1
|
Liu P, Jiang J, Chen Y, Gao F, Wang S, Yu M, Liu Y, Guo R, Zhang L, Xu Z, Wang C, Qi X, Zhang Y, Cui H, Duan Y, Wu S, Gao Y. Identification of Cables1 as a critical host factor that promotes ALV-J replication via genome-wide CRISPR/Cas9 gene knockout screening. J Biol Chem 2024; 300:107804. [PMID: 39307305 DOI: 10.1016/j.jbc.2024.107804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 10/21/2024] Open
Abstract
Avian leukosis virus subgroup J (ALV-J), a member of the genus Alpharetrovirus, possesses a small genome and exploits a vast array of host factors during its replication cycle. To identify host factors required for ALV-J replication and potentially guide the development of key therapeutic targets for ALV-J prevention, we employed a chicken genome-wide CRISPR/Cas9 knockout library to screen host factors involved in ALV-J infection within DF-1 cells. This screening revealed 42 host factors critical for ALV-J infection. Subsequent knockout assays showed that the absence of the genes encoding cycle-regulatory proteins, namely, Cables1, CDK1, and DHFR, significantly inhibited ALV-J replication. Notably, Cables1 knockout cell lines displayed the most pronounced inhibitory effect. Conversely, overexpression assays confirmed that Cables1 significantly promotes ALV-J replication. Immunoprecipitation assays further indicated that Cables1 specifically interacts with the viral protein p15 (viral protease) among all ALV-J proteins, enhancing ALV-J p15 polyubiquitination. Additionally, we identified 26 lysine residues of ALV-J p15 as key sites for ubiquitination, and their replacement with arginine attenuated the replication ability of ALV-J in both in vitro and in vivo assays. This study demonstrates that Cables1 is a critical replication-dependent host factor of ALV-J by enhancing p15 ubiquitination and thereby promoting viral replication. Overall, these findings contribute to a deeper understanding of the ALJ-V replication mechanism and offer a potential target for the prevention and control of ALV-J infection.
Collapse
Affiliation(s)
- Peng Liu
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Jinghua Jiang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Yuntong Chen
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Fei Gao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Suyan Wang
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Mengmeng Yu
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Yongzhen Liu
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Ru Guo
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Li Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Zhuangzhuang Xu
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Caiying Wang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Xiaole Qi
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Yanping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Hongyu Cui
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Yulu Duan
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China
| | - Sen Wu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, PR China; Frontiers Science Center for Molecular Design Breeding, China Agricultural University, Beijing, China.
| | - Yulong Gao
- State Key Laboratory for Animal Disease Control and Prevention, Avian Immunosuppressive Diseases Division, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin, PR China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou, PR China; National Poultry Laboratory Animal Resource Center, Harbin, PR China.
| |
Collapse
|
2
|
Tripp JA, Berrio A, McGraw LA, Matz MV, Davis JK, Inoue K, Thomas JW, Young LJ, Phelps SM. Comparative neurotranscriptomics reveal widespread species differences associated with bonding. BMC Genomics 2021; 22:399. [PMID: 34058981 PMCID: PMC8165761 DOI: 10.1186/s12864-021-07720-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/20/2021] [Indexed: 11/28/2022] Open
Abstract
Background Pair bonding with a reproductive partner is rare among mammals but is an important feature of human social behavior. Decades of research on monogamous prairie voles (Microtus ochrogaster), along with comparative studies using the related non-bonding meadow vole (M. pennsylvanicus), have revealed many of the neural and molecular mechanisms necessary for pair-bond formation in that species. However, these studies have largely focused on just a few neuromodulatory systems. To test the hypothesis that neural gene expression differences underlie differential capacities to bond, we performed RNA-sequencing on tissue from three brain regions important for bonding and other social behaviors across bond-forming prairie voles and non-bonding meadow voles. We examined gene expression in the amygdala, hypothalamus, and combined ventral pallidum/nucleus accumbens in virgins and at three time points after mating to understand species differences in gene expression at baseline, in response to mating, and during bond formation. Results We first identified species and brain region as the factors most strongly associated with gene expression in our samples. Next, we found gene categories related to cell structure, translation, and metabolism that differed in expression across species in virgins, as well as categories associated with cell structure, synaptic and neuroendocrine signaling, and transcription and translation that varied among the focal regions in our study. Additionally, we identified genes that were differentially expressed across species after mating in each of our regions of interest. These include genes involved in regulating transcription, neuron structure, and synaptic plasticity. Finally, we identified modules of co-regulated genes that were strongly correlated with brain region in both species, and modules that were correlated with post-mating time points in prairie voles but not meadow voles. Conclusions These results reinforce the importance of pre-mating differences that confer the ability to form pair bonds in prairie voles but not promiscuous species such as meadow voles. Gene ontology analysis supports the hypothesis that pair-bond formation involves transcriptional regulation, and changes in neuronal structure. Together, our results expand knowledge of the genes involved in the pair bonding process and open new avenues of research in the molecular mechanisms of bond formation. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07720-0.
Collapse
Affiliation(s)
- Joel A Tripp
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Alejandro Berrio
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, 78712, USA.,Present Address: Department of Biology, Duke University, Durham, NC, 27708, USA
| | - Lisa A McGraw
- Center for Translational Social Neuroscience, Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Mikhail V Matz
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Jamie K Davis
- Centers for Disease Control and Prevention, Atlanta, GA, 30333, USA
| | - Kiyoshi Inoue
- Center for Translational Social Neuroscience, Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - James W Thomas
- National Institutes of Health Intramural Sequencing Center, National Human Genome Research Institute, National Institutes of Health, Rockville, MD, USA
| | - Larry J Young
- Center for Translational Social Neuroscience, Department of Psychiatry and Behavioral Sciences, Yerkes National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Steven M Phelps
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
3
|
Hasan ASH, Dinh TTH, Le HT, Mizuno-Iijima S, Daitoku Y, Ishida M, Tanimoto Y, Kato K, Yoshiki A, Murata K, Mizuno S, Sugiyama F. Characterization of a bicistronic knock-in reporter mouse model for investigating the role of CABLES2 in vivo. Exp Anim 2021; 70:22-30. [PMID: 32779618 PMCID: PMC7887623 DOI: 10.1538/expanim.20-0063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/05/2020] [Indexed: 12/16/2022] Open
Abstract
Two members of the CDK5 and ABL enzyme substrate (CABLES) family, CABLES1 and CABLES2, share a highly homologous C-terminus. They interact and associate with cyclin-dependent kinase 3 (CDK3), CDK5, and c-ABL. CABLES1 mediates tumor suppression, regulates cell proliferation, and prevents protein degradation. Although Cables2 is ubiquitously expressed in adult mouse tissues at RNA level, the role of CABLES2 in vivo remains unknown. Here, we generated bicistronic Cables2 knock-in reporter mice that expressed CABLES2 tagged with 3×FLAG and 2A-mediated fluorescent reporter tdTomato. Cables2-3×FLAG-2A-tdTomato (Cables2Tom) mice confirmed the expression of Cables2 in various mouse tissues. Interestingly, high intensity of tdTomato fluorescence was observed in the brain, testis and ovary, especially in the corpus luteum. Furthermore, immunoprecipitation analysis using the brain and testis in Cables2Tom/Tom revealed interaction of CABLES2 with CDK5. Collectively, our new Cables2 knock-in reporter model will enable the comprehensive analysis of in vivo CABLES2 function.
Collapse
Affiliation(s)
- Ammar Shaker Hamed Hasan
- Laboratory Animal Resource Center, Trans-Border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Doctor's Program in Biomedical Sciences, Graduate School of Comprehensive Human Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Ministry of Works, Municipalities Affairs and Urban Planning, Building 86, Block 318, Sheikh Hamad Street 1802, Manama Diplomatic Area, Manama, Bahrain
| | - Tra Thi Huong Dinh
- Laboratory Animal Resource Center, Trans-Border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hoai Thu Le
- Laboratory Animal Resource Center, Trans-Border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors (SIGMA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Saori Mizuno-Iijima
- Experimental Animal Division, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Yoko Daitoku
- Laboratory Animal Resource Center, Trans-Border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Miyuki Ishida
- Laboratory Animal Resource Center, Trans-Border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yoko Tanimoto
- Laboratory Animal Resource Center, Trans-Border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Kanako Kato
- Laboratory Animal Resource Center, Trans-Border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Atsushi Yoshiki
- Experimental Animal Division, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Kazuya Murata
- Laboratory Animal Resource Center, Trans-Border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Trans-Border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, Trans-Border Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
4
|
Shen AJJ, King J, Scott H, Colman P, Yates CJ. Insights into pituitary tumorigenesis: from Sanger sequencing to next-generation sequencing and beyond. Expert Rev Endocrinol Metab 2019; 14:399-418. [PMID: 31793361 DOI: 10.1080/17446651.2019.1689120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
Introduction: This review explores insights provided by next-generation sequencing (NGS) of pituitary tumors and the clinical implications.Areas covered: Although syndromic forms account for just 5% of pituitary tumours, past Sanger sequencing studies pragmatically focused on them. These studies identified mutations in MEN1, CDKN1B, PRKAR1A, GNAS and SDHx causing Multiple Endocrine Neoplasia-1 (MEN1), MEN4, Carney Complex-1, McCune Albright Syndrome and 3P association syndromes, respectively. Furthermore, linkage analysis of single-nucleotide polymorphisms identified AIP mutations in 20% with familial isolated pituitary adenomas (FIPA). NGS has enabled further investigation of sporadic tumours. Thus, mutations of USP8 and CABLES1 were identified in corticotrophinomas, BRAF in papillary craniopharyngiomas and CTNNB1 in adamantinomatous craniopharyngiomas. NGS also revealed that pituitary tumours occur in the DICER1 syndrome, due to DICER1 mutations, and CDH23 mutations occur in FIPA. These discoveries revealed novel therapeutic targets and studies are underway of BRAF inhibitors for papillary craniopharyngiomas, and EGFR and USP8 inhibitors for corticotrophinomas.Expert opinion: It has become apparent that single-nucleotide variants and small insertion/deletion DNA mutations cannot explain all pituitary tumorigenesis. Integrated and improved analyses including whole-genome sequencing, copy number, and structural variation analyses, RNA sequencing and epigenomic analyses, with improved genomic technologies, are likely to further define the genomic landscape.
Collapse
Affiliation(s)
| | - James King
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Australia
| | - Hamish Scott
- Department of Genetics and Molecular Pathology, Center for Cancer Biology, SA Pathology, Adelaide, Australia
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, Australia
- School of Medicine, University of Adelaide, Adelaide, Australia
- Australian Cancer Research Foundation Genomics Facility, Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | - Peter Colman
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Diabetes and Endocrinology, The Royal Melbourne Hospital, Parkville, Australia
| | - Christopher J Yates
- Department of Medicine, The University of Melbourne, Parkville, Australia
- Department of Diabetes and Endocrinology, The Royal Melbourne Hospital, Parkville, Australia
| |
Collapse
|
5
|
CABLES1 Deficiency Impairs Quiescence and Stress Responses of Hematopoietic Stem Cells in Intrinsic and Extrinsic Manners. Stem Cell Reports 2019; 13:274-290. [PMID: 31327733 PMCID: PMC6700604 DOI: 10.1016/j.stemcr.2019.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/20/2022] Open
Abstract
Bone marrow (BM) niche cells help to keep adult hematopoietic stem cells (HSCs) in a quiescent state via secreted factors and induction of cell-cycle inhibitors. Here, we demonstrate that the adapter protein CABLES1 is a key regulator of long-term hematopoietic homeostasis during stress and aging. Young mice lacking Cables1 displayed hyperproliferation of hematopoietic progenitor cells. This defect was cell intrinsic, since it was reproduced in BM transplantation assays using wild-type animals as recipients. Overexpression and short hairpin RNA-mediated depletion of CABLES1 protein resulted in p21Cip/waf up- and downregulation, respectively. Aged mice lacking Cables1 displayed abnormalities in peripheral blood cell counts accompanied by a significant reduction in HSC compartment, concomitant with an increased mobilization of progenitor cells. In addition, Cables1−/− mice displayed increased sensitivity to the chemotherapeutic agent 5-fluorouracil due to an abnormal microenvironment. Altogether, our findings uncover a key role for CABLES1 in HSC homeostasis and stress hematopoiesis. CABLES1 is expressed in immature hematopoietic progenitor cells and niche cells CABLES1 in an intrinsic negative cell-cycle regulator of hematopoietic progenitor cells CABLES1 regulates p21Cip/waf protein levels The abnormal stress responses of Cables1−/− HSC during aging are niche cell dependent
Collapse
|
6
|
Hernández-Ramírez LC, Gam R, Valdés N, Lodish MB, Pankratz N, Balsalobre A, Gauthier Y, Faucz FR, Trivellin G, Chittiboina P, Lane J, Kay DM, Dimopoulos A, Gaillard S, Neou M, Bertherat J, Assié G, Villa C, Mills JL, Drouin J, Stratakis CA. Loss-of-function mutations in the CABLES1 gene are a novel cause of Cushing's disease. Endocr Relat Cancer 2017; 24:379-392. [PMID: 28533356 PMCID: PMC5510591 DOI: 10.1530/erc-17-0131] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 05/18/2017] [Indexed: 12/11/2022]
Abstract
The CABLES1 cell cycle regulator participates in the adrenal-pituitary negative feedback, and its expression is reduced in corticotropinomas, pituitary tumors with a largely unexplained genetic basis. We investigated the presence of CABLES1 mutations/copy number variations (CNVs) and their associated clinical, histopathological and molecular features in patients with Cushing's disease (CD). Samples from 146 pediatric (118 germline DNA only/28 germline and tumor DNA) and 35 adult (tumor DNA) CD patients were screened for CABLES1 mutations. CNVs were assessed in 116 pediatric CD patients (87 germline DNA only/29 germline and tumor DNA). Four potentially pathogenic missense variants in CABLES1 were identified, two in young adults (c.532G > A, p.E178K and c.718C > T, p.L240F) and two in children (c.935G > A, p.G312D and c.1388A > G, and p.D463G) with CD; no CNVs were found. The four variants affected residues within or close to the predicted cyclin-dependent kinase-3 (CDK3)-binding region of the CABLES1 protein and impaired its ability to block cell growth in a mouse corticotropinoma cell line (AtT20/D16v-F2). The four patients had macroadenomas. We provide evidence for a role of CABLES1 as a novel pituitary tumor-predisposing gene. Its function might link two of the main molecular mechanisms altered in corticotropinomas: the cyclin-dependent kinase/cyclin group of cell cycle regulators and the epidermal growth factor receptor signaling pathway. Further studies are needed to assess the prevalence of CABLES1 mutations among patients with other types of pituitary adenomas and to elucidate the pituitary-specific functions of this gene.
Collapse
Affiliation(s)
- Laura C Hernández-Ramírez
- Section on Endocrinology and GeneticsEunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Ryhem Gam
- Laboratoire de Génétique MoléculaireInstitut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Nuria Valdés
- Section on Endocrinology and GeneticsEunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
- Service of Endocrinology and NutritionHospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain
| | - Maya B Lodish
- Section on Endocrinology and GeneticsEunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine and PathologyUniversity of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Aurelio Balsalobre
- Laboratoire de Génétique MoléculaireInstitut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Yves Gauthier
- Laboratoire de Génétique MoléculaireInstitut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Fabio R Faucz
- Section on Endocrinology and GeneticsEunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Giampaolo Trivellin
- Section on Endocrinology and GeneticsEunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Prashant Chittiboina
- Surgical Neurology BranchNational Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - John Lane
- Department of Laboratory Medicine and PathologyUniversity of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Denise M Kay
- Newborn Screening ProgramWadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Aggeliki Dimopoulos
- Division of Intramural Population Health ResearchEpidemiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Stephan Gaillard
- Institut CochinINSERM U1016, CNRS UMR8104, Université Paris Descartes, Paris, France
- Department of NeurosurgeryHôpital Foch, Suresnes, France
| | - Mario Neou
- Institut CochinINSERM U1016, CNRS UMR8104, Université Paris Descartes, Paris, France
| | - Jérôme Bertherat
- Institut CochinINSERM U1016, CNRS UMR8104, Université Paris Descartes, Paris, France
- Service d'EndocrinologieCochin Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Guillaume Assié
- Institut CochinINSERM U1016, CNRS UMR8104, Université Paris Descartes, Paris, France
- Service d'EndocrinologieCochin Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Chiara Villa
- Institut CochinINSERM U1016, CNRS UMR8104, Université Paris Descartes, Paris, France
- Department of Pathological Cytology and AnatomyHôpital Foch, Suresnes, France
- Department of EndocrinologyCHU de Liège, University of Liège, Liège, Belgium
| | - James L Mills
- Division of Intramural Population Health ResearchEpidemiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Jacques Drouin
- Laboratoire de Génétique MoléculaireInstitut de Recherches Cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Constantine A Stratakis
- Section on Endocrinology and GeneticsEunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
7
|
Liu J, Wang Y, Cui J, Sun M, Pu Z, Wang C, Du W, Liu X, Wu J, Hou J, Zhang S, Yu B. miR199a-3p regulates P53 by targeting CABLES1 in mouse cardiac c-kit + cells to promote proliferation and inhibit apoptosis through a negative feedback loop. Stem Cell Res Ther 2017; 8:127. [PMID: 28583208 PMCID: PMC5460483 DOI: 10.1186/s13287-017-0515-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 02/09/2017] [Accepted: 02/21/2017] [Indexed: 01/08/2023] Open
Abstract
Background MicroRNAs (miRNAs) have emerged as crucial factors that regulate proliferation and apoptosis of cardiac c-kit+ cells. Although much is known about their role in maintaining cardiac c-kit+ cell pluripotency, the mechanisms by which they affect cell fate decisions that are an essential part of the repair of heart failure remain poorly understood. Methods Cardiac c-kit+ cells were obtained from Balb/c mice and cultured in vitro. Lentiviral vectors of miR199a-3p, its corresponding anti-miRNA, or short hairpin RNA against Cables1 were transfected into cells. The proliferation of cardiac c-kit+ cells was evaluated using EdU and flow cytometry. Furthermore, we examined cell apoptosis by flow cytometry under treatment with 200nM angiotensin II for 48 h. The levels of miR199a-3p and Cables1 mRNA were measured by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot was performed to examine the expression of Cables1 and P53 proteins. Results We demonstrated a significantly decreased expression of miR199a-3p in heart failure samples compared with healthy donors. Meanwhile, we identified miR199a-3p as a proliferation- and apoptosis-associated regulator impacted through Cdk5 and Abl enzyme substrate 1 (CABLES1) targeting, and also attributed their repression to P53 protein expression. We further demonstrated that P53 induced miR199a-3p expression and, in turn, miR199-3p decreased P53 activity. Conclusion Collectively, our findings uncover one new mechanism by which P53 induced miR199a-3p expression and, in turn, miR199-3p decreased P53 activity. Therefore, miR199a-3p and P53 are coupled through CABLES1 and comprise a novel negative feedback loop that likely contributes to cardiac c-kit+ cell proliferation and apoptosis.
Collapse
Affiliation(s)
- Jingjin Liu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Yongshun Wang
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Jinjin Cui
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Meng Sun
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Zhongyue Pu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Chao Wang
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Wenjuan Du
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Xinxin Liu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China.,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China
| | - Jian Wu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Jingbo Hou
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Shuo Zhang
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China
| | - Bo Yu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150086, China. .,Key Laboratories of the Education Ministry for Myocardial Ischemia Mechanisms and Treatment, Harbin, Heilongjiang Province, China.
| |
Collapse
|
8
|
Pu Z, Wang Y, Liu X, Liu J, Cui J, Wang Y, Lv B, Yu B. Cables1 Inhibits Proliferation and Induces Senescence by Angiotensin II via a p21-Dependent Pathway in Human Umbilical Vein Endothelial Cells. J Vasc Res 2017; 54:13-21. [DOI: 10.1159/000452409] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 10/08/2016] [Indexed: 11/19/2022] Open
|
9
|
Huang JR, Tan GM, Li Y, Shi Z. The Emerging Role of Cables1 in Cancer and Other Diseases. Mol Pharmacol 2017; 92:240-245. [PMID: 28119482 DOI: 10.1124/mol.116.107730] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cdk5 and Abl enzyme substrate 1 (Cables1) is an adaptor protein that links cyclin-dependent kinase (Cdks) with nonreceptor tyrosine kinases and regulates the activity of Cdks by enhancing their Y15 phosphorylation. Emerging evidence also shows that Cables1 can interact with, for example, p53 family proteins, 14-3-3, and β-catenin, suggesting that Cables1 may be a signaling hub for the regulation of cell growth. Abnormal expression of Cables1 has been observed in multiple types of cancers and other diseases. In this review, we summarize the characteristics of Cables1 and highlight the molecular mechanisms through which Cables1 regulates the development of cancer and other diseases. Finally, we discuss future challenges in demonstrating the role and potential application of Cables1 in cancer and other diseases.
Collapse
Affiliation(s)
- Jia-Rong Huang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, People's Republic of China (J.-R.H., Z.S.); Department of Head & Neck Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China (G.-M.T.); and Department of Gastrointestinal Surgery & General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, People's Republic of China (Y.L.)
| | - Guang-Mou Tan
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, People's Republic of China (J.-R.H., Z.S.); Department of Head & Neck Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China (G.-M.T.); and Department of Gastrointestinal Surgery & General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, People's Republic of China (Y.L.)
| | - Yong Li
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, People's Republic of China (J.-R.H., Z.S.); Department of Head & Neck Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China (G.-M.T.); and Department of Gastrointestinal Surgery & General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, People's Republic of China (Y.L.)
| | - Zhi Shi
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, People's Republic of China (J.-R.H., Z.S.); Department of Head & Neck Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China (G.-M.T.); and Department of Gastrointestinal Surgery & General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, People's Republic of China (Y.L.)
| |
Collapse
|
10
|
Shi Z, Park HR, Du Y, Li Z, Cheng K, Sun SY, Li Z, Fu H, Khuri FR. Cables1 complex couples survival signaling to the cell death machinery. Cancer Res 2014; 75:147-158. [PMID: 25361894 DOI: 10.1158/0008-5472.can-14-0036] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cables1 is a candidate tumor suppressor that negatively regulates cell growth by inhibiting cyclin-dependent kinases. Cables1 expression is lost frequently in human cancer but little is known about its regulation. Here, we report that Cables1 levels are controlled by a phosphorylation and 14-3-3-dependent mechanism. Mutagenic analyses identified two residues, T44 and T150, that are specifically critical for 14-3-3 binding and that serve as substrates for phosphorylation by the cell survival kinase Akt, which by binding directly to Cables1 recruits 14-3-3 to the complex. In cells, Cables1 overexpression induced apoptosis and inhibited cell growth in part by stabilizing p21 and decreasing Cdk2 kinase activity. Ectopic expression of activated Akt (AKT1) prevented Cables1-induced apoptosis. Clinically, levels of phosphorylated Cables1 and phosphorylated Akt correlated with each other in human lung cancer specimens, consistent with pathophysiologic significance. Together, our results illuminated a dynamic regulatory system through which activated Akt and 14-3-3 work directly together to neutralize a potent tumor suppressor function of Cables1.
Collapse
Affiliation(s)
- Zhi Shi
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,Department of Pharmacology, Emory University, Atlanta, Georgia 30322
| | - Hae Ryon Park
- Department of Pharmacology, Emory University, Atlanta, Georgia 30322.,Department of Oral Pathology, School of Dentistry, Pusan National University, Pusan, South Korea
| | - Yuhong Du
- Department of Pharmacology, Emory University, Atlanta, Georgia 30322.,Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322
| | - Zijian Li
- Department of Pharmacology, Emory University, Atlanta, Georgia 30322.,Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Kejun Cheng
- Department of Pharmacology, Emory University, Atlanta, Georgia 30322.,Chemical Biology Center, Lishui Institute of Agricultural Sciences, Lishui, China
| | - Shi-Yong Sun
- Department of Hematology & Medical Oncology and Winship Cancer Institute, Emory University, Atlanta, Georgia 30322
| | - Zenggang Li
- Department of Pharmacology, Emory University, Atlanta, Georgia 30322
| | - Haian Fu
- Department of Pharmacology, Emory University, Atlanta, Georgia 30322.,Department of Hematology & Medical Oncology and Winship Cancer Institute, Emory University, Atlanta, Georgia 30322.,Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322
| | - Fadlo R Khuri
- Department of Hematology & Medical Oncology and Winship Cancer Institute, Emory University, Atlanta, Georgia 30322.,Emory Chemical Biology Discovery Center, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
11
|
Holocarboxylase synthetase interacts physically with nuclear receptor co-repressor, histone deacetylase 1 and a novel splicing variant of histone deacetylase 1 to repress repeats. Biochem J 2014; 461:477-86. [PMID: 24840043 DOI: 10.1042/bj20131208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
HLCS (holocarboxylase synthetase) is a nuclear protein that catalyses the binding of biotin to distinct lysine residues in chromatin proteins. HLCS-dependent epigenetic marks are over-represented in repressed genomic loci, particularly in repeats. Evidence is mounting that HLCS is a member of a multi-protein gene repression complex, which determines its localization in chromatin. In the present study we tested the hypothesis that HLCS interacts physically with N-CoR (nuclear receptor co-repressor) and HDAC1 (histone deacetylase 1), thereby contributing toward the removal of H3K9ac (Lys⁹-acetylated histone H3) gene activation marks and the repression of repeats. Physical interactions between HLCS and N-CoR, HDAC1 and a novel splicing variant of HDAC1 were confirmed by co-immunoprecipitation, limited proteolysis and split luciferase complementation assays. When HLCS was overexpressed, the abundance of H3K9ac marks decreased by 50% and 68% in LTRs (long terminal repeats) 15 and 22 respectively in HEK (human embryonic kidney)-293 cells compared with the controls. This loss of H3K9ac marks was linked with an 83% decrease in mRNA coding for LTRs. Similar patterns were seen in pericentromeric alpha satellite repeats in chromosomes 1 and 4. We conclude that interactions of HLCS with N-CoR and HDACs contribute towards the transcriptional repression of repeats, presumably increasing genome stability.
Collapse
|
12
|
Cables1 controls p21/Cip1 protein stability by antagonizing proteasome subunit alpha type 3. Oncogene 2014; 34:2538-45. [PMID: 24975575 PMCID: PMC4617825 DOI: 10.1038/onc.2014.171] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 12/15/2022]
Abstract
The cyclin-dependent kinase inhibitor 1A (CDKN1A), p21/Cip1, is a vital cell cycle regulator, dysregulation of which has been associated with a large number of human malignancies. One critical mechanism that controls p21 function is through its degradation, which allows the activation of its associated cell cycle promoting kinases, CDK2 and CDK4. Thus, delineating how p21 is stabilized and degraded will enhance our understanding of cell growth control and offer a basis for potential therapeutic interventions. Here, we report a novel regulatory mechanism that controls the dynamic status of p21 through its interaction with Cdk5 and Ablenzyme substrate 1 (Cables1). Cables1 has a proposed role as a tumor suppressor. We found that upregulation of Cables1 protein was correlated with increased half-life of p21 protein, which was attributed to Cables1/p21 complex formation and supported by their co-localization in the nucleus. Mechanistically, Cables1 interferes with the proteasome (Prosome, Macropain) subunit alpha type 3 (PSMA3) binding to p21 and protects p21 from PSMA3-mediated proteasomal degradation. Moreover, silencing of p21 partially reverses the ability of Cables1 to induce cell death and inhibit cell proliferation. In further support of a potential pathophysiological role of Cables1, the expression level of Cables1 is tightly associated with p21 in both cancer cell lines and human lung cancer patient tumor samples. Together, these results suggest Cables1 as a novel p21 regulator through maintaining p21 stability, and support the model that the tumor suppressive function of Cables1 occurs at least in part through enhancing the tumor suppressive activity of p21.
Collapse
|
13
|
Mizuno S, Tra DTH, Mizobuchi A, Iseki H, Mizuno-Iijima S, Kim JD, Ishida J, Matsuda Y, Kunita S, Fukamizu A, Sugiyama F, Yagami KI. Truncated Cables1 causes agenesis of the corpus callosum in mice. J Transl Med 2014; 94:321-30. [PMID: 24336072 DOI: 10.1038/labinvest.2013.146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/07/2013] [Accepted: 11/08/2013] [Indexed: 12/16/2022] Open
Abstract
Agenesis of the corpus callosum (ACC) is a congenital abnormality of the brain structure. More than 60 genes are known to be involved in corpus callosum development. However, the molecular mechanisms underlying ACC are not fully understood. Previously, we produced a novel transgenic mouse strain, TAS, carrying genes of the tetracycline-inducible expression system that are not involved in brain development, and inherited ACC was observed in the brains of all homozygous TAS mice. Although ACC was probably induced by transgene insertion mutation, the causative gene and the molecular mechanism of its pathogenesis remain unclear. Here, we first performed interphase three-color fluorescence in situ hybridization (FISH) analysis to determine the genomic insertion site. Transgenes were inserted into chromosome 18 ∼12.0 Mb from the centromere. Gene expression analysis and genomic PCR walking showed that the genomic region containing exon 4 of Cables1 was deleted by transgene insertion and the other exons of Cables1 were intact. The mutant allele was designated as Cables1(TAS). Interestingly, Cables1(TAS) mRNA consisted of exons 1-3 of Cables1 and part of the transgene that encoded a novel truncated Cables1 protein. Homozygous TAS mice exhibited mRNA expression of Cables1(TAS) in the fetal cerebrum, but not that of wild-type Cables1. To investigate whether a dominant negative effect of Cables1(TAS) or complete loss of function of Cables1 gives rise to ACC, we produced Cables1-null mutant mice. ACC was not observed in Cables1-null mutant mice, suggesting that a dominant negative effect of Cables1(TAS) impairs callosal formation. Moreover, ACC frequency in Cables1(+/TAS) mice was significantly lower than that in Cables1(-/TAS) mice, indicating that wild-type Cables1 interfered with the dominant negative effect of Cables1(TAS). This study indicated that truncated Cables1 causes ACC and wild-type Cables1 contributes to callosal formation.
Collapse
Affiliation(s)
- Seiya Mizuno
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Japan
| | - Dinh T H Tra
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Japan
| | - Atsushi Mizobuchi
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Japan
| | - Hiroyoshi Iseki
- 1] Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Japan [2] Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | | | - Jun-Dal Kim
- Life Science Center, Tsukuba Advanced Research Alliance, Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Junji Ishida
- Life Science Center, Tsukuba Advanced Research Alliance, Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Yoichi Matsuda
- Laboratory of Animal Genetics, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Satoshi Kunita
- Center for Experimental Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Akiyoshi Fukamizu
- Life Science Center, Tsukuba Advanced Research Alliance, Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Japan
| | - Ken-ichi Yagami
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
14
|
Reduced expression of BTBD10, an Akt activator, leads to motor neuron death. Cell Death Differ 2012; 19:1398-407. [PMID: 22388351 DOI: 10.1038/cdd.2012.19] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BTBD10, an Akt interactor, activates Akt by decreasing the protein phosphatase 2A-mediated dephosphorylation and inactivation of Akt. Overexpression of BTBD10 suppresses motor neuron death that is induced by a familial amyotrophic lateral sclerosis (ALS)-linked superoxide dismutase 1 (SOD1) mutant, G93A-SOD1 in vitro. In this study, we further investigated the BTBD10-mediated suppression of motor neuron death. We found that the small interfering RNA-mediated inhibition of BTBD10 expression led to the death of cultured motor neurons. In Caenorhabditis elegans (C. elegans), disruption of the btbd-10 gene caused not only loss of neurons, including both motor and touch-receptor neurons, but also a locomotion defect. In addition, we found that the expression of BTBD10 was generally decreased in the motor neurons from patients of sporadic ALS and transgenic mice overexpressing G93A-SOD1 (G93A-SOD1-transgenic mice). Collectively, these results suggest that the reduced expression of BTBD10 leads to motor neuron death both in vitro and in vivo.
Collapse
|
15
|
Groeneweg JW, White YAR, Kokel D, Peterson RT, Zukerberg LR, Berin I, Rueda BR, Wood AW. cables1 is required for embryonic neural development: molecular, cellular, and behavioral evidence from the zebrafish. Mol Reprod Dev 2010; 78:22-32. [PMID: 21268180 DOI: 10.1002/mrd.21263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 11/11/2010] [Indexed: 12/19/2022]
Abstract
In vitro studies have suggested that the Cables1 gene regulates epithelial cell proliferation, whereas other studies suggest a role in promoting neural differentiation. In efforts to clarify the functions of Cables1 in vivo, we conducted gain- and loss-of-function studies targeting its ortholog (cables1) in the zebrafish embryo. Similar to rodents, zebrafish cables1 mRNA expression is detected most robustly in embryonic neural tissues. Antisense knockdown of cables1 leads to increased numbers of apoptotic cells, particularly in brain tissue, in addition to a distinct behavioral phenotype, characterized by hyperactivity in response to stimulation. Apoptosis and the behavioral abnormality could be rescued by co-expression of a morpholino-resistant cables1 construct. Suppression of p53 expression in cables1 morphants partially rescued both apoptosis and the behavioral phenotype, suggesting that the phenotype of cables1 morphants is due in part to p53-dependent apoptosis. Alterations in the expression patterns of several neural transcription factors were observed in cables1 morphants during early neurulation, suggesting that cables1 is required for early neural differentiation. Ectopic overexpression of cables1 strongly disrupted embryonic morphogenesis, while overexpression of a cables1 mutant lacking the C-terminal cyclin box had little effect, suggesting functional importance of the cyclin box. Lastly, marked reductions in p35, but not Cdk5, were observed in cables1 morphants. Collectively, these data suggest that cables1 is important for neural differentiation during embryogenesis, in a mechanism that likely involves interactions with the Cdk5/p35 kinase pathway.
Collapse
Affiliation(s)
- Jolijn W Groeneweg
- Vincent Center for Reproductive Biology, MGH Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Cables1 protects p63 from proteasomal degradation to ensure deletion of cells after genotoxic stress. EMBO Rep 2010; 11:633-9. [PMID: 20559324 PMCID: PMC2920443 DOI: 10.1038/embor.2010.82] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 05/11/2010] [Accepted: 05/18/2010] [Indexed: 01/04/2023] Open
Abstract
Cables1 interacts with TAp63 to protect it from ubiquitin-mediated degradation after exposure to genotoxic stress. Oocytes lacking Cables1 accumulate less phosphorylated TAp63 in response to genotoxic stress, which enhances the survival of these cells. The p63 gene product regulates epithelial morphogenesis and female germline integrity. In this study, we show that cyclin-dependent kinase 5 and Abl enzyme substrate 1 (Cables1) interacts with the trans-activating (TA) p63α isoform to protect it from proteasomal degradation. Using the female germline of Cables1-null mice as an in vivo model, we demonstrate further that oocytes lacking Cables1 exhibit lower basal levels of TAp63α and reduced accumulation of phosphorylated TAp63α in response to genotoxic stress. This in turn enhances the survival of these cells after ionizing radiation exposure. Thus, Cables1 modulates p63 protein stability and function during genotoxic stress.
Collapse
|
17
|
Torii K, Nishizawa K, Kawasaki A, Yamashita Y, Katada M, Ito M, Nishimoto I, Terashita K, Aiso S, Matsuoka M. Anti-apoptotic action of Wnt5a in dermal fibroblasts is mediated by the PKA signaling pathways. Cell Signal 2008; 20:1256-66. [DOI: 10.1016/j.cellsig.2008.02.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Revised: 02/18/2008] [Accepted: 02/18/2008] [Indexed: 01/22/2023]
|
18
|
Park DY, Sakamoto H, Kirley SD, Ogino S, Kawasaki T, Kwon E, Mino-Kenudson M, Lauwers GY, Chung DC, Rueda BR, Zukerberg LR. The Cables gene on chromosome 18q is silenced by promoter hypermethylation and allelic loss in human colorectal cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 171:1509-19. [PMID: 17982127 DOI: 10.2353/ajpath.2007.070331] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cables is a cyclin-dependent kinase-binding nuclear protein that maps to chromosome 18q11-12. Here, we assessed Cables expression in 160 colorectal cancers (CRCs), its role in colon cancer cell growth, and the potential mechanisms of Cables inactivation. Expression levels, promoter methylation, and mutational status of Cables were investigated in colon cancer cell lines and primary colon tumors. Chromosome 18q loss of heterozygosity (LOH) was evaluated with multiple polymorphic markers. Cables inhibited cellular proliferation and colony formation in colon cancer cell lines. Cables expression was reduced in 65% of primary CRCs. No mutations were detected in 10 exons of Cables in 20 primary colon tumors. Cables promoter was methylated in cell lines with decreased Cables expression and vice versa. 5-Aza-2'-deoxycytidine resulted in increased Cables expression in methylated cell lines. There was a significant correlation between promoter methylation and Cables gene expression in primary colon tumors. Sixty-five percent of primary colon tumors demonstrated chromosome 18q LOH. LOH involving the Cables region was observed in 35% of cases, including those in which more distal portions of chromosome 18q were retained, and Cables expression was decreased in all such cases. Loss of Cables expression in 65% of CRCs suggests that it is a common event in colonic carcinogenesis, with promoter methylation and LOH appearing to be important mechanisms of Cables gene inactivation.
Collapse
Affiliation(s)
- Do Youn Park
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kurita M, Suzuki H, Kawano Y, Aiso S, Matsuoka M. CR/periphilin is a transcriptional co-repressor involved in cell cycle progression. Biochem Biophys Res Commun 2007; 364:930-6. [DOI: 10.1016/j.bbrc.2007.10.090] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2007] [Accepted: 10/16/2007] [Indexed: 11/30/2022]
|
20
|
Sasabe J, Chiba T, Yamada M, Okamoto K, Nishimoto I, Matsuoka M, Aiso S. D-serine is a key determinant of glutamate toxicity in amyotrophic lateral sclerosis. EMBO J 2007; 26:4149-59. [PMID: 17762863 PMCID: PMC2230675 DOI: 10.1038/sj.emboj.7601840] [Citation(s) in RCA: 211] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Accepted: 07/30/2007] [Indexed: 11/09/2022] Open
Abstract
Excitotoxicity has been implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS). More recently, glial involvement has been shown to be essential for ALS-related motoneuronal death. Here, we identified an N-methyl-D-aspartate (NMDA) receptor co-agonist, D-serine (D-Ser), as a glia-derived enhancer of glutamate (Glu) toxicity to ALS motoneurons. Cell death assay indicated that primary spinal cord neurons from ALS mice were more vulnerable to NMDA toxicity than those from control mice, in a D-Ser-dependent manner. Levels of D-Ser and its producing enzyme, serine racemase, in spinal cords of ALS mice were progressively elevated, dominantly in glia, with disease progression. In vitro, expression of serine racemase was induced not only by an extracellular pro-inflammatory factor, but also by transiently expressed G93A-superoxide dismutase1 in microglial cells. Furthermore, increases of D-Ser levels were also observed in spinal cords of both familial and sporadic ALS patients. Collectively, Glu toxicity enhanced by D-Ser overproduced in glia is proposed as a novel mechanism underlying ALS motoneuronal death, and this mechanism may be regarded as a potential therapeutic target for ALS.
Collapse
Affiliation(s)
- Jumpei Sasabe
- Department of Anatomy, KEIO University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Cell Biology and Neuroscience, KEIO University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Tomohiro Chiba
- Department of Anatomy, KEIO University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Cell Biology and Neuroscience, KEIO University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Marina Yamada
- Department of Anatomy, KEIO University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Cell Biology and Neuroscience, KEIO University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Koichi Okamoto
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | | | - Masaaki Matsuoka
- Department of Anatomy, KEIO University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Cell Biology and Neuroscience, KEIO University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Cell Biology and Neuroscience/Anatomy, KEIO University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan. Tel.: +81 3 5363 8427; Fax: +81 3 5363 8428; E-mail:
| | - Sadakazu Aiso
- Department of Anatomy, KEIO University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Cell Biology and Neuroscience, KEIO University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
21
|
Tomasini R, Seux M, Nowak J, Bontemps C, Carrier A, Dagorn JC, Pébusque MJ, Iovanna JL, Dusetti NJ. TP53INP1 is a novel p73 target gene that induces cell cycle arrest and cell death by modulating p73 transcriptional activity. Oncogene 2006; 24:8093-104. [PMID: 16044147 DOI: 10.1038/sj.onc.1208951] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
TP53INP1 is an alternatively spliced gene encoding two nuclear protein isoforms (TP53INP1alpha and TP53INP1beta), whose transcription is activated by p53. When overexpressed, both isoforms induce cell cycle arrest in G1 and enhance p53-mediated apoptosis. TP53INP1s also interact with the p53 gene and regulate p53 transcriptional activity. We report here that TP53INP1 expression is induced during experimental acute pancreatitis in p53-/- mice and in cisplatin-treated p53-/- mouse embryo fibroblasts (MEFs). We demonstrate that ectopic expression of p73, a p53 homologue, leads to TP53INP1 induction in p53-deficient cells. In turn, TP53INP1s alters the transactivation capacity of p73 on several p53-target genes, including TP53INP1 itself, demonstrating a functional association between p73 and TP53INP1s. Also, when overexpressed in p53-deficient cells, TP53INP1s inhibit cell growth and promote cell death as assessed by cell cycle analysis and colony formation assays. Finally, we show that TP53INP1s potentiate the capacity of p73 to inhibit cell growth, that effect being prevented when the p53 mutant R175H is expressed or when p73 expression is blocked by a siRNA. These results suggest that TP53INP1s are functionally associated with p73 to regulate cell cycle progression and apoptosis, independently from p53.
Collapse
Affiliation(s)
- Richard Tomasini
- INSERM U624, Stress Cellulaire, IFR 137-Institut de Cancérologie et Immunologie de Marseille, Université de la Méditerranée, Marseille, France
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Suzuki H, Kurita M, Mizumoto K, Moriyama M, Aiso S, Nishimoto I, Matsuoka M. The ARF tumor suppressor inhibits BCL6-mediated transcriptional repression. Biochem Biophys Res Commun 2005; 326:242-8. [PMID: 15567177 DOI: 10.1016/j.bbrc.2004.11.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2004] [Indexed: 10/26/2022]
Abstract
The ARF tumor suppressor gene antagonizes generation of various tumors. ARF-mediated tumor suppression occurs in a p53-independent manner as well as in a p53-dependent manner. We here demonstrate that BCL6 is a target of the ARF tumor suppressor. Either mouse p19(ARF) or human p14(ARF) binds to BCL6 and downregulates BCL6-induced transcriptional repression. ARF-mediated downregulation of the BCL6 activity may account in part for ARF-mediated tumor suppression.
Collapse
Affiliation(s)
- Hiroaki Suzuki
- Department of Pharmacology, KEIO University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo160-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
23
|
Kurita M, Suzuki H, Masai H, Mizumoto K, Ogata E, Nishimoto I, Aiso S, Matsuoka M. Overexpression of CR/periphilin downregulates Cdc7 expression and induces S-phase arrest. Biochem Biophys Res Commun 2004; 324:554-61. [PMID: 15474462 DOI: 10.1016/j.bbrc.2004.09.083] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2004] [Indexed: 10/26/2022]
Abstract
Cdc7 expression repressor (CR)/periphilin has been originally cloned as an interactor with periplakin, a precursor of the cornified cell envelope, and suggested to constitute a new type of nuclear matrix. We here show that CR/periphilin is a ubiquitously expressed nuclear protein with speckled distribution. Overexpression of CR/periphilin induces S-phase arrest. Analysis of expression of regulators involved in DNA replication has revealed that both mRNA and protein expression of Cdc7, a regulator of the initiation and continuation of DNA replication, are markedly downregulated by overexpression of CR/periphilin. However, co-expression of Cdc7 only marginally rescues S-phase arrest induced by CR, indicating that CR retards S-phase progression by modifying expression of some genes including Cdc7, which are involved in progression of DNA replication or coordination of DNA replication and S-phase progression.
Collapse
Affiliation(s)
- Megumi Kurita
- Department of Pharmacology, KEIO University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Dong Q, Kirley S, Rueda B, Zhao C, Zukerberg L, Oliva E. Loss of cables, a novel gene on chromosome 18q, in ovarian cancer. Mod Pathol 2004; 16:863-8. [PMID: 13679449 DOI: 10.1097/01.mp.0000084434.88269.0a] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cables, a cyclin-dependent kinase (cdk) interacting protein, has recently been identified and mapped to human chromosome 18q11. Cables appears to be primarily involved in cell cycle regulation and cell proliferation. Overexpression of Cables in Hela and other cell lines inhibits cell proliferation and tumor formation. We hypothesize that loss of Cables expression is associated with ovarian cancer. To test our hypothesis, we examined Cables expression in the four most common subtypes of ovarian carcinomas: serous, endometrioid, mucinous, and clear cell. In addition, mucinous and serous borderline tumors were also included. Loss of Cables expression was observed at high frequency in ovarian serous (11 of 14 cases, 79%) and endometrioid (5 of 10 cases, 50%) carcinomas. In contrast, strong Cables staining was detected in all clear cell carcinomas (10 cases) and mucinous tumors (5 carcinomas and 5 borderline tumors). The majority of serous borderline tumors (11 of 14 cases, 79%) showed positive Cables staining, with the rest showing focal loss of Cables expression. Furthermore, RT-PCR revealed the lack of Cables mRNA in a human ovarian cancer xenograft. No correlation was noted between loss of Cables and histologic grade, tumor stage, and survival. In conclusion, our results indicate that loss of Cables is common in ovarian serous and endometrioid carcinomas and imply that Cables may be involved in the pathogenesis of these two types of ovarian carcinomas.
Collapse
Affiliation(s)
- Qun Dong
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
25
|
Zukerberg LR, DeBernardo RL, Kirley SD, D'Apuzzo M, Lynch MP, Littell RD, Duska LR, Boring L, Rueda BR. Loss of cables, a cyclin-dependent kinase regulatory protein, is associated with the development of endometrial hyperplasia and endometrial cancer. Cancer Res 2004; 64:202-8. [PMID: 14729625 DOI: 10.1158/0008-5472.can-03-2833] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endometrial cancer is the most common gynecological cancer in Western industrialized countries. Cables, a cyclin-dependent kinase binding protein, plays a role in proliferation and/or differentiation. Cables mutant mice are viable, but develop endometrial hyperplasia and carcinoma in situ at a young age. Exposure to chronic low levels of estrogen results in development of endometrial cancer, similar to that observed in the postmenopausal female. In vitro and in vivo studies demonstrate that levels of Cables mRNA in benign human endometrial epithelium are up-regulated by progesterone and down-regulated by estrogen. Furthermore, nuclear immunostaining for Cables is lost in a high percentage of cases of human endometrial hyperplasia and adenocarcinoma, which are likely the product of unopposed estrogen. The loss of Cables immunostaining in the human endometrial cancer samples correlates with a marked decrease in Cables mRNA. Ectopic expression of Cables in human endometrial cells dramatically slows cell proliferation. Collectively, these data provide evidence that Cables is hormonally regulated and is involved in regulating endometrial cell proliferation. In addition, loss or suppression of Cables may be an early step in the development of endometrial cancer.
Collapse
Affiliation(s)
- Lawrence R Zukerberg
- Departments of Pathology and Urology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Matsuoka M, Sudo H, Tsuji K, Sato H, Kurita M, Suzuki H, Nishimoto I, Ogata E. ik3-2, a relative to ik3-1/Cables, is involved in both p53-mediated and p53-independent apoptotic pathways. Biochem Biophys Res Commun 2004; 312:520-9. [PMID: 14637168 DOI: 10.1016/j.bbrc.2003.10.142] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ik3-2 is a close relative to ik3-1/Cables, an associator with cdk3 and cdk5. ik3-1/Cables has been identified to be a candidate tumor suppressor for colon and head/neck cancers. In agreement, it has been pointed out that ik3-1/Cables is a regulator for both p53- and p73-induced apoptosis [J. Biol. Chem. 277 (2002) 2951] although ectopic expression of ik3-1/Cables does not induce apoptosis. Here we show that adenovirus-mediated overexpression of ik3-2 results in apoptosis of p53-intact U2OS cells. ik3-2 binds to p53 in vivo and ectopic coexpression of ik3-2 enhances apoptosis induced by adenovirus-mediated expression of p53. Furthermore, ectopic expression of ik3-2 results in apoptosis of primary p53/Mdm2- and p53/ARF-null mouse embryo fibroblasts, indicating that ik3-2-induced apoptosis is partially p53-independent. Both the highly conserved C-terminal cyclin box-homologous domain (ik3-2-C) and the N-terminal region consisting of 70 amino acids (ik3-2-N) are responsible for ik3-2-mediated enhancement of p53-induced apoptosis. In contrast, ik3-2-induced p53-independent apoptosis is mediated through ik3-2-N. We thus identified ik3-2 as a proapoptotic factor involved in both p53-mediated and p53-independent apoptotic pathways.
Collapse
Affiliation(s)
- Masaaki Matsuoka
- Department of Pharmacology, University of KEIO School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Suzuki H, Kurita M, Mizumoto K, Nishimoto I, Ogata E, Matsuoka M. p19ARF-induced p53-independent apoptosis largely occurs through BAX. Biochem Biophys Res Commun 2003; 312:1273-7. [PMID: 14652011 DOI: 10.1016/j.bbrc.2003.11.071] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Combined disruption of the ARF gene and the p53 gene causes mouse predisposition to tumors of a wider variety and at a higher frequency than disruption of the p53 gene, indicating that the ARF gene has p53-independent anti-tumor function in addition to p53-dependent function. Coincidentally with this notion, ectopic expression of the p19(ARF) induces apoptosis for wild-type mouse embryo fibroblasts which have been immortalized by introduction of the SV40 virus genome (SV40-MEFs). The protein expression levels of p53, p21(Cip1), and Bax were not upregulated by ectopic expression of p19(ARF) in SV40-MEFs, indicating that expression of p19(ARF) induced apoptosis through p53-independent pathways in this system. Ectopic expression of p19(ARF) induced prominent apoptosis even in SV40-Bak-/-MEFs. In contrast, expression of p19(ARF) induced only a very low grade of apoptosis in Bax-/- or Bax-/-/Bak-/-SV40-MEFs. Remarkable attenuation of p19(ARF)-induced apoptosis by disruption of the Bax gene thus leads to the conclusion that Bax plays a major role in p53-independent apoptosis induced by p19(ARF).
Collapse
Affiliation(s)
- Hiroaki Suzuki
- Department of Pharmacology, KEIO University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
28
|
Gaiddon C, Lokshin M, Gross I, Levasseur D, Taya Y, Loeffler JP, Prives C. Cyclin-dependent kinases phosphorylate p73 at threonine 86 in a cell cycle-dependent manner and negatively regulate p73. J Biol Chem 2003; 278:27421-31. [PMID: 12676926 DOI: 10.1074/jbc.m300251200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
p73 transcription factors are members of the p53 family and participate in developmental processes and DNA damage response. p73 expression was shown to be regulated during the cell cycle, suggesting that p73 might play a role in cell growth and might be a target for cyclin-dependent kinases. Consistent with this hypothesis, we discovered that p73 interacts physically with various cyclins (A, B, D, and E). Furthermore, cyclin A/CDK1/2, cyclin B/CDK1/2, and cyclin E/CDK2 complexes can phosphorylate multiple p73 isoforms in vitro at threonine 86. A specific antibody directed against phosphorylated Thr86 showed that this site is phosphorylated in vivo and that such phosphorylation is regulated in a cell cycle-dependent manner. Thr86 phosphorylation is induced during S phase and is maximal in the G2/M phase. Accordingly inhibitors of cell growth, such as p16 and serum starvation, reduce Thr86 phosphorylation. Finally, we found that cyclin-dependent kinase (CDK)-dependent Thr86 phosphorylation represses the ability of p73 to induce endogenous p21 expression. Our results demonstrate that p73 proteins are targets of CDK complexes and that phosphorylation on Thr86 by CDKs regulates p73 functions.
Collapse
Affiliation(s)
- Christian Gaiddon
- Equipe d'Accecil Signalisations Moléculaires et Neurodégénéréscence, Université Louis Pasteur, Strasbourg 67000, France.
| | | | | | | | | | | | | |
Collapse
|
29
|
Matsuoka M, Kurita M, Sudo H, Mizumoto K, Nishimoto I, Ogata E. Multiple domains of the mouse p19ARF tumor suppressor are involved in p53-independent apoptosis. Biochem Biophys Res Commun 2003; 301:1000-10. [PMID: 12589812 DOI: 10.1016/s0006-291x(03)00080-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The ARF (p19ARF for the mouse ARF consisting of 169 amino acids and p14ARF for the human ARF consisting of 132 amino acids) genes upregulate p53 activities to induce cell cycle arrest and sensitize cells to apoptosis by inhibiting Mdm2 activity. p53-independent apoptosis also is induced by ectopic expression of p19ARF. We constructed various deletion mutants of p19ARF with a cre/loxP-regulated adenoviral vector to determine the regions of p19ARF which are responsible for p53-independent apoptosis. Ectopic expression of the C-terminal region (named C40) of p19ARF whose primary sequence is unique to the rodent ARF induced prominent apoptosis in p53-deficient mouse embryo fibroblasts. Relatively low-grade but significant apoptosis also was induced in p53-deficient mouse embryo fibroblasts by ectopic expression of p19ARF1-129, a p19ARF deletion mutant deficient in the C40 region. In contrast, ectopic expression of the wild-type p14ARF did not induce significant apoptosis in human cells. Taken together, we concluded that p53-independent apoptosis was mediated through multiple regions of the mouse ARF including C40, and the ability of the ARF gene to mediate p53-independent apoptosis has been not well conserved during mammalian evolution.
Collapse
Affiliation(s)
- Masaaki Matsuoka
- Department of Pharmacology, KEIO University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
30
|
Tsuji K, Mizumoto K, Sudo H, Kouyama K, Ogata E, Matsuoka M. p53-independent apoptosis is induced by the p19ARF tumor suppressor. Biochem Biophys Res Commun 2002; 295:621-9. [PMID: 12099684 DOI: 10.1016/s0006-291x(02)00723-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
p19(ARF) is a potent tumor suppressor. By inactivating Mdm2, p19(ARF) upregulates p53 activities to induce cell cycle arrest and sensitize cells to apoptosis in the presence of collateral signals. It has also been demonstrated that cell cycle arrest is induced by overexpressed p19(ARF) in p53-deficient mouse embryonic fibroblasts, only in the absence of the Mdm2 gene. Here, we show that apoptosis can be induced without additional apoptosis signals by expression of p19(ARF) using an adenovirus-mediated expression system in p53-intact cell lines as well as p53-deficient cell lines. Also, in primary mouse embryonic fibroblasts (MEFs) lacking p53/ARF, p53-independent apoptosis is induced irrespective of Mdm2 status by expression of p19(ARF). In agreement, p19(ARF)-mediated apoptosis in U2OS cells, but not in Saos2 cells, was attenuated by coexpression of Mdm2. We thus conclude that there is a p53-independent pathway for p19(ARF)-induced apoptosis that is insensitive to inhibition by Mdm2.
Collapse
Affiliation(s)
- Keitaro Tsuji
- Department of Pharmacology, KEIO University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | |
Collapse
|