1
|
Baldazzi C, Bandini L, Robustelli V, Patuelli A, Venturi C, Grassi A, Marzocchi G, Ielpo A, Solli V, Bochicchio MT, Paolini S, Sartor C, Zingarelli F, Curti A, Ottaviani E, Testoni N. Emergence and Cytogenetic Clonal Evolution of Chromosome 7 Abnormalities in Myeloid Malignancies: Investigating the Role of Telomere Dysfunction. Int J Mol Sci 2025; 26:1162. [PMID: 39940930 PMCID: PMC11817968 DOI: 10.3390/ijms26031162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/16/2025] Open
Abstract
Monosomy 7 and deletion 7q are common chromosomal abnormalities in myeloid malignancies, and they are associated with a poor prognosis. The mechanism underlying their acquisition remains elusive. We identified a cohort of 24 patients exhibiting clones with different chromosome 7 abnormalities, such as deletion 7q, unstable derivatives (ring chromosomes or 'naked' centromeres), and monosomy 7. We designated this group as having cytogenetic clonal evolution of chromosome 7 abnormalities (CCE7). In some cases, CCE7 correlated with disease progression, suggesting that deletions or other derivatives involving the q-arm of chromosome 7 may arise early in the disease course. These abnormalities may be transient but can potentially evolve into monosomy 7. Within the CCE7 group, telomere loss or shortening may contribute to chromosomal instability and the emergence of unstable derivatives, as the chromosome 7 derivatives displayed loss or rearrangement of subtelomeric regions. Moreover, we identified variants in genes implicated in telomere biology disorders and observed specific genetic mutation profiles associated with different chromosome 7 abnormalities. These findings shed light on a potential mechanism leading to monosomy 7 through the evolution of chromosome 7q abnormalities. Identifying patients at risk of developing monosomy 7, based on the presence of unstable derivatives with telomere loss or a specific mutation profile, could potentially enhance patient management and guide the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Carmen Baldazzi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy
| | - Lorenza Bandini
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy
| | - Valentina Robustelli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy
| | - Agnese Patuelli
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy
| | - Claudia Venturi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy
| | - Alessandra Grassi
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy
| | - Giulia Marzocchi
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy
| | - Angela Ielpo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy
| | - Vincenza Solli
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy
| | - Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Stefania Paolini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy
| | - Chiara Sartor
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy
| | - Federico Zingarelli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy
| | - Antonio Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy
| | - Emanuela Ottaviani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy
| | - Nicoletta Testoni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli”, 40138 Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, 40138 Bologna, Italy
| |
Collapse
|
2
|
Li J, Wang X, Chen Y, Sun X, Fu L, Xin Q, Zhang H, Qin B, Sun N, Li Y, Xu Y, Yang H, Huo D, Dong Y, Wang S, Zhao M, Lin Q, Wang F, Yue B, Gao Y, Jiang Y, Guo R. Exosome-Mediated Lectin Pathway and Resistin-MIF-AA Metabolism Axis Drive Immune Dysfunction in Immune Thrombocytopenia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412378. [PMID: 39792656 DOI: 10.1002/advs.202412378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/01/2024] [Indexed: 01/12/2025]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disorder characterized by reduced platelet levels and heightened susceptibility to bleeding resulting from augmented autologous platelet destruction and diminished thrombopoiesis. Although antibody-mediated autoimmune reactions are widely recognized as primary factors, the precise etiological agents that trigger ITP remain unidentified. The pathogenesis of ITP remains unclear owing to the absence of comprehensive high-throughput data, except for the belated emergence of autoreactive antibodies. In this study, using flow cytometry (FCM), proteomics, and single-cell RNA sequencing of samples from patients with ITP, it is shown that exosome-mediated lectin complement pathway is involved in the pathogenesis of ITP, which triggers and enlarges the complement activation cascade without effective regulation because of downregulated CD55. The activated complement system enhances the immune response and resistin and further Macrophage Migration Inhibitory Factor (MIF) triggers several proinflammatory signaling pathways, which contribute to the survival of hyperactivated immune cells and dysfunctional arachidonic acid (AA) metabolism. The resistin and MIF are also identified as potential contributors to resistance to glucocorticoid therapy. Taken together, the findings indicate that the lectin pathway of the complement system, resistin, MIF, and AA metabolism may serve as promising targets for ITP treatment, offering novel perspectives on potential therapeutic interventions.
Collapse
Affiliation(s)
- Jin Li
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Xiaoqian Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yaoyao Chen
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Xianlei Sun
- Basic Medical Research Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Liyan Fu
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Qingxuan Xin
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Huilin Zhang
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Bo Qin
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Nannan Sun
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yingmei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yan Xu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Hui Yang
- Translational Cancer Research Center, Peking University First Hospital, Beijing, 100034, China
| | - Dawei Huo
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Zhejiang University, Hang Zhou, Zhejiang, 311100, China
| | - Yong Dong
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Shuya Wang
- Department of Blood Transfusion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Mengyun Zhao
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, 450000, China
| | - Quande Lin
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, 450000, China
| | - Fang Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Baohong Yue
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, 450046, China
| | - Yanxia Gao
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yong Jiang
- Henan International Joint Laboratory of Infection and Immunity, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Henan Key Laboratory of Critical Care Medicine, Department of Emergency Medicine, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, 450001, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, China
| | - Rongqun Guo
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| |
Collapse
|
3
|
Fajac A, Simeonova I, Leemput J, Gabriel M, Morin A, Lejour V, Hamon A, Rakotopare J, Vaysse-Zinkhöfer W, Eldawra E, Pinskaya M, Morillon A, Bourdon JC, Bardot B, Toledo F. Mutant mice lacking alternatively spliced p53 isoforms unveil Ackr4 as a male-specific prognostic factor in Myc-driven B-cell lymphomas. eLife 2024; 13:RP92774. [PMID: 39298333 PMCID: PMC11412721 DOI: 10.7554/elife.92774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024] Open
Abstract
The Trp53 gene encodes several isoforms of elusive biological significance. Here, we show that mice lacking the Trp53 alternatively spliced (AS) exon, thereby expressing the canonical p53 protein but not isoforms with the AS C-terminus, have unexpectedly lost a male-specific protection against Myc-induced B-cell lymphomas. Lymphomagenesis was delayed in Trp53+/+Eμ-Myc males compared to Trp53ΔAS/ΔAS Eμ-Myc males, but also compared to Trp53+/+Eμ-Myc and Trp53ΔAS/ΔAS Eμ-Myc females. Pre-tumoral splenic cells from Trp53+/+Eμ-Myc males exhibited a higher expression of Ackr4, encoding an atypical chemokine receptor with tumor suppressive effects. We identified Ackr4 as a p53 target gene whose p53-mediated transactivation is inhibited by estrogens, and as a male-specific factor of good prognosis relevant for murine Eμ-Myc-induced and human Burkitt lymphomas. Furthermore, the knockout of ACKR4 increased the chemokine-guided migration of Burkitt lymphoma cells. These data demonstrate the functional relevance of alternatively spliced p53 isoforms and reveal sex disparities in Myc-driven lymphomagenesis.
Collapse
Affiliation(s)
- Anne Fajac
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Iva Simeonova
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Julia Leemput
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Marc Gabriel
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
- Non Coding RNA, Epigenetic and Genome Fluidity, Institut CurieParisFrance
| | - Aurélie Morin
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Vincent Lejour
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Annaïg Hamon
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Jeanne Rakotopare
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Wilhelm Vaysse-Zinkhöfer
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Eliana Eldawra
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Marina Pinskaya
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
- Non Coding RNA, Epigenetic and Genome Fluidity, Institut CurieParisFrance
| | - Antonin Morillon
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
- School of Medicine, Ninewells Hospital, University of DundeeDundeeUnited Kingdom
| | | | - Boris Bardot
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Franck Toledo
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| |
Collapse
|
4
|
Rakotopare J, Toledo F. p53 in the Molecular Circuitry of Bone Marrow Failure Syndromes. Int J Mol Sci 2023; 24:14940. [PMID: 37834388 PMCID: PMC10573108 DOI: 10.3390/ijms241914940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023] Open
Abstract
Mice with a constitutive increase in p53 activity exhibited features of dyskeratosis congenita (DC), a bone marrow failure syndrome (BMFS) caused by defective telomere maintenance. Further studies confirmed, in humans and mice, that germline mutations affecting TP53 or its regulator MDM4 may cause short telomeres and alter hematopoiesis, but also revealed features of Diamond-Blackfan anemia (DBA) or Fanconi anemia (FA), two BMFSs, respectively, caused by defects in ribosomal function or DNA repair. p53 downregulates several genes mutated in DC, either by binding to promoter sequences (DKC1) or indirectly via the DREAM repressor complex (RTEL1, DCLRE1B), and the p53-DREAM pathway represses 22 additional telomere-related genes. Interestingly, mutations in any DC-causal gene will cause telomere dysfunction and subsequent p53 activation to further promote the repression of p53-DREAM targets. Similarly, ribosomal dysfunction and DNA lesions cause p53 activation, and p53-DREAM targets include the DBA-causal gene TSR2, at least 9 FA-causal genes, and 38 other genes affecting ribosomes or the FA pathway. Furthermore, patients with BMFSs may exhibit brain abnormalities, and p53-DREAM represses 16 genes mutated in microcephaly or cerebellar hypoplasia. In sum, positive feedback loops and the repertoire of p53-DREAM targets likely contribute to partial phenotypic overlaps between BMFSs of distinct molecular origins.
Collapse
Affiliation(s)
- Jeanne Rakotopare
- Genetics of Tumor Suppression, Institut Curie, CEDEX 05, 75248 Paris, France;
- CNRS UMR3244, 75005 Paris, France
- Faculty of Science and Engineering, Sorbonne University, 75005 Paris, France
- Institut Curie, PSL Research University, 75005 Paris, France
| | - Franck Toledo
- Genetics of Tumor Suppression, Institut Curie, CEDEX 05, 75248 Paris, France;
- CNRS UMR3244, 75005 Paris, France
- Faculty of Science and Engineering, Sorbonne University, 75005 Paris, France
- Institut Curie, PSL Research University, 75005 Paris, France
| |
Collapse
|
5
|
Rakotopare J, Lejour V, Duval C, Eldawra E, Escoffier H, Toledo F. A systematic approach identifies p53-DREAM pathway target genes associated with blood or brain abnormalities. Dis Model Mech 2023; 16:dmm050376. [PMID: 37661832 PMCID: PMC10581385 DOI: 10.1242/dmm.050376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023] Open
Abstract
p53 (encoded by Trp53) is a tumor suppressor, but mouse models have revealed that increased p53 activity may cause bone marrow failure, likely through dimerization partner, RB-like, E2F4/E2F5 and MuvB (DREAM) complex-mediated gene repression. Here, we designed a systematic approach to identify p53-DREAM pathway targets, the repression of which might contribute to abnormal hematopoiesis. We used Gene Ontology analysis to study transcriptomic changes associated with bone marrow cell differentiation, then chromatin immunoprecipitation-sequencing (ChIP-seq) data to identify DREAM-bound promoters. We next created positional frequency matrices to identify evolutionary conserved sequence elements potentially bound by DREAM. The same approach was developed to find p53-DREAM targets associated with brain abnormalities, also observed in mice with increased p53 activity. Putative DREAM-binding sites were found for 151 candidate target genes, of which 106 are mutated in a blood or brain genetic disorder. Twenty-one DREAM-binding sites were tested and found to impact gene expression in luciferase assays, to notably regulate genes mutated in dyskeratosis congenita (Rtel1), Fanconi anemia (Fanca), Diamond-Blackfan anemia (Tsr2), primary microcephaly [Casc5 (or Knl1), Ncaph and Wdr62] and pontocerebellar hypoplasia (Toe1). These results provide clues on the role of the p53-DREAM pathway in regulating hematopoiesis and brain development, with implications for tumorigenesis.
Collapse
Affiliation(s)
- Jeanne Rakotopare
- Genetics of Tumor Suppression, Institut Curie, Paris 75248 Cedex 05, France
- CNRS UMR3244, Paris 75005, France
- Sorbonne University, Paris 75005, France
- PSL Research University, Paris 75005, France
| | - Vincent Lejour
- Genetics of Tumor Suppression, Institut Curie, Paris 75248 Cedex 05, France
- CNRS UMR3244, Paris 75005, France
- Sorbonne University, Paris 75005, France
- PSL Research University, Paris 75005, France
| | - Carla Duval
- Genetics of Tumor Suppression, Institut Curie, Paris 75248 Cedex 05, France
- CNRS UMR3244, Paris 75005, France
- Sorbonne University, Paris 75005, France
- PSL Research University, Paris 75005, France
| | - Eliana Eldawra
- Genetics of Tumor Suppression, Institut Curie, Paris 75248 Cedex 05, France
- CNRS UMR3244, Paris 75005, France
- Sorbonne University, Paris 75005, France
- PSL Research University, Paris 75005, France
| | | | - Franck Toledo
- Genetics of Tumor Suppression, Institut Curie, Paris 75248 Cedex 05, France
- CNRS UMR3244, Paris 75005, France
- Sorbonne University, Paris 75005, France
- PSL Research University, Paris 75005, France
| |
Collapse
|
6
|
Zhang L, Zhang J, Xuan X, Wu D, Yu J, Wang P, Yang X, Zhang J, Gan W, He M, Liu XM, Zhou J, Wang D, Gu W, Li D. A p53/LINC00324 positive feedback loop suppresses tumor growth by counteracting SET-mediated transcriptional repression. Cell Rep 2023; 42:112833. [PMID: 37480565 DOI: 10.1016/j.celrep.2023.112833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 05/26/2023] [Accepted: 07/03/2023] [Indexed: 07/24/2023] Open
Abstract
The p53 tumor suppressor exerts antitumor functions through its ability to regulate the transcription of its downstream targets. Long noncoding RNAs (lncRNAs) act as oncogenes or tumor suppressors implicated in tumorigenesis and tumor progression. Here, we identify the lncRNA LINC00324 (long intergenic noncoding RNA 00324) as a direct p53 transcriptional target. Knockdown of LINC00324 expression promotes tumor growth by reducing p53 transcriptional activity, whereas ectopic LINC00324 expression demonstrates a reverse effect. Notably, LINC00324 is present in the endogenous p53 complex in tumor cells and directly binds to the C-terminal domain of p53 in vitro. Mechanistically, LINC00324 enables p53 transactivation by competitively disrupting the p53-SET interaction, resulting in an increase of p300/CBP-mediated H3K18 and H3K27 acetylation on the p53 target promoters. Lower LINC00324 expression is associated with more aggressive disease status and predicts worse overall survival of patients with cancer. Our study identifies a p53/LINC00324 positive feedback loop that suppresses tumor growth by counteracting SET-mediated transcriptional repression.
Collapse
Affiliation(s)
- Ling Zhang
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Jun Zhang
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xiaofeng Xuan
- Department of Respiratory & Critical Care Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Di Wu
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Jianfeng Yu
- Department of Life Science and Technology, Changshu Institute of Technology, 99 South Third Ring Road, Suzhou 215500, China
| | - Peizhen Wang
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Xiaomei Yang
- Department of Emergency, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Jieru Zhang
- Department of Respiratory & Critical Care Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Wenjuan Gan
- Department of Pathology, Dushu Lake Hospital Affiliated to Soochow University, 9 Chongwen Road, Suzhou 215300, China
| | - Mengfan He
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China
| | - Xiao-Min Liu
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jun Zhou
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Donglai Wang
- State Key Laboratory of Medical Molecular Biology and Department of Medical Genetics, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Dawei Li
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou Medical College of Soochow University, 68 Jiyang West Road, Suzhou 215600, China.
| |
Collapse
|
7
|
Yi J, Tavana O, Li H, Wang D, Baer RJ, Gu W. Targeting USP2 regulation of VPRBP-mediated degradation of p53 and PD-L1 for cancer therapy. Nat Commun 2023; 14:1941. [PMID: 37024504 PMCID: PMC10079682 DOI: 10.1038/s41467-023-37617-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 03/24/2023] [Indexed: 04/08/2023] Open
Abstract
Since Mdm2 (Mouse double minute 2) inhibitors show serious toxicity in clinic studies, different approaches to achieve therapeutic reactivation of p53-mediated tumor suppression in cancers need to be explored. Here, we identify the USP2 (ubiquitin specific peptidase 2)-VPRBP (viral protein R binding protein) axis as an important pathway for p53 regulation. Like Mdm2, VPRBP is a potent repressor of p53 but VPRBP stability is controlled by USP2. Interestingly, the USP2-VPRBP axis also regulates PD-L1 (programmed death-ligand 1) expression. Strikingly, the combination of a small-molecule USP2 inhibitor and anti-PD1 monoclonal antibody leads to complete regression of the tumors expressing wild-type p53. In contrast to Mdm2, knockout of Usp2 in mice has no obvious effect in normal tissues. Moreover, no obvious toxicity is observed upon the USP2 inhibitor treatment in vivo as Mdm2-mediated regulation of p53 remains intact. Our study reveals a promising strategy for p53-based therapy by circumventing the toxicity issue.
Collapse
Affiliation(s)
- Jingjie Yi
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Omid Tavana
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Huan Li
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Donglai Wang
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Richard J Baer
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.
| |
Collapse
|
8
|
Webster SF, Ghalei H. Maturation of small nucleolar RNAs: from production to function. RNA Biol 2023; 20:715-736. [PMID: 37796118 PMCID: PMC10557570 DOI: 10.1080/15476286.2023.2254540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2023] [Indexed: 10/06/2023] Open
Abstract
Small Nucleolar RNAs (snoRNAs) are an abundant group of non-coding RNAs with well-defined roles in ribosomal RNA processing, folding and chemical modification. Besides their classic roles in ribosome biogenesis, snoRNAs are also implicated in several other cellular activities including regulation of splicing, transcription, RNA editing, cellular trafficking, and miRNA-like functions. Mature snoRNAs must undergo a series of processing steps tightly regulated by transiently associating factors and coordinated with other cellular processes including transcription and splicing. In addition to their mature forms, snoRNAs can contribute to gene expression regulation through their derivatives and degradation products. Here, we review the current knowledge on mechanisms of snoRNA maturation, including the different pathways of processing, and the regulatory mechanisms that control snoRNA levels and complex assembly. We also discuss the significance of studying snoRNA maturation, highlight the gaps in the current knowledge and suggest directions for future research in this area.
Collapse
Affiliation(s)
- Sarah F. Webster
- Biochemistry, Cell, and Developmental Biology Graduate Program, Emory University, Atlanta, Georgia, USA
- Department of Biochemistry, Emory University, Atlanta, Georgia, USA
| | - Homa Ghalei
- Department of Biochemistry, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
9
|
Lundine D, Annor GK, Chavez V, Maimos S, Syed Z, Jiang S, Ellison V, Bargonetti J. The C-terminus of Gain-of-Function Mutant p53 R273H Is Required for Association with PARP1 and Poly-ADP-Ribose. Mol Cancer Res 2022; 20:1799-1810. [PMID: 36074101 PMCID: PMC9716242 DOI: 10.1158/1541-7786.mcr-22-0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/02/2022] [Accepted: 09/02/2022] [Indexed: 01/15/2023]
Abstract
The TP53 gene is mutated in 80% of triple-negative breast cancers. Cells that harbor the hot-spot p53 gene mutation R273H produce an oncogenic mutant p53 (mtp53) that enhances cell proliferative and metastatic properties. The enhanced activities of mtp53 are collectively referred to as gain-of-function (GOF), and may include transcription-independent chromatin-based activities shared with wild-type p53 (wtp53) such as association with replicating DNA and DNA replication associated proteins like PARP1. However, how mtp53 upregulates cell proliferation is not well understood. wtp53 interacts with PARP1 using a portion of its C-terminus. The wtp53 oligomerization and far C-terminal domain (CTD) located within the C-terminus constitute putative GOF-associated domains, because mtp53 R273H expressing breast cancer cells lacking both domains manifest slow proliferation phenotypes. We addressed if the C-terminal region of mtp53 R273H is important for chromatin interaction and breast cancer cell proliferation using CRISPR-Cas9 mutated MDA-MB-468 cells endogenously expressing mtp53 R273H C-terminal deleted isoforms (R273HΔ381-388 and R273HΔ347-393). The mtp53 R273HΔ347-393 lacks the CTD and a portion of the oligomerization domain. We observed that cells harboring mtp53 R273HΔ347-393 (compared with mtp53 R273H full-length) manifest a significant reduction in chromatin, PARP1, poly-ADP-ribose (PAR), and replicating DNA binding. These cells also exhibited impaired response to hydroxyurea replicative stress, decreased sensitivity to the PARP-trapping drug combination temozolomide-talazoparib, and increased phosphorylated 53BP1 foci, suggesting reduced Okazaki fragment processing. IMPLICATIONS The C-terminal region of mtp53 confers GOF activity that mediates mtp53-PARP1 and PAR interactions assisting DNA replication, thus implicating new biomarkers for PARP inhibitor therapy.
Collapse
Affiliation(s)
- Devon Lundine
- The Department of Biological Sciences, Hunter College, Belfer Building, City University of New York, New York
- The Graduate Center Biology and Biochemistry Programs, City University of New York, New York
| | - George K. Annor
- The Department of Biological Sciences, Hunter College, Belfer Building, City University of New York, New York
- The Graduate Center Biology and Biochemistry Programs, City University of New York, New York
| | - Valery Chavez
- The Department of Biological Sciences, Hunter College, Belfer Building, City University of New York, New York
- The Graduate Center Biology and Biochemistry Programs, City University of New York, New York
| | - Styliana Maimos
- The Department of Biological Sciences, Hunter College, Belfer Building, City University of New York, New York
| | - Zafar Syed
- The Department of Biological Sciences, Hunter College, Belfer Building, City University of New York, New York
| | - Shuhong Jiang
- The Department of Biological Sciences, Hunter College, Belfer Building, City University of New York, New York
| | - Viola Ellison
- The Department of Biological Sciences, Hunter College, Belfer Building, City University of New York, New York
| | - Jill Bargonetti
- The Department of Biological Sciences, Hunter College, Belfer Building, City University of New York, New York
- The Graduate Center Biology and Biochemistry Programs, City University of New York, New York
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York
| |
Collapse
|
10
|
Deciphering the acetylation code of p53 in transcription regulation and tumor suppression. Oncogene 2022; 41:3039-3050. [PMID: 35487975 PMCID: PMC9149126 DOI: 10.1038/s41388-022-02331-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/14/2022] [Accepted: 04/20/2022] [Indexed: 12/16/2022]
Abstract
Although it is well established that p53-mediated tumor suppression mainly acts through its ability in transcriptional regulation, the molecular mechanisms of this regulation are not completely understood. Among a number of regulatory modes, acetylation of p53 attracts great interests. p53 was one of the first non-histone proteins found to be functionally regulated by acetylation and deacetylation, and subsequent work has established that reversible acetylation is a general mechanism for regulation of non-histone proteins. Unlike other types of post-translational modifications occurred during stress responses, the role of p53 acetylation has been recently validated in vivo by using the knockin mice with both acetylation-defective and acetylation-mimicking p53 mutants. Here, we review the role of acetylation in p53-mediated activities, with a focus on which specific acetylation sites are critical for p53-dependent transcription regulation during tumor suppression and how acetylation of p53 recruits specific “readers” to execute its promoter-specific regulation of different targets. We also discuss the role of p53 acetylation in differentially regulating its classic activities in cell cycle arrest, senescence and apoptosis as well as newly identified unconventional functions such as cell metabolism and ferroptosis.
Collapse
|
11
|
Kumar RD, Tosur M, Lalani SR, Mahoney DH, Bertuch AA. The germline p53 activation syndrome: A new patient further refines the clinical phenotype. Am J Med Genet A 2022; 188:2204-2208. [PMID: 35362179 DOI: 10.1002/ajmg.a.62749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/01/2022] [Accepted: 03/18/2022] [Indexed: 11/11/2022]
Abstract
The tumor suppressor p53 has well known roles in cancer development and germline cancer predisposition disorders, but increasing evidence supports the role of activation of this transcription factor in the pathogenesis of inherited bone marrow failure and chromosomal instability disorders. Here we report a patient with red cell aplasia, which was steroid responsive, as well as intellectual disability, seizures, microcephaly, short stature, cellular radiosensitivity, and normal telomere lengths, who had a germline heterozygous C-terminal frameshift variant in TP53 similar to others that activate the transcription factor. This is the third reported individual with a germline p53 activation syndrome, with several unique features that refine the clinical disease associated with these variants.
Collapse
Affiliation(s)
- Runjun D Kumar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Mustafa Tosur
- Department of Pediatrics, Division of Diabetes and Endocrinology, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA
| | - Seema R Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA
| | - Donald H Mahoney
- Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Division of Hematology/Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Alison A Bertuch
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Division of Hematology/Oncology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
12
|
Fedorova D, Ovsyannikova G, Kurnikova M, Pavlova A, Konyukhova T, Pshonkin A, Smetanina N. De novo TP53 germline activating mutations in two patients with the phenotype mimicking Diamond-Blackfan anemia. Pediatr Blood Cancer 2022; 69:e29558. [PMID: 35084091 DOI: 10.1002/pbc.29558] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 11/09/2022]
Abstract
Diamond-Blackfan anemia (DBA) is an inherited bone marrow failure syndrome, associated with mutations in ribosomal protein (RP) genes. Growing data on mutations in non-RP genes in patients with DBA-like phenotype became available over recent years. We describe two patients with the phenotype of DBA (onset of macrocytic anemia within the first year of life, paucity of erythroid precursors in bone marrow) and germline de novo variants in the TP53 gene. Both patients became transfusion independent, probably due to L-leucine therapy. The possible role of TP53 variants should be considered in patients with DBA-like phenotype and no mutations in RP genes.
Collapse
Affiliation(s)
- Daria Fedorova
- Dmitry Rogachev National Research Medical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Galina Ovsyannikova
- Dmitry Rogachev National Research Medical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Maria Kurnikova
- Dmitry Rogachev National Research Medical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Anna Pavlova
- Dmitry Rogachev National Research Medical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Tatiana Konyukhova
- Dmitry Rogachev National Research Medical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Alexey Pshonkin
- Dmitry Rogachev National Research Medical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| | - Nataliya Smetanina
- Dmitry Rogachev National Research Medical Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
| |
Collapse
|
13
|
Akter J, Katai Y, Sultana P, Takenobu H, Haruta M, Sugino RP, Mukae K, Satoh S, Wada T, Ohira M, Ando K, Kamijo T. Loss of p53 suppresses replication stress-induced DNA damage in ATRX-deficient neuroblastoma. Oncogenesis 2021; 10:73. [PMID: 34743173 PMCID: PMC8572175 DOI: 10.1038/s41389-021-00363-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/03/2021] [Accepted: 10/11/2021] [Indexed: 11/09/2022] Open
Abstract
Genetic aberrations are present in the ATRX gene in older high-risk neuroblastoma (NB) patients with very poor clinical outcomes. Its loss-of-function (LoF) facilitates the alternative lengthening of telomeres (ALT) pathway in tumor cells and is strongly linked to replication stress (RS) and DNA damage through G-quadruplex (G4) DNA secondary structures. However, limited information is available on ATRX alteration-related NB tumorigenesis. We herein knocked out (KO) ATRX in MYCN-amplified (NGP) and MYCN single copy (SK-N-AS) NB cells with wild-type (wt) and truncated TP53 at the C terminus, respectively, using CRISPR/Cas9 technologies. The loss of ATRX increased DNA damage and G4 formation related to RS in TP53 wt isogenic ATRX KO NGP cells, but not in SK-N-AS clones. A gene set enrichment analysis (GSEA) showed that the gene sets related to DNA double-strand break repair, negative cell cycle regulation, the G2M checkpoint, and p53 pathway activation were enriched in NGP clones. The accumulation of DNA damage activated the ATM/CHK2/p53 pathway, leading to cell cycle arrest in NGP clones. Interestingly, ATRX loss did not induce RS related to DNA damage response (DDR) in TP53-truncated SK-N-AS cells. p53 inactivation abrogated cell cycle arrest and reduced G4 accumulation in NGP clones. The loss of p53 also induced G4 DNA helicases or Fanconi anemia group D2 protein (FANCD2) with ATRX deficiency, suggesting that ATRX maintained genome integrity and p53 deficiency attenuated RS-induced DNA damage in NB cells featuring inactivated ATRX by regulating DNA repair mechanisms and replication fork stability.
Collapse
Affiliation(s)
- Jesmin Akter
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Yutaka Katai
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Parvin Sultana
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan.,Laboratory of Tumor Molecular Biology, Department of Graduate School of Science and Engineering, Saitama University, Saitama, Japan
| | - Hisanori Takenobu
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Masayuki Haruta
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Ryuichi P Sugino
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Kyosuke Mukae
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Shunpei Satoh
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Tomoko Wada
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Miki Ohira
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Kiyohiro Ando
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Takehiko Kamijo
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan. .,Laboratory of Tumor Molecular Biology, Department of Graduate School of Science and Engineering, Saitama University, Saitama, Japan.
| |
Collapse
|
14
|
Wang M, Attardi LD. A Balancing Act: p53 Activity from Tumor Suppression to Pathology and Therapeutic Implications. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 17:205-226. [PMID: 34699262 DOI: 10.1146/annurev-pathol-042320-025840] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
TP53, encoding the p53 transcription factor, is the most frequently mutated tumor suppressor gene across all human cancer types. While p53 has long been appreciated to induce antiproliferative cell cycle arrest, apoptosis, and senescence programs in response to diverse stress signals, various studies in recent years have revealed additional important functions for p53 that likely also contribute to tumor suppression, including roles in regulating tumor metabolism, ferroptosis, signaling in the tumor microenvironment, and stem cell self-renewal/differentiation. Not only does p53 loss or mutation cause cancer, but hyperactive p53 also drives various pathologies, including developmental phenotypes, premature aging, neurodegeneration, and side effects of cancer therapies. These findings underscore the importance of balanced p53 activity and influence our thinking of how to best develop cancer therapies based on modulating the p53 pathway. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 17 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Mengxiong Wang
- Department of Radiation Oncology, Division of Radiation and Cancer Biology, Stanford University School of Medicine, Stanford, California 94305, USA;
| | - Laura D Attardi
- Department of Radiation Oncology, Division of Radiation and Cancer Biology, Stanford University School of Medicine, Stanford, California 94305, USA; .,Department of Genetics and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
15
|
Tsai YY, Su CH, Tarn WY. p53 Activation in Genetic Disorders: Different Routes to the Same Destination. Int J Mol Sci 2021; 22:9307. [PMID: 34502215 PMCID: PMC8430931 DOI: 10.3390/ijms22179307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/18/2022] Open
Abstract
The tumor suppressor p53 is critical for preventing neoplastic transformation and tumor progression. Inappropriate activation of p53, however, has been observed in a number of human inherited disorders that most often affect development of the brain, craniofacial region, limb skeleton, and hematopoietic system. Genes related to these developmental disorders are essentially involved in transcriptional regulation/chromatin remodeling, rRNA metabolism, DNA damage-repair pathways, telomere maintenance, and centrosome biogenesis. Perturbation of these activities or cellular processes may result in p53 accumulation in cell cultures, animal models, and perhaps humans as well. Mouse models of several p53 activation-associated disorders essentially recapitulate human traits, and inactivation of p53 in these models can alleviate disorder-related phenotypes. In the present review, we focus on how dysfunction of the aforementioned biological processes causes developmental defects via excessive p53 activation. Notably, several disease-related genes exert a pleiotropic effect on those cellular processes, which may modulate the magnitude of p53 activation and establish or disrupt regulatory loops. Finally, we discuss potential therapeutic strategies for genetic disorders associated with p53 misactivation.
Collapse
|
16
|
Kon N, Gu W. p53 activation vs. stabilization: an acetylation tale from the C-terminal tail. Oncoscience 2021; 8:58-60. [PMID: 33997110 PMCID: PMC8115982 DOI: 10.18632/oncoscience.534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/06/2021] [Indexed: 12/04/2022] Open
Affiliation(s)
- Ning Kon
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.,Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.,Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
17
|
Kon N, Churchill M, Li H, Mukherjee S, Manfredi JJ, Gu W. Robust p53 Stabilization Is Dispensable for Its Activation and Tumor Suppressor Function. Cancer Res 2021; 81:935-944. [PMID: 33323382 PMCID: PMC8026563 DOI: 10.1158/0008-5472.can-20-1804] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 11/06/2020] [Accepted: 12/10/2020] [Indexed: 11/16/2022]
Abstract
p53 is a short-lived protein with low basal levels under normal homeostasis conditions. However, upon DNA damage, levels of p53 dramatically increase for its activation. Although robust stabilization of p53 serves as a "trademark" for DNA damage responses, the requirement for such dramatic protein stabilization in tumor suppression has not been well addressed. Here we generated a mutant p53KQ mouse where all the C-terminal domain lysine residues were mutated to glutamines (K to Q mutations at K367, K369, K370, K378, K379, K383, and K384) to mimic constitutive acetylation of the p53 C-terminus. Because of p53 activation, p53KQ/KQ mice were perinatal lethal, yet this lethality was averted in p53KQ/- mice, which displayed normal postnatal development. Nevertheless, p53KQ/- mice died prematurely due to anemia and hematopoiesis failure. Further analyses indicated that expression of the acetylation-mimicking p53 mutant in vivo induces activation of p53 targets in various tissues without obviously increasing p53 levels. In the well-established pancreatic ductal adenocarcinoma (PDAC) mouse model, expression of the acetylation-mimicking p53-mutant protein effectively suppressed K-Ras-induced PDAC development in the absence of robust p53 stabilization. Together, our results provide proof-of-principle evidence that p53-mediated transcriptional function and tumor suppression can be achieved independently of its robust stabilization and reveal an alternative approach to activate p53 function for therapeutic purposes. SIGNIFICANCE: Although robust p53 stabilization is critical for acute p53 responses such as DNA damage, this study underscores the important role of low basal p53 protein levels in p53 activation and tumor suppression.
Collapse
Affiliation(s)
- Ning Kon
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, New York, New York
| | - Michael Churchill
- Department of Medicine and Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, New York, New York
| | - Huan Li
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, New York, New York
| | - Siddhartha Mukherjee
- Department of Medicine and Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, New York, New York
| | - James J Manfredi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Wei Gu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, New York, New York.
| |
Collapse
|
18
|
Phosphatase magnesium-dependent 1 δ (PPM1D), serine/threonine protein phosphatase and novel pharmacological target in cancer. Biochem Pharmacol 2020; 184:114362. [PMID: 33309518 DOI: 10.1016/j.bcp.2020.114362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022]
Abstract
Aberrations in DNA damage response genes are recognized mediators of tumorigenesis and resistance to chemo- and radiotherapy. While protein phosphatase magnesium-dependent 1 δ (PPM1D), located on the long arm of chromosome 17 at 17q22-23, is a key regulator of cellular responses to DNA damage, amplification, overexpression, or mutation of this gene is important in a wide range of pathologic processes. In this review, we describe the physiologic function of PPM1D, as well as its role in diverse processes, including fertility, development, stemness, immunity, tumorigenesis, and treatment responsiveness. We highlight both the advances and limitations of current approaches to targeting malignant processes mediated by pathogenic alterations in PPM1D with the goal of providing rationale for continued research and development of clinically viable treatment approaches for PPM1D-associated diseases.
Collapse
|
19
|
Park D, Bergin SM, Jones D, Ru P, Koivisto CS, Jeon YJ, Sizemore GM, Kladney RD, Hadjis A, Shakya R, Ludwig T. Ablation of the Brca1-Palb2 Interaction Phenocopies Fanconi Anemia in Mice. Cancer Res 2020; 80:4172-4184. [PMID: 32732220 DOI: 10.1158/0008-5472.can-20-0486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/01/2020] [Accepted: 07/21/2020] [Indexed: 11/16/2022]
Abstract
Heterozygous mutations in the BRCA1 gene predispose women to breast and ovarian cancer, while biallelic BRCA1 mutations are a cause of Fanconi anemia (FA), a rare genetic disorder characterized by developmental abnormalities, early-onset bone marrow failure, increased risk of cancers, and hypersensitivity to DNA-crosslinking agents. BRCA1 is critical for homologous recombination of DNA double-strand breaks (DSB). Through its coiled-coil domain, BRCA1 interacts with an essential partner, PALB2, recruiting BRCA2 and RAD51 to sites of DNA damage. Missense mutations within the coiled-coil domain of BRCA1 (e.g., L1407P) that affect the interaction with PALB2 have been reported in familial breast cancer. We hypothesized that if PALB2 regulates or mediates BRCA1 tumor suppressor function, ablation of the BRCA1-PALB2 interaction may also elicit genomic instability and tumor susceptibility. We generated mice defective for the Brca1-Palb2 interaction (Brca1 L1363P in mice) and established MEF cells from these mice. Brca1 L1363P/L1363P MEF exhibited hypersensitivity to DNA-damaging agents and failed to recruit Rad51 to DSB. Brca1 L1363P/L1363P mice were viable but exhibited various FA symptoms including growth retardation, hyperpigmentation, skeletal abnormalities, and male/female infertility. Furthermore, all Brca1 L1363P/L1363P mice exhibited macrocytosis and died due to bone marrow failure or lymphoblastic lymphoma/leukemia with activating Notch1 mutations. These phenotypes closely recapitulate clinical features observed in patients with FA. Collectively, this model effectively demonstrates the significance of the BRCA1-PALB2 interaction in genome integrity and provides an FA model to investigate hematopoietic stem cells for mechanisms underlying progressive failure of hematopoiesis and associated development of leukemia/lymphoma, and other FA phenotypes. SIGNIFICANCE: A new Brca1 mouse model for Fanconi anemia (FA) complementation group S provides a system in which to study phenotypes observed in human FA patients including bone marrow failure.See related commentary by Her and Bunting, p. 4044.
Collapse
Affiliation(s)
- Dongju Park
- Department of Cancer Biology and Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.
| | - Stephen M Bergin
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Dan Jones
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio.,The James Polaris Molecular Laboratory, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio.,Division of Molecular Pathology, Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Peng Ru
- The James Polaris Molecular Laboratory, The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Christopher S Koivisto
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Young-Jun Jeon
- Department of Cancer Biology and Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, South Korea
| | - Gina M Sizemore
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio.,Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Raleigh D Kladney
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Ashley Hadjis
- Department of Cancer Biology and Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Reena Shakya
- The Ohio State University Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Thomas Ludwig
- Department of Cancer Biology and Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.
| |
Collapse
|
20
|
Toufektchan E, Lejour V, Durand R, Giri N, Draskovic I, Bardot B, Laplante P, Jaber S, Alter BP, Londono-Vallejo JA, Savage SA, Toledo F. Germline mutation of MDM4, a major p53 regulator, in a familial syndrome of defective telomere maintenance. SCIENCE ADVANCES 2020; 6:eaay3511. [PMID: 32300648 PMCID: PMC7148086 DOI: 10.1126/sciadv.aay3511] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 01/16/2020] [Indexed: 05/08/2023]
Abstract
Dyskeratosis congenita is a cancer-prone inherited bone marrow failure syndrome caused by telomere dysfunction. A mouse model recently suggested that p53 regulates telomere metabolism, but the clinical relevance of this finding remained uncertain. Here, a germline missense mutation of MDM4, a negative regulator of p53, was found in a family with features suggestive of dyskeratosis congenita, e.g., bone marrow hypocellularity, short telomeres, tongue squamous cell carcinoma, and acute myeloid leukemia. Using a mouse model, we show that this mutation (p.T454M) leads to increased p53 activity, decreased telomere length, and bone marrow failure. Variations in p53 activity markedly altered the phenotype of Mdm4 mutant mice, suggesting an explanation for the variable expressivity of disease symptoms in the family. Our data indicate that a germline activation of the p53 pathway may cause telomere dysfunction and point to polymorphisms affecting this pathway as potential genetic modifiers of telomere biology and bone marrow function.
Collapse
Affiliation(s)
- Eléonore Toufektchan
- Genetics of Tumor Suppression, Institut Curie, Paris, France
- CNRS UMR 3244, Paris, France
- Sorbonne Université, Paris, France
- PSL Research University, Paris, France
| | - Vincent Lejour
- Genetics of Tumor Suppression, Institut Curie, Paris, France
- CNRS UMR 3244, Paris, France
- Sorbonne Université, Paris, France
- PSL Research University, Paris, France
| | - Romane Durand
- Genetics of Tumor Suppression, Institut Curie, Paris, France
- CNRS UMR 3244, Paris, France
- Sorbonne Université, Paris, France
- PSL Research University, Paris, France
| | - Neelam Giri
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Irena Draskovic
- CNRS UMR 3244, Paris, France
- Sorbonne Université, Paris, France
- PSL Research University, Paris, France
- Telomeres and Cancer, Institut Curie, Paris, France
| | - Boris Bardot
- Genetics of Tumor Suppression, Institut Curie, Paris, France
- CNRS UMR 3244, Paris, France
- Sorbonne Université, Paris, France
- PSL Research University, Paris, France
| | - Pierre Laplante
- Genetics of Tumor Suppression, Institut Curie, Paris, France
- CNRS UMR 3244, Paris, France
- Sorbonne Université, Paris, France
- PSL Research University, Paris, France
| | - Sara Jaber
- Genetics of Tumor Suppression, Institut Curie, Paris, France
- CNRS UMR 3244, Paris, France
- Sorbonne Université, Paris, France
- PSL Research University, Paris, France
| | - Blanche P. Alter
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - José-Arturo Londono-Vallejo
- CNRS UMR 3244, Paris, France
- Sorbonne Université, Paris, France
- PSL Research University, Paris, France
- Telomeres and Cancer, Institut Curie, Paris, France
| | - Sharon A. Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Franck Toledo
- Genetics of Tumor Suppression, Institut Curie, Paris, France
- CNRS UMR 3244, Paris, France
- Sorbonne Université, Paris, France
- PSL Research University, Paris, France
- Corresponding author.
| |
Collapse
|
21
|
Timofeev O, Klimovich B, Schneikert J, Wanzel M, Pavlakis E, Noll J, Mutlu S, Elmshäuser S, Nist A, Mernberger M, Lamp B, Wenig U, Brobeil A, Gattenlöhner S, Köhler K, Stiewe T. Residual apoptotic activity of a tumorigenic p53 mutant improves cancer therapy responses. EMBO J 2019; 38:e102096. [PMID: 31483066 PMCID: PMC6792016 DOI: 10.15252/embj.2019102096] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/30/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022] Open
Abstract
Engineered p53 mutant mice are valuable tools for delineating p53 functions in tumor suppression and cancer therapy. Here, we have introduced the R178E mutation into the Trp53 gene of mice to specifically ablate the cooperative nature of p53 DNA binding. Trp53R178E mice show no detectable target gene regulation and, at first sight, are largely indistinguishable from Trp53−/− mice. Surprisingly, stabilization of p53R178E in Mdm2−/− mice nevertheless triggers extensive apoptosis, indicative of residual wild‐type activities. Although this apoptotic activity suffices to trigger lethality of Trp53R178E;Mdm2−/− embryos, it proves insufficient for suppression of spontaneous and oncogene‐driven tumorigenesis. Trp53R178E mice develop tumors indistinguishably from Trp53−/− mice and tumors retain and even stabilize the p53R178E protein, further attesting to the lack of significant tumor suppressor activity. However, Trp53R178E tumors exhibit remarkably better chemotherapy responses than Trp53−/− ones, resulting in enhanced eradication of p53‐mutated tumor cells. Together, this provides genetic proof‐of‐principle evidence that a p53 mutant can be highly tumorigenic and yet retain apoptotic activity which provides a survival benefit in the context of cancer therapy.
Collapse
Affiliation(s)
- Oleg Timofeev
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Boris Klimovich
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Jean Schneikert
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Michael Wanzel
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany.,German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Marburg, Germany
| | | | - Julia Noll
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Samet Mutlu
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Andrea Nist
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Boris Lamp
- Genomics Core Facility, Philipps University, Marburg, Germany
| | - Ulrich Wenig
- Institute of Pathology, Justus Liebig University, Giessen, Germany
| | | | | | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University, Giessen, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany.,German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Marburg, Germany.,Genomics Core Facility, Philipps University, Marburg, Germany
| |
Collapse
|
22
|
Fischbach A, Krüger A, Hampp S, Assmann G, Rank L, Hufnagel M, Stöckl MT, Fischer JMF, Veith S, Rossatti P, Ganz M, Ferrando-May E, Hartwig A, Hauser K, Wiesmüller L, Bürkle A, Mangerich A. The C-terminal domain of p53 orchestrates the interplay between non-covalent and covalent poly(ADP-ribosyl)ation of p53 by PARP1. Nucleic Acids Res 2019; 46:804-822. [PMID: 29216372 PMCID: PMC5778597 DOI: 10.1093/nar/gkx1205] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/22/2017] [Indexed: 01/25/2023] Open
Abstract
The post-translational modification poly(ADP-ribosyl)ation (PARylation) plays key roles in genome maintenance and transcription. Both non-covalent poly(ADP-ribose) binding and covalent PARylation control protein functions, however, it is unknown how the two modes of modification crosstalk mechanistically. Employing the tumor suppressor p53 as a model substrate, this study provides detailed insights into the interplay between non-covalent and covalent PARylation and unravels its functional significance in the regulation of p53. We reveal that the multifunctional C-terminal domain (CTD) of p53 acts as the central hub in the PARylation-dependent regulation of p53. Specifically, p53 bound to auto-PARylated PARP1 via highly specific non–covalent PAR-CTD interaction, which conveyed target specificity for its covalent PARylation by PARP1. Strikingly, fusing the p53-CTD to a protein that is normally not PARylated, renders this a target for covalent PARylation as well. Functional studies revealed that the p53–PAR interaction had substantial implications on molecular and cellular levels. Thus, PAR significantly influenced the complex p53–DNA binding properties and controlled p53 functions, with major implications on the p53-dependent interactome, transcription, and replication-associated recombination. Remarkably, this mechanism potentially also applies to other PARylation targets, since a bioinformatics analysis revealed that CTD-like regions are highly enriched in the PARylated proteome.
Collapse
Affiliation(s)
- Arthur Fischbach
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany.,Konstanz Research School Chemical Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Annika Krüger
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany.,Konstanz Research School Chemical Biology, University of Konstanz, 78457 Konstanz, Germany.,Department of Chemistry, University of Konstanz, 78457 Konstanz, Germany
| | - Stephanie Hampp
- Department of Obstetrics and Gynaecology, University of Ulm, 89075 Ulm, Germany
| | - Greta Assmann
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany.,Konstanz Research School Chemical Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Lisa Rank
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Matthias Hufnagel
- Department of Food Chemistry and Toxicology, Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), 76131 Karlsruhe, Germany
| | - Martin T Stöckl
- Bioimaging Center, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Jan M F Fischer
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany.,Konstanz Research School Chemical Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Sebastian Veith
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany.,Research Training Group 1331, University of Konstanz, 78457 Konstanz, Germany
| | - Pascal Rossatti
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Magdalena Ganz
- Bioimaging Center, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Elisa Ferrando-May
- Bioimaging Center, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Andrea Hartwig
- Department of Food Chemistry and Toxicology, Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), 76131 Karlsruhe, Germany
| | - Karin Hauser
- Department of Chemistry, University of Konstanz, 78457 Konstanz, Germany
| | - Lisa Wiesmüller
- Department of Obstetrics and Gynaecology, University of Ulm, 89075 Ulm, Germany
| | - Alexander Bürkle
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Aswin Mangerich
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| |
Collapse
|
23
|
Benyelles M, Episkopou H, O'Donohue M, Kermasson L, Frange P, Poulain F, Burcu Belen F, Polat M, Bole‐Feysot C, Langa‐Vives F, Gleizes P, de Villartay J, Callebaut I, Decottignies A, Revy P. Impaired telomere integrity and rRNA biogenesis in PARN-deficient patients and knock-out models. EMBO Mol Med 2019; 11:e10201. [PMID: 31273937 PMCID: PMC6609912 DOI: 10.15252/emmm.201810201] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/24/2019] [Accepted: 05/09/2019] [Indexed: 12/12/2022] Open
Abstract
PARN, poly(A)-specific ribonuclease, regulates the turnover of mRNAs and the maturation and stabilization of the hTR RNA component of telomerase. Biallelic PARN mutations were associated with Høyeraal-Hreidarsson (HH) syndrome, a rare telomere biology disorder that, because of its severity, is likely not exclusively due to hTR down-regulation. Whether PARN deficiency was affecting the expression of telomere-related genes was still unclear. Using cells from two unrelated HH individuals carrying novel PARN mutations and a human PARN knock-out (KO) cell line with inducible PARN complementation, we found that PARN deficiency affects both telomere length and stability and down-regulates the expression of TRF1, TRF2, TPP1, RAP1, and POT1 shelterin transcripts. Down-regulation of dyskerin-encoding DKC1 mRNA was also observed and found to result from p53 activation in PARN-deficient cells. We further showed that PARN deficiency compromises ribosomal RNA biogenesis in patients' fibroblasts and cells from heterozygous Parn KO mice. Homozygous Parn KO however resulted in early embryonic lethality that was not overcome by p53 KO. Our results refine our knowledge on the pleiotropic cellular consequences of PARN deficiency.
Collapse
Affiliation(s)
- Maname Benyelles
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| | | | - Marie‐Françoise O'Donohue
- Laboratoire de Biologie Moléculaire EucaryoteCentre de Biologie Intégrative (CBI)CNRS, UPSUniversité de ToulouseToulouseFrance
| | - Laëtitia Kermasson
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| | - Pierre Frange
- EA 7327, Université Paris Descartes, Sorbonne Paris‐CitéParisFrance
- Laboratoire de Microbiologie clinique & Unité d'ImmunologieHématologie et Rhumatologie PédiatriquesAP‐HP, Hôpital Necker, Enfants MaladesParisFrance
| | - Florian Poulain
- de Duve InstituteUniversité catholique de LouvainBrusselsBelgium
| | - Fatma Burcu Belen
- Pediatric HematologyFaculty of MedicineBaskent UniversityAnkaraTurkey
| | - Meltem Polat
- Pediatric Infectious DiseasesDepartment of Pediatric Infectious DiseasesPamukkale University Medical FacultyDenizliTurkey
| | - Christine Bole‐Feysot
- INSERM, UMR 1163Genomics platform, Imagine InstituteParis Descartes–Sorbonne Paris Cité UniversityParisFrance
- Genomic Core FacilityImagine Institute‐Structure Fédérative de Recherche NeckerINSERM U1163ParisFrance
| | | | - Pierre‐Emmanuel Gleizes
- Laboratoire de Biologie Moléculaire EucaryoteCentre de Biologie Intégrative (CBI)CNRS, UPSUniversité de ToulouseToulouseFrance
| | - Jean‐Pierre de Villartay
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| | - Isabelle Callebaut
- Muséum National d'Histoire NaturelleUMR CNRS 7590Institut de Minéralogiede Physique des Matériaux et de Cosmochimie, IMPMCSorbonne UniversitéParisFrance
| | | | - Patrick Revy
- Laboratory of Genome Dynamics in the Immune SystemINSERM, UMR 1163ParisFrance
- Laboratoire labellisé LigueImagine InstituteParis Descartes–Sorbonne Paris Cite UniversityParisFrance
| |
Collapse
|
24
|
Elevated telomere dysfunction in cells containing the African-centric Pro47Ser cancer-risk variant of TP53. Oncotarget 2019; 10:3581-3591. [PMID: 31217894 PMCID: PMC6557208 DOI: 10.18632/oncotarget.26980] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/13/2019] [Indexed: 11/25/2022] Open
Abstract
Subtelomeric transcription and chromatin can have a significant impact on telomere repeat maintenance and chromosome stability. We have previously found that tumor suppressor protein p53 (TP53) can bind to retrotransposon-like elements in a majority of human subtelomeres to regulate TERRA transcription and telomeric histone acetylation in response to DNA damage. TP53 also prevents the accumulation of γH2AX DNA-damage signaling at telomeres. We now show that the inherited TP53 polymorphism Pro47Ser (hereafter S47), which is enriched in populations of African descent, is associated with elevated marks of telomere dysfunction. We found that human and mouse cells carrying the S47 variant show increased γH2AX DNA-damage signals at telomeres, as well as reduced TERRA transcription and subtelomeric histone acetylation in response to DNA damage stress. Cell-lines containing inducible genes for P47 or S47 versions of p53, as well mouse embryo fibroblasts (MEFs) reconstituted with human p53, showed elevated telomere-induced DNA damage foci and metaphase telomere signal loss in cells with S47. Human lymphoblastoid cell lines (LCLs) derived from individuals homozygous for S47, show increased accumulation of subtelomeric γH2AX and unstable telomere repeats in response to DNA damage relative to age matched LCLs homozygous for P47. Furthermore, LCLs with S47 had reduced replicative lifespan. These studies indicate that the naturally occurring S47 variant of p53 can affect telomeric chromatin, telomere repeat stability, and replicative capacity. We discuss the potential evolutionary significance of the S47 variant to African populations with respect to telomere regulation and the implications for inherited health disparities.
Collapse
|
25
|
Interaction between p53 N terminus and core domain regulates specific and nonspecific DNA binding. Proc Natl Acad Sci U S A 2019; 116:8859-8868. [PMID: 30988205 DOI: 10.1073/pnas.1903077116] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The p53 tumor suppressor is a sequence-specific DNA binding protein that activates gene transcription to regulate cell survival and proliferation. Dynamic control of p53 degradation and DNA binding in response to stress signals are critical for tumor suppression. The p53 N terminus (NT) contains two transactivation domains (TAD1 and TAD2), a proline-rich region (PRR), and multiple phosphorylation sites. Previous work revealed the p53 NT reduced DNA binding in vitro. Here, we show that TAD2 and the PRR inhibit DNA binding by directly interacting with the sequence-specific DNA binding domain (DBD). NMR spectroscopy revealed that TAD2 and the PRR interact with the DBD at or near the DNA binding surface, possibly acting as a nucleic acid mimetic to competitively block DNA binding. In vitro and in vivo DNA binding analyses showed that the NT reduced p53 DNA binding affinity but improved the ability of p53 to distinguish between specific and nonspecific sequences. MDMX inhibits p53 binding to specific target promoters but stimulates binding to nonspecific chromatin sites. The results suggest that the p53 NT regulates the affinity and specificity of DNA binding by the DBD. The p53 NT-interacting proteins and posttranslational modifications may regulate DNA binding, partly by modulating the NT-DBD interaction.
Collapse
|
26
|
Kon N, Wang D, Gu W. Loss of SET reveals both the p53-dependent and the p53-independent functions in vivo. Cell Death Dis 2019; 10:237. [PMID: 30858352 PMCID: PMC6411979 DOI: 10.1038/s41419-019-1484-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/28/2019] [Accepted: 02/20/2019] [Indexed: 11/30/2022]
Abstract
Our previous study showed that the oncoprotein SET acts as a new reader of unacetylated p53 for transcriptional repression. To further elucidate the physiological significance of SET in vivo, we generated set knockout mice. Set knockout mice died during embryonic development between day 11.5 and day 12.5 post coitum, exhibiting cardiac edema and open neural tube, among other developmental defects. Further analyses revealed that loss of SET leads to upregulation of p53 target genes including p21 and puma without any obvious effect on p53 stability in set knockout embryos. Notably, the developmental defects of set knockout mice were significantly, but nonetheless partially, rescued by concomitant deletion of p53. The failure to obtain fully live set/p53 double knockout mice suggested that p53-independent targets of SET also contribute to the embryonic lethality of set knockout mice. Indeed, we found that FOXO1 acts as an important target of SET and that SET-mediated regulation of FOXO1 is also acetylation-dependent. Taken together, these data underscore the importance of SET oncoprotein during embryonic development and reveal both of the p53-dependent and the p53-independent functions of SET in vivo.
Collapse
Affiliation(s)
- Ning Kon
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Donglai Wang
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.,National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.,Department of Medical Genetics, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China
| | - Wei Gu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.
| |
Collapse
|
27
|
Hafner A, Bulyk ML, Jambhekar A, Lahav G. The multiple mechanisms that regulate p53 activity and cell fate. Nat Rev Mol Cell Biol 2019; 20:199-210. [DOI: 10.1038/s41580-019-0110-x] [Citation(s) in RCA: 452] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
28
|
Bowen ME, Attardi LD. The role of p53 in developmental syndromes. J Mol Cell Biol 2019; 11:200-211. [PMID: 30624728 PMCID: PMC6478128 DOI: 10.1093/jmcb/mjy087] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/22/2018] [Accepted: 01/06/2019] [Indexed: 12/17/2022] Open
Abstract
While it is well appreciated that loss of the p53 tumor suppressor protein promotes cancer, growing evidence indicates that increased p53 activity underlies the developmental defects in a wide range of genetic syndromes. The inherited or de novo mutations that cause these syndromes affect diverse cellular processes, such as ribosome biogenesis, DNA repair, and centriole duplication, and analysis of human patient samples and mouse models demonstrates that disrupting these cellular processes can activate the p53 pathway. Importantly, many of the developmental defects in mouse models of these syndromes can be rescued by loss of p53, indicating that inappropriate p53 activation directly contributes to their pathogenesis. A role for p53 in driving developmental defects is further supported by the observation that mouse strains with broad p53 hyperactivation, due to mutations affecting p53 pathway components, display a host of tissue-specific developmental defects, including hematopoietic, neuronal, craniofacial, cardiovascular, and pigmentation defects. Furthermore, germline activating mutations in TP53 were recently identified in two human patients exhibiting bone marrow failure and other developmental defects. Studies in mice suggest that p53 drives developmental defects by inducing apoptosis, restraining proliferation, or modulating other developmental programs in a cell type-dependent manner. Here, we review the growing body of evidence from mouse models that implicates p53 as a driver of tissue-specific developmental defects in diverse genetic syndromes.
Collapse
Affiliation(s)
- Margot E Bowen
- Division of Radiation and Cancer Biology in the Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology in the Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
29
|
Hayman L, Chaudhry WR, Revin VV, Zhelev N, Bourdon JC. What is the potential of p53 isoforms as a predictive biomarker in the treatment of cancer? Expert Rev Mol Diagn 2019; 19:149-159. [DOI: 10.1080/14737159.2019.1563484] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Liam Hayman
- School of Science, Engineering and Technology, Abertay University, Dundee, Scotland
| | - Wajeeh Raza Chaudhry
- School of Medicine, University of Dundee, Dundee Cancer Centre, Dundee, Scotland
| | - Victor V. Revin
- Department of Biotechnology, Bioengineering and Biochemistry, Faculty of Biotechnology and Biology, Federal state-financed academic institution of higher education, National Research Ogarev Mordovia State University, Saransk, Republic of Mordovia, Russia
| | - Nikolai Zhelev
- School of Science, Engineering and Technology, Abertay University, Dundee, Scotland
| | | |
Collapse
|
30
|
De Novo Mutations Activating Germline TP53 in an Inherited Bone-Marrow-Failure Syndrome. Am J Hum Genet 2018; 103:440-447. [PMID: 30146126 DOI: 10.1016/j.ajhg.2018.07.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/24/2018] [Indexed: 01/09/2023] Open
Abstract
Inherited bone-marrow-failure syndromes (IBMFSs) include heterogeneous genetic disorders characterized by bone-marrow failure, congenital anomalies, and an increased risk of malignancy. Many lines of evidence have suggested that p53 activation might be central to the pathogenesis of IBMFSs, including Diamond-Blackfan anemia (DBA) and dyskeratosis congenita (DC). However, the exact role of p53 activation in each clinical feature remains unknown. Here, we report unique de novo TP53 germline variants found in two individuals with an IBMFS accompanied by hypogammaglobulinemia, growth retardation, and microcephaly mimicking DBA and DC. TP53 is a tumor-suppressor gene most frequently mutated in human cancers, and occasional germline variants occur in Li-Fraumeni cancer-predisposition syndrome. Most of these mutations affect the core DNA-binding domain, leading to compromised transcriptional activities. In contrast, the variants found in the two individuals studied here caused the same truncation of the protein, resulting in the loss of 32 residues from the C-terminal domain (CTD). Unexpectedly, the p53 mutant had augmented transcriptional activities, an observation not previously described in humans. When we expressed this mutant in zebrafish and human-induced pluripotent stem cells, we observed impaired erythrocyte production. These findings together with close similarities to published knock-in mouse models of TP53 lacking the CTD demonstrate that the CTD-truncation mutations of TP53 cause IBMFS, providing important insights into the previously postulated connection between p53 and IBMFSs.
Collapse
|
31
|
Marcel V, Nguyen Van Long F, Diaz JJ. 40 Years of Research Put p53 in Translation. Cancers (Basel) 2018; 10:E152. [PMID: 29883412 PMCID: PMC5977125 DOI: 10.3390/cancers10050152] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/15/2018] [Accepted: 05/18/2018] [Indexed: 12/18/2022] Open
Abstract
Since its discovery in 1979, p53 has shown multiple facets. Initially the tumor suppressor p53 protein was considered as a stress sensor able to maintain the genome integrity by regulating transcription of genes involved in cell cycle arrest, apoptosis and DNA repair. However, it rapidly came into light that p53 regulates gene expression to control a wider range of biological processes allowing rapid cell adaptation to environmental context. Among them, those related to cancer have been extensively documented. In addition to its role as transcription factor, scattered studies reported that p53 regulates miRNA processing, modulates protein activity by direct interaction or exhibits RNA-binding activity, thus suggesting a role of p53 in regulating several layers of gene expression not restricted to transcription. After 40 years of research, it appears more and more clearly that p53 is strongly implicated in translational regulation as well as in the control of the production and activity of the translational machinery. Translation control of specific mRNAs could provide yet unsuspected capabilities to this well-known guardian of the genome.
Collapse
Affiliation(s)
- Virginie Marcel
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France.
| | - Flora Nguyen Van Long
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France.
| | - Jean-Jacques Diaz
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France.
| |
Collapse
|
32
|
The Guardian of the Genome Revisited: p53 Downregulates Genes Required for Telomere Maintenance, DNA Repair, and Centromere Structure. Cancers (Basel) 2018; 10:cancers10050135. [PMID: 29734785 PMCID: PMC5977108 DOI: 10.3390/cancers10050135] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 02/06/2023] Open
Abstract
The p53 protein has been extensively studied for its capacity to prevent proliferation of cells with a damaged genome. Surprisingly, however, our recent analysis of mice expressing a hyperactive mutant p53 that lacks the C-terminal domain revealed that increased p53 activity may alter genome maintenance. We showed that p53 downregulates genes essential for telomere metabolism, DNA repair, and centromere structure and that a sustained p53 activity leads to phenotypic traits associated with dyskeratosis congenita and Fanconi anemia. This downregulation is largely conserved in human cells, which suggests that our findings could be relevant to better understand processes involved in bone marrow failure as well as aging and tumor suppression.
Collapse
|
33
|
Sullivan KD, Galbraith MD, Andrysik Z, Espinosa JM. Mechanisms of transcriptional regulation by p53. Cell Death Differ 2017; 25:133-143. [PMID: 29125602 PMCID: PMC5729533 DOI: 10.1038/cdd.2017.174] [Citation(s) in RCA: 306] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/25/2017] [Accepted: 08/31/2017] [Indexed: 12/19/2022] Open
Abstract
p53 is a transcription factor that suppresses tumor growth through regulation of dozens of target genes with diverse biological functions. The activity of this master transcription factor is inactivated in nearly all tumors, either by mutations in the TP53 locus or by oncogenic events that decrease the activity of the wild-type protein, such as overexpression of the p53 repressor MDM2. However, despite decades of intensive research, our collective understanding of the p53 signaling cascade remains incomplete. In this review, we focus on recent advances in our understanding of mechanisms of p53-dependent transcriptional control as they relate to five key areas: (1) the functionally distinct N-terminal transactivation domains, (2) the diverse regulatory roles of its C-terminal domain, (3) evidence that p53 is solely a direct transcriptional activator, not a direct repressor, (4) the ability of p53 to recognize many of its enhancers across diverse chromatin environments, and (5) mechanisms that modify the p53-dependent transcriptional program in a context-dependent manner.
Collapse
Affiliation(s)
- Kelly D Sullivan
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.,Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Matthew D Galbraith
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.,Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Zdenek Andrysik
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.,Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Joaquin M Espinosa
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.,Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO 80045, USA.,Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80203, USA
| |
Collapse
|
34
|
Control of Cellular Aging, Tissue Function, and Cancer by p53 Downstream of Telomeres. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026088. [PMID: 28289249 DOI: 10.1101/cshperspect.a026088] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Telomeres, the nucleoprotein complex at the ends of eukaryotic chromosomes, perform an essential cellular role in part by preventing the chromosomal end from initiating a DNA-damage response. This function of telomeres can be compromised as telomeres erode either as a consequence of cell division in culture or as a normal part of cellular ageing in proliferative tissues. Telomere dysfunction in this context leads to DNA-damage signaling and activation of the tumor-suppressor protein p53, which then can prompt either cellular senescence or apoptosis. By culling cells with dysfunctional telomeres, p53 plays a critical role in protecting tissues against the effects of critically short telomeres. However, as telomere dysfunction worsens, p53 likely exacerbates short telomere-driven tissue failure diseases, including pulmonary fibrosis, aplastic anemia, and liver cirrhosis. In cells lacking p53, unchecked telomere shortening drives chromosomal end-to-end fusions and cycles of chromosome fusion-bridge-breakage. Incipient cancer cells confronting these telomere barriers must disable p53 signaling to avoid senescence and eventually up-regulate telomerase to achieve cellular immortality. The recent findings of highly recurrent activating mutations in the promoter for the telomerase reverse transcriptase (TERT) gene in diverse human cancers, together with the widespread mutations in p53 in cancer, provide support for the idea that circumvention of a telomere-p53 checkpoint is essential for malignant progression in human cancer.
Collapse
|
35
|
Kominakis A, Hager-Theodorides AL, Zoidis E, Saridaki A, Antonakos G, Tsiamis G. Combined GWAS and 'guilt by association'-based prioritization analysis identifies functional candidate genes for body size in sheep. Genet Sel Evol 2017; 49:41. [PMID: 28454565 PMCID: PMC5408376 DOI: 10.1186/s12711-017-0316-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 04/19/2017] [Indexed: 12/30/2022] Open
Abstract
Background Body size in sheep is an important indicator of productivity, growth and health as well as of environmental adaptation. It is a composite quantitative trait that has been studied with high-throughput genomic methods, i.e. genome-wide association studies (GWAS) in various mammalian species. Several genomic markers have been associated with body size traits and genes have been identified as causative candidates in humans, dog and cattle. A limited number of related GWAS have been performed in various sheep breeds and have identified genomic regions and candidate genes that partly account for body size variability. Here, we conducted a GWAS in Frizarta dairy sheep with phenotypic data from 10 body size measurements and genotypic data (from Illumina ovineSNP50 BeadChip) for 459 ewes. Results The 10 body size measurements were subjected to principal component analysis and three independent principal components (PC) were constructed, interpretable as width, height and length dimensions, respectively. The GWAS performed for each PC identified 11 significant SNPs, at the chromosome level, one on each of the chromosomes 3, 8, 9, 10, 11, 12, 19, 20, 23 and two on chromosome 25. Nine out of the 11 SNPs were located on previously identified quantitative trait loci for sheep meat, production or reproduction. One hundred and ninety-seven positional candidate genes within a 1-Mb distance from each significant SNP were found. A guilt-by-association-based (GBA) prioritization analysis (PA) was performed to identify the most plausible functional candidate genes. GBA-based PA identified 39 genes that were significantly associated with gene networks relevant to body size traits. Prioritized genes were identified in the vicinity of all significant SNPs except for those on chromosomes 10 and 12. The top five ranking genes were TP53, BMPR1A, PIK3R5, RPL26 and PRKDC. Conclusions The results of this GWAS provide evidence for 39 causative candidate genes across nine chromosomal regions for body size traits, some of which are novel and some are previously identified candidates from other studies (e.g. TP53, NTN1 and ZNF521). GBA-based PA has proved to be a useful tool to identify genes with increased biological relevance but it is subjected to certain limitations. Electronic supplementary material The online version of this article (doi:10.1186/s12711-017-0316-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Antonios Kominakis
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece
| | - Ariadne L Hager-Theodorides
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece.
| | - Evangelos Zoidis
- Department of Animal Science and Aquaculture, Agricultural University of Athens, Iera Odos 75, 11855, Athens, Greece
| | - Aggeliki Saridaki
- Department of Environmental and Natural Resources Management, University of Patras, Seferi 2, 30100, Agrinio, Greece
| | - George Antonakos
- Agricultural and Livestock Union of Western Greece, 13rd Km N.R. Agrinio-Ioannina, 30100, Lepenou, Greece
| | - George Tsiamis
- Department of Environmental and Natural Resources Management, University of Patras, Seferi 2, 30100, Agrinio, Greece
| |
Collapse
|
36
|
Chen J, Crutchley J, Zhang D, Owzar K, Kastan MB. Identification of a DNA Damage-Induced Alternative Splicing Pathway That Regulates p53 and Cellular Senescence Markers. Cancer Discov 2017; 7:766-781. [PMID: 28288992 DOI: 10.1158/2159-8290.cd-16-0908] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/14/2016] [Accepted: 03/09/2017] [Indexed: 01/10/2023]
Abstract
Cellular responses to DNA damage are critical determinants of cancer development and aging-associated pathogenesis. Here, we identify and characterize a DNA-damage response (DDR) pathway that regulates alternative splicing of numerous gene products, including the human tumor suppressor TP53, and controls DNA damage-induced cellular senescence. In brief, ionizing radiation (IR) inhibits the activity of SMG1, a phosphoinositide-3-kinase-like kinase family member, reducing the binding of SMG1 to a specific region near exon 9 of p53 precursor mRNA and promoting the binding of ribosomal protein L26 (RPL26) to p53 pre-mRNA. RPL26, in turn, is required for the recruitment of the serine/arginine-rich splicing factor SRSF7 to p53 pre-mRNA and generation of alternatively spliced p53β RNA. Disruption of this pathway via selective knockout of p53β by CRISPR/Cas9 or downregulation of pathway constituents significantly reduces IR-induced senescence markers, and cells lacking p53β expression fail to transcriptionally repress negative regulators of cellular senescence and aging.Significance: We identified a new component of the DDR pathway that regulates alternative splicing of messenger RNAs, including human TP53 mRNA. Modulation of this regulatory pathway affects DNA-damage induction of cellular senescence markers. Cancer Discov; 7(7); 766-81. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 653.
Collapse
Affiliation(s)
- Jing Chen
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina.,Duke Cancer Institute, Duke University, Durham, North Carolina
| | - John Crutchley
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina.,Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Dadong Zhang
- Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Kouros Owzar
- Duke Cancer Institute, Duke University, Durham, North Carolina.,Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Michael B Kastan
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina. .,Duke Cancer Institute, Duke University, Durham, North Carolina
| |
Collapse
|
37
|
Martínez P, Blasco MA. Telomere-driven diseases and telomere-targeting therapies. J Cell Biol 2017; 216:875-887. [PMID: 28254828 PMCID: PMC5379954 DOI: 10.1083/jcb.201610111] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/03/2017] [Accepted: 01/09/2017] [Indexed: 01/19/2023] Open
Abstract
Martínez and Blasco review the molecular mechanisms underlying diseases associated with telomere dysfunction, including telomeropathies, age-related disorders, and cancer. Current and future therapeutic strategies to treat and prevent these diseases, including preclinical development of telomere-targeted therapies using mouse models, are discussed. Telomeres, the protective ends of linear chromosomes, shorten throughout an individual’s lifetime. Telomere shortening is proposed to be a primary molecular cause of aging. Short telomeres block the proliferative capacity of stem cells, affecting their potential to regenerate tissues, and trigger the development of age-associated diseases. Mutations in telomere maintenance genes are associated with pathologies referred to as telomere syndromes, including Hoyeraal-Hreidarsson syndrome, dyskeratosis congenita, pulmonary fibrosis, aplastic anemia, and liver fibrosis. Telomere shortening induces chromosomal instability that, in the absence of functional tumor suppressor genes, can contribute to tumorigenesis. In addition, mutations in telomere length maintenance genes and in shelterin components, the protein complex that protects telomeres, have been found to be associated with different types of cancer. These observations have encouraged the development of therapeutic strategies to treat and prevent telomere-associated diseases, namely aging-related diseases, including cancer. Here we review the molecular mechanisms underlying telomere-driven diseases and highlight recent advances in the preclinical development of telomere-targeted therapies using mouse models.
Collapse
Affiliation(s)
- Paula Martínez
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre, Madrid E-28029, Spain
| | - Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre, Madrid E-28029, Spain
| |
Collapse
|
38
|
Abstract
The p53 tumor suppressor has been studied for decades, and still there are many questions left unanswered. In this review, we first describe the current understanding of the wild-type p53 functions that determine cell survival or death, and regulation of the protein, with a particular focus on the negative regulators, the murine double minute family of proteins. We also summarize tissue-, stress-, and age-specific p53 activities and the potential underlying mechanisms. Among all p53 gene alterations identified in human cancers, p53 missense mutations predominate, suggesting an inherent biological advantage. Numerous gain-of-function activities of mutant p53 in different model systems and contexts have been identified. The emerging theme is that mutant p53, which retains a potent transcriptional activation domain, also retains the ability to modify gene transcription, albeit indirectly. Lastly, because mutant p53 stability is necessary for its gain of function, we summarize the mechanisms through which mutant p53 is specifically stabilized. A deeper understanding of the multiple pathways that impinge upon wild-type and mutant p53 activities and how these, in turn, regulate cell behavior will help identify vulnerabilities and therapeutic opportunities.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Guillermina Lozano
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
39
|
Filipescu D, Naughtin M, Podsypanina K, Lejour V, Wilson L, Gurard-Levin ZA, Orsi GA, Simeonova I, Toufektchan E, Attardi LD, Toledo F, Almouzni G. Essential role for centromeric factors following p53 loss and oncogenic transformation. Genes Dev 2017; 31:463-480. [PMID: 28356341 PMCID: PMC5393061 DOI: 10.1101/gad.290924.116] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/24/2017] [Indexed: 11/25/2022]
Abstract
In mammals, centromere definition involves the histone variant CENP-A (centromere protein A), deposited by its chaperone, HJURP (Holliday junction recognition protein). Alterations in this process impair chromosome segregation and genome stability, which are also compromised by p53 inactivation in cancer. Here we found that CENP-A and HJURP are transcriptionally up-regulated in p53-null human tumors. Using an established mouse embryonic fibroblast (MEF) model combining p53 inactivation with E1A or HRas-V12 oncogene expression, we reproduced a similar up-regulation of HJURP and CENP-A. We delineate functional CDE/CHR motifs within the Hjurp and Cenpa promoters and demonstrate their roles in p53-mediated repression. To assess the importance of HJURP up-regulation in transformed murine and human cells, we used a CRISPR/Cas9 approach. Remarkably, depletion of HJURP leads to distinct outcomes depending on their p53 status. Functional p53 elicits a cell cycle arrest response, whereas, in p53-null transformed cells, the absence of arrest enables the loss of HJURP to induce severe aneuploidy and, ultimately, apoptotic cell death. We thus tested the impact of HJURP depletion in pre-established allograft tumors in mice and revealed a major block of tumor progression in vivo. We discuss a model in which an "epigenetic addiction" to the HJURP chaperone represents an Achilles' heel in p53-deficient transformed cells.
Collapse
Affiliation(s)
- Dan Filipescu
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, UMR3664, Centre Nationnal de la Recherche Scientifique (CNRS), Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Université Paris 06, UMR3664, CNRS, F-75005 Paris, France
| | - Monica Naughtin
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, UMR3664, Centre Nationnal de la Recherche Scientifique (CNRS), Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Université Paris 06, UMR3664, CNRS, F-75005 Paris, France
| | - Katrina Podsypanina
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, UMR3664, Centre Nationnal de la Recherche Scientifique (CNRS), Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Université Paris 06, UMR3664, CNRS, F-75005 Paris, France
| | - Vincent Lejour
- Institut Curie, PSL Research University, UMR3244, CNRS, Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France
- Sorbonne Universités, UPMC Université Paris 06, UMR3244, CNRS, F-75005 Paris, France
| | - Laurence Wilson
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, UMR3664, Centre Nationnal de la Recherche Scientifique (CNRS), Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Université Paris 06, UMR3664, CNRS, F-75005 Paris, France
| | - Zachary A Gurard-Levin
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, UMR3664, Centre Nationnal de la Recherche Scientifique (CNRS), Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Université Paris 06, UMR3664, CNRS, F-75005 Paris, France
| | - Guillermo A Orsi
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, UMR3664, Centre Nationnal de la Recherche Scientifique (CNRS), Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Université Paris 06, UMR3664, CNRS, F-75005 Paris, France
| | - Iva Simeonova
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, UMR3664, Centre Nationnal de la Recherche Scientifique (CNRS), Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Université Paris 06, UMR3664, CNRS, F-75005 Paris, France
| | - Eleonore Toufektchan
- Institut Curie, PSL Research University, UMR3244, CNRS, Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France
- Sorbonne Universités, UPMC Université Paris 06, UMR3244, CNRS, F-75005 Paris, France
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Franck Toledo
- Institut Curie, PSL Research University, UMR3244, CNRS, Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France
- Sorbonne Universités, UPMC Université Paris 06, UMR3244, CNRS, F-75005 Paris, France
| | - Geneviève Almouzni
- Institut Curie, Paris Sciences et Lettres (PSL) Research University, UMR3664, Centre Nationnal de la Recherche Scientifique (CNRS), Equipe Labellisée Ligue contre le Cancer, F-75005 Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Université Paris 06, UMR3664, CNRS, F-75005 Paris, France
| |
Collapse
|
40
|
Abstract
MDM4, an essential negative regulator of the P53 tumor suppressor, is frequently overexpressed in cancer cells that harbor a wild-type P53. By a mechanism based on alternative splicing, the MDM4 gene generates two mutually exclusive isoforms: MDM4-FL, which encodes the full-length MDM4 protein, and a shorter splice variant called MDM4-S. Previous results suggested that the MDM4-S isoform could be an important driver of tumor development. In this short review, we discuss a recent set of data indicating that MDM4-S is more likely a passenger isoform during tumorigenesis and that targeting MDM4 splicing to prevent MDM4-FL protein expression appears as a promising strategy to reactivate p53 in cancer cells. The benefits and risks associated with this strategy are also discussed.
Collapse
|
41
|
Toufektchan E, Jaber S, Toledo F. [Dangerous liaisons: p53, dyskeratosis congenita and Fanconi anemia]. Med Sci (Paris) 2017; 33:95-98. [PMID: 28120765 DOI: 10.1051/medsci/20173301018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Eléonore Toufektchan
- Génétique de la suppression tumorale, Équipe Labellisée Ligue, Institut Curie, Centre de recherche, 26, rue d'Ulm, 75248 Paris Cedex 05, France - Sorbonne Universités, UPMC, Université Paris 6, Paris, France - CNRS UMR 3244, Paris, France - PSL Research University, Paris, France
| | - Sara Jaber
- Génétique de la suppression tumorale, Équipe Labellisée Ligue, Institut Curie, Centre de recherche, 26, rue d'Ulm, 75248 Paris Cedex 05, France - Sorbonne Universités, UPMC, Université Paris 6, Paris, France - CNRS UMR 3244, Paris, France - PSL Research University, Paris, France
| | - Franck Toledo
- Génétique de la suppression tumorale, Équipe Labellisée Ligue, Institut Curie, Centre de recherche, 26, rue d'Ulm, 75248 Paris Cedex 05, France - Sorbonne Universités, UPMC, Université Paris 6, Paris, France - CNRS UMR 3244, Paris, France - PSL Research University, Paris, France
| |
Collapse
|
42
|
Barton MC. Acidic shield puts a chink in p53's armour. Nature 2016; 538:45-46. [DOI: 10.1038/nature19469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
43
|
Laptenko O, Tong DR, Manfredi J, Prives C. The Tail That Wags the Dog: How the Disordered C-Terminal Domain Controls the Transcriptional Activities of the p53 Tumor-Suppressor Protein. Trends Biochem Sci 2016; 41:1022-1034. [PMID: 27669647 DOI: 10.1016/j.tibs.2016.08.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 01/22/2023]
Abstract
The p53 tumor suppressor is a transcription factor (TF) that exerts antitumor functions through its ability to regulate the expression of multiple genes. Within the p53 protein resides a relatively short unstructured C-terminal domain (CTD) that remarkably participates in virtually every aspect of p53 performance as a TF. Because these aspects are often interdependent and it is not always possible to dissect them experimentally, there has been a great deal of controversy about the CTD. In this review we evaluate the significance and key features of this interesting region of p53 and its impact on the many aspects of p53 function in light of previous and more recent findings.
Collapse
Affiliation(s)
- Oleg Laptenko
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - David R Tong
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - James Manfredi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
44
|
Wang D, Kon N, Lasso G, Jiang L, Leng W, Zhu WG, Qin J, Honig B, Gu W. Acetylation-regulated interaction between p53 and SET reveals a widespread regulatory mode. Nature 2016; 538:118-122. [PMID: 27626385 PMCID: PMC5333498 DOI: 10.1038/nature19759] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 08/19/2016] [Indexed: 12/14/2022]
Abstract
Although lysine acetylation is now recognized as a general protein
modification for both histones and non-histone proteins1-3, the mechanisms of acetylation mediated actions are not
completely understood. Acetylation of the C-terminal domain (CTD) of p53 was the
first example for non-histone protein acetylation4. Yet the precise role of the CTD acetylation
remains elusive. Lysine acetylation often creates binding sites for
bromodomain-containing “reader” proteins5,6;
surprisingly, in a proteomic screen, we identified SET as a major cellular
factor whose binding with p53 is totally dependent on the CTD acetylation
status. SET profoundly inhibits p53 transcriptional activity in unstressed cells
but SET-mediated repression is completely abolished by stress-induced p53 CTD
acetylation. Moreover, loss of the interaction with SET activates p53, resulting
in tumor regression in mouse xenograft models. Notably, the acidic domain of SET
acts as a “reader” for unacetylated CTD of p53 and this mechanism
of acetylation-dependent regulation is widespread in nature. For example, p53
acetylation also modulates its interactions with similar acidic domains found in
other p53 regulators including VPRBP, DAXX and PELP1 (refs. 7-9),
and computational analysis of the proteome identified numerous proteins with the
potential to serve as the acidic domain readers and lysine-rich ligands. Unlike
bromodomain readers, which preferentially bind the acetylated forms of their
cognate ligands, the acidic domain readers specifically recognize the
unacetylated forms of their ligands. Finally, the acetylation-dependent
regulation of p53 was further validated in vivo by using a
knockin mouse model expressing an acetylation-mimicking form of p53. These
results reveal that the acidic domain-containing factors act as a new class of
acetylation-dependent regulators by targeting p53 and potentially, beyond.
Collapse
Affiliation(s)
- Donglai Wang
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
| | - Ning Kon
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
| | - Gorka Lasso
- Department of Biochemistry and Molecular Biophysics and Systems Biology, Center for Computational Biology and Bioinformatics, Howard Hughes Medical Institute, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
| | - Le Jiang
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
| | - Wenchuan Leng
- State Key Laboratory of Proteomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, Shenzhen University School of Medicine, Shenzhen 518060, China
| | - Jun Qin
- State Key Laboratory of Proteomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China.,Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Barry Honig
- Department of Biochemistry and Molecular Biophysics and Systems Biology, Center for Computational Biology and Bioinformatics, Howard Hughes Medical Institute, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY 10032, USA
| |
Collapse
|
45
|
Lieberman PM. Retrotransposon-derived p53 binding sites enhance telomere maintenance and genome protection. Bioessays 2016; 38:943-9. [PMID: 27539745 DOI: 10.1002/bies.201600078] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Tumor suppressor protein 53 (p53) plays a central role in the control of genome stability, acting primarily through the transcriptional activation of stress-response genes. However, many p53 binding sites are located at genomic locations with no obvious regulatory-link to known stress-response genes. We recently discovered p53 binding sites within retrotransposon-derived elements in human and mouse subtelomeres. These retrotransposon-derived p53 binding sites protected chromosome ends through transcription activation of telomere repeat RNA, as well as through the direct modification of local chromatin structure in response to DNA damage. Based on these findings, I hypothesize that a class of p53 binding sites, including the retrotransposon-derived p53-sites found in subtlomeres, provide a primary function in genome stability by mounting a direct and local protective chromatin-response to DNA damage. I speculate that retrotransposon-derived p53 binding sites share features with telomere-repeats through an evolutionary drive to monitor and maintain genome integrity.
Collapse
|
46
|
Toledo F. p53: A two-faced regulator of telomere metabolism? (comment on DOI 10.1002/bies.201600078). Bioessays 2016; 38:938. [PMID: 27538365 DOI: 10.1002/bies.201600149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Franck Toledo
- Genetics of Tumor Suppression - Equipe Labellisée Ligue, Institut Curie, Centre de Recherche - PSL Research University, Sorbonne Universités, Paris, France.
| |
Collapse
|
47
|
You L, Li L, Zou J, Yan K, Belle J, Nijnik A, Wang E, Yang XJ. BRPF1 is essential for development of fetal hematopoietic stem cells. J Clin Invest 2016; 126:3247-62. [PMID: 27500495 DOI: 10.1172/jci80711] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 06/16/2016] [Indexed: 12/21/2022] Open
Abstract
Hematopoietic stem cells (HSCs) serve as a life-long reservoir for all blood cell types and are clinically useful for a variety of HSC transplantation-based therapies. Understanding the role of chromatin organization and regulation in HSC homeostasis may provide important insights into HSC development. Bromodomain- and PHD finger-containing protein 1 (BRPF1) is a multivalent chromatin regulator that possesses 4 nucleosome-binding domains and activates 3 lysine acetyltransferases (KAT6A, KAT6B, and KAT7), suggesting that this protein has the potential to stimulate crosstalk between different chromatin modifications. Here, we investigated the function of BRPF1 in hematopoiesis by selectively deleting its gene in murine blood cells. Brpf1-deficient pups experienced early lethality due to acute bone marrow failure and aplastic anemia. The mutant bone marrow and fetal liver exhibited severe deficiency in HSCs and hematopoietic progenitors, along with elevated reactive oxygen species, senescence, and apoptosis. BRPF1 deficiency also reduced the expression of multipotency genes, including Slamf1, Mecom, Hoxa9, Hlf, Gfi1, Egr, and Gata3. Furthermore, BRPF1 was required for acetylation of histone H3 at lysine 23, a highly abundant but not well-characterized epigenetic mark. These results identify an essential role of the multivalent chromatin regulator BRPF1 in definitive hematopoiesis and illuminate a potentially new avenue for studying epigenetic networks that govern HSC ontogeny.
Collapse
|
48
|
Abstract
It is poorly understood how a single protein, p53, can be responsive to so many stress signals and orchestrates very diverse cell responses to maintain/restore cell/tissue functions. The uncovering that TP53 gene physiologically expresses, in a tissue-dependent manner, several p53 splice variants (isoforms) provides an explanation to its pleiotropic biological activities. Here, we summarize a decade of research on p53 isoforms. The clinical studies and the diverse cellular and animal models of p53 isoforms (zebrafish, Drosophila, and mouse) lead us to realize that a p53-mediated cell response is, in fact, the sum of the intrinsic activities of the coexpressed p53 isoforms and that unbalancing expression of different p53 isoforms leads to cancer, premature aging, (neuro)degenerative diseases, inflammation, embryo malformations, or defects in tissue regeneration. Cracking the p53 isoforms' code is, thus, a necessary step to improve cancer treatment. It also opens new exciting perspectives in tissue regeneration.
Collapse
Affiliation(s)
- Sebastien M Joruiz
- Dundee Cancer Centre, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom
| | - Jean-Christophe Bourdon
- Dundee Cancer Centre, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom
| |
Collapse
|
49
|
Laptenko O, Shiff I, Freed-Pastor W, Zupnick A, Mattia M, Freulich E, Shamir I, Kadouri N, Kahan T, Manfredi J, Simon I, Prives C. The p53 C terminus controls site-specific DNA binding and promotes structural changes within the central DNA binding domain. Mol Cell 2016; 57:1034-1046. [PMID: 25794615 DOI: 10.1016/j.molcel.2015.02.015] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/23/2014] [Accepted: 02/06/2015] [Indexed: 11/19/2022]
Abstract
DNA binding by numerous transcription factors including the p53 tumor suppressor protein constitutes a vital early step in transcriptional activation. While the role of the central core DNA binding domain (DBD) of p53 in site-specific DNA binding has been established, the contribution of the sequence-independent C-terminal domain (CTD) is still not well understood. We investigated the DNA-binding properties of a series of p53 CTD variants using a combination of in vitro biochemical analyses and in vivo binding experiments. Our results provide several unanticipated and interconnected findings. First, the CTD enables DNA binding in a sequence-dependent manner that is drastically altered by either its modification or deletion. Second, dependence on the CTD correlates with the extent to which the p53 binding site deviates from the canonical consensus sequence. Third, the CTD enables stable formation of p53-DNA complexes to divergent binding sites via DNA-induced conformational changes within the DBD itself.
Collapse
Affiliation(s)
- Oleg Laptenko
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Idit Shiff
- Department of Microbiology and Molecular Genetics, Hebrew University Medical School, IMRIC, Jerusalem 91120, Israel
| | - Will Freed-Pastor
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Andrew Zupnick
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Melissa Mattia
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Ella Freulich
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Inbal Shamir
- Department of Microbiology and Molecular Genetics, Hebrew University Medical School, IMRIC, Jerusalem 91120, Israel
| | - Noam Kadouri
- Department of Microbiology and Molecular Genetics, Hebrew University Medical School, IMRIC, Jerusalem 91120, Israel
| | - Tamar Kahan
- Department of Microbiology and Molecular Genetics, Hebrew University Medical School, IMRIC, Jerusalem 91120, Israel
| | - James Manfredi
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Itamar Simon
- Department of Microbiology and Molecular Genetics, Hebrew University Medical School, IMRIC, Jerusalem 91120, Israel.
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
50
|
Barthelery NJ, Manfredi JJ. Cerebellum Development and Tumorigenesis: A p53-Centric Perspective. Trends Mol Med 2016; 22:404-413. [PMID: 27085812 DOI: 10.1016/j.molmed.2016.03.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/19/2016] [Accepted: 03/19/2016] [Indexed: 12/30/2022]
Abstract
The p53 protein has been extensively studied for its role in suppressing tumorigenesis, in part through surveillance and maintenance of genomic stability. p53 has been associated with the induction of a variety of cellular outcomes including cell cycle arrest, senescence, and apoptosis. This occurs primarily, but not exclusively, through transcriptional activation of specific target genes. By contrast, the participation of p53 in normal developmental processes has been largely understudied. This review focuses on possible functions of p53 in cerebellar development. It can be argued that a better understanding of such mechanisms will provide needed insight into the genesis of certain embryonic cancers including medulloblastomas, and thus lead to more effective therapies.
Collapse
Affiliation(s)
- Nicolas J Barthelery
- Department of Oncological Sciences and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - James J Manfredi
- Department of Oncological Sciences and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA.
| |
Collapse
|