.); [email protected] (F.P.) 3 Department of Medicine (DAME), University of Udine, 33100 Udine, Italy * Correspondence: [email protected]; Tel.: +39-0434-659779; Fax: +39-0434-659429 † These authors equally contributed to the work. ‡ Present address: Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran. Simple Summary: Altered regulation of the cell cycle is a hallmark of cancer. The recent clinical success of the inhibitors of CDK4 and CDK6 has convincingly demonstrated that targeting cell cycle components may represent an effective anti-cancer strategy, at least in some cancer types. However, possible applications of CDK4/6 inhibitors in patients with ovarian cancer is still under evaluation. Citation: Dall’Acqua, A.; Bartoletti, M.; Masoudi-Khoram, N.; Sorio, R.; Here, we describe the possible biological role of CDK4 and CDK6 complexes in ovarian cancer and Puglisi, F.; Belletti, B.; Baldassarre, G. provide the rationale for the use of CDK4/6 inhibitors in this pathology, alone or in combination Inhibition of CDK4/6 as Therapeutic with other drugs.
    [Show full text]
  • The Role of Cdks and Cdkis in Murine Development
    International Journal of Molecular Sciences Review The Role of CDKs and CDKIs in Murine Development Grace Jean Campbell , Emma Langdale Hands and Mathew Van de Pette * Epigenetic Mechanisms of Toxicology Lab, MRC Toxicology Unit, Cambridge University, Cambridge CB2 1QR, UK; [email protected] (G.J.C.); [email protected] (E.L.H.) * Correspondence: [email protected] Received: 8 July 2020; Accepted: 26 July 2020; Published: 28 July 2020 Abstract: Cyclin-dependent kinases (CDKs) and their inhibitors (CDKIs) play pivotal roles in the regulation of the cell cycle. As a result of these functions, it may be extrapolated that they are essential for appropriate embryonic development. The twenty known mouse CDKs and eight CDKIs have been studied to varying degrees in the developing mouse, but only a handful of CDKs and a single CDKI have been shown to be absolutely required for murine embryonic development. What has become apparent, as more studies have shone light on these family members, is that in addition to their primary functional role in regulating the cell cycle, many of these genes are also controlling specific cell fates by directing differentiation in various tissues. Here we review the extensive mouse models that have been generated to study the functions of CDKs and CDKIs, and discuss their varying roles in murine embryonic development, with a particular focus on the brain, pancreas and fertility. Keywords: cyclin-dependent kinase; CDK inhibitors; mouse; development; knock-out models 1. Introduction Cyclin-dependent kinases (CDKs) are proteins that, by definition, require the binding of partner cyclin proteins in order to phosphorylate a series of target proteins.
    [Show full text]
  • MEN4 and CDKN1B Mutations 24:10 T195–T208 Thematic Review
    2410 R Alrezk et al. MEN4 and CDKN1B mutations 24:10 T195–T208 Thematic Review MEN4 and CDKN1B mutations: the latest of the MEN syndromes Rami Alrezk1, Fady Hannah-Shmouni2 and Constantine A Stratakis2 1 The National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Correspondence Maryland, USA should be addressed 2 Section on Endocrinology & Genetics, the Eunice Kennedy Shriver National Institute of Child Health and Human to C A Stratakis Development, NIH, Bethesda, Maryland, USA Email [email protected] Abstract Multiple endocrine neoplasia (MEN) refers to a group of autosomal dominant disorders Key Words with generally high penetrance that lead to the development of a wide spectrum of f multiple endocrine endocrine and non-endocrine manifestations. The most frequent among these conditions neoplasia is MEN type 1 (MEN1), which is caused by germline heterozygous loss-of-function f MEN4 mutations in the tumor suppressor gene MEN1. MEN1 is characterized by primary f MEN1 hyperparathyroidism (PHPT) and functional or nonfunctional pancreatic neuroendocrine f neuroendocrine tumors tumors and pituitary adenomas. Approximately 10% of patients with familial or sporadic f CDKN1B MEN1-like phenotype do not have MEN1 mutations or deletions. A novel MEN syndrome f p27 was discovered, initially in rats (MENX), and later in humans (MEN4), which is caused by germline mutations in the putative tumor suppressor CDKN1B. The most common phenotype of the 19 established cases of MEN4 that have been described to date is PHPT Endocrine-Related Cancer Endocrine-Related followed by pituitary adenomas. Recently, somatic or germline mutations in CDKN1B were also identified in patients with sporadic PHPT, small intestinal neuroendocrine tumors, lymphoma and breast cancer, demonstrating a novel role for CDKN1B as a tumor susceptibility gene for other neoplasms.
    [Show full text]