1
|
Levine AJ, Carpten JD, Murphy M, Hainaut P. Exploring the genetic and molecular basis of differences in multiple myeloma of individuals of African and European descent. Cell Death Differ 2024; 31:1-8. [PMID: 38001255 PMCID: PMC10781774 DOI: 10.1038/s41418-023-01236-8] [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: 08/02/2023] [Revised: 10/08/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Multiple Myeloma is a typical example of a neoplasm that shows significant differences in incidence, age of onset, type, and frequency of genetic alterations between patients of African and European ancestry. This perspective explores the hypothesis that both genetic polymorphisms and spontaneous somatic mutations in the TP53 tumor suppressor gene are determinants of these differences. In the US, the rates of occurrence of MM are at least twice as high in African Americans (AA) as in Caucasian Americans (CA). Strikingly, somatic TP53 mutations occur in large excess (at least 4-6-fold) in CA versus AA. On the other hand, TP53 contains polymorphisms specifying amino-acid differences that are under natural selection by the latitude of a population and have evolved during the migrations of humans over several hundred thousand years. The p53 protein plays important roles in DNA strand break repair and, therefore, in the surveillance of aberrant DNA recombination, leading to the B-cell translocations that are causal in the pathogenesis of MM. We posit that polymorphisms in one region of the TP53 gene (introns 2 and 3, and the proline-rich domain) specify a concentration of the p53 protein with a higher capacity to repress translocations in CA than AA patients. This, in turn, results in a higher risk of acquiring inactivating, somatic mutations in a different region of the TP53 gene (DNA binding domain) in CA than in AA patients. Such a mechanism, by which the polymorphic status of a gene influencing its own "spontaneous" mutation frequency, may provide a genetic basis to address ethnicity-related differences in the incidence and phenotypes of many different forms of cancer.
Collapse
Affiliation(s)
- Arnold J Levine
- Simons Center for Systems Biology, Institute for Advanced Study, Princeton, NJ, USA.
| | - John D Carpten
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Pierre Hainaut
- Institute for Advanced Biosciences, Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
2
|
Huang Y, Roig I. Genetic control of meiosis surveillance mechanisms in mammals. Front Cell Dev Biol 2023; 11:1127440. [PMID: 36910159 PMCID: PMC9996228 DOI: 10.3389/fcell.2023.1127440] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Meiosis is a specialized cell division that generates haploid gametes and is critical for successful sexual reproduction. During the extended meiotic prophase I, homologous chromosomes progressively pair, synapse and desynapse. These chromosomal dynamics are tightly integrated with meiotic recombination (MR), during which programmed DNA double-strand breaks (DSBs) are formed and subsequently repaired. Consequently, parental chromosome arms reciprocally exchange, ultimately ensuring accurate homolog segregation and genetic diversity in the offspring. Surveillance mechanisms carefully monitor the MR and homologous chromosome synapsis during meiotic prophase I to avoid producing aberrant chromosomes and defective gametes. Errors in these critical processes would lead to aneuploidy and/or genetic instability. Studies of mutation in mouse models, coupled with advances in genomic technologies, lead us to more clearly understand how meiosis is controlled and how meiotic errors are linked to mammalian infertility. Here, we review the genetic regulations of these major meiotic events in mice and highlight our current understanding of their surveillance mechanisms. Furthermore, we summarize meiotic prophase genes, the mutations that activate the surveillance system leading to meiotic prophase arrest in mouse models, and their corresponding genetic variants identified in human infertile patients. Finally, we discuss their value for the diagnosis of causes of meiosis-based infertility in humans.
Collapse
Affiliation(s)
- Yan Huang
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Histology Unit, Department of Cell Biology, Physiology, and Immunology, Cytology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ignasi Roig
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Histology Unit, Department of Cell Biology, Physiology, and Immunology, Cytology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
3
|
p53 Controls Meiotic Prophase Progression and Crossover Formation. Int J Mol Sci 2022; 23:ijms23179818. [PMID: 36077210 PMCID: PMC9456223 DOI: 10.3390/ijms23179818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
Meiosis initiates with the formation of double strand breaks (DSBs) throughout the genome. To avoid genomic instability, these DSBs need to be correctly repaired by homologous recombination. Surveillance mechanisms involving the DNA damage response (DDR) pathway ATM-CHK2-p53 can detect the persistence of unrepaired DBSs and activate the recombination-dependent arrest at the pachytene stage. However, a complete understanding of p53 functions under normal physiological conditions remains lacking. Here, we report a detailed analysis of the p53 role during meiotic prophase in mice spermatocytes. We show that the absence of p53 regulates prophase progression by slowing down the pachytene stage when the recombination-dependent arrest occurs. Furthermore, our results show that p53 is necessary for proper crossover (CO) formation and localization. Our study contributes to a deeper understanding of p53 roles during the meiotic prophase.
Collapse
|
4
|
Wylie A, Jones AE, Das S, Lu WJ, Abrams JM. Distinct p53 isoforms code for opposing transcriptional outcomes. Dev Cell 2022; 57:1833-1846.e6. [PMID: 35820415 PMCID: PMC9378576 DOI: 10.1016/j.devcel.2022.06.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/15/2022] [Accepted: 06/15/2022] [Indexed: 12/19/2022]
Abstract
p53 genes are conserved transcriptional activators that respond to stress. These proteins can also downregulate genes, but the mechanisms are not understood and are generally assumed to be indirect. Here, we investigate synthetic and native cis-regulatory elements in Drosophila to examine opposing features of p53-mediated transcriptional control in vivo. We show that transcriptional repression by p53 operates continuously through canonical DNA binding sites that confer p53-dependent transactivation at earlier developmental stages. p53 transrepression is correlated with local H3K9me3 chromatin marks and occurs without the need for stress or Chk2. In sufficiency tests, two p53 isoforms qualify as transrepressors and a third qualifies as a transcriptional activator. Targeted isoform-specific knockouts dissociate these opposing transcriptional activities, highlighting features that are dispensable for transactivation but critical for repression and for proper germ cell formation. Together, these results demonstrate that certain p53 isoforms function as constitutive tissue-specific repressors, raising important implications for tumor suppression by the human counterpart.
Collapse
Affiliation(s)
- Annika Wylie
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Amanda E Jones
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Simanti Das
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wan-Jin Lu
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John M Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
5
|
Liu C, Moten A, Ma Z, Lin HK. The foundational framework of tumors: Gametogenesis, p53, and cancer. Semin Cancer Biol 2022; 81:193-205. [PMID: 33940178 PMCID: PMC9382687 DOI: 10.1016/j.semcancer.2021.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022]
Abstract
The completion-of-tumor hypothesis involved in the dynamic interplay between the initiating oncogenic event and progression is essential to better recognize the foundational framework of tumors. Here we review and extend the gametogenesis-related hypothesis of tumors, because high embryonic/germ cell traits are common in tumors. The century-old gametogenesis-related hypothesis of tumors postulated that tumors arise from displaced/activated trophoblasts, displaced (lost) germ cells, and the reprogramming/reactivation of gametogenic program in somatic cells. Early primordial germ cells (PGCs), embryonic stem (ES) cells, embryonic germ cells (EGCs), and pre-implantation embryos at the stage from two-cell stage to blastocysts originating from fertilization or parthenogenesis have the potential to develop teratomas/teratocarcinomas. In addition, the teratomas/teratocarcinomas/germ cells occur in gonads and extra-gonads. Undoubtedly, the findings provide strong support for the hypothesis. However, it was thought that these tumor types were an exception rather than verification. In fact, there are extensive similarities between somatic tumor types and embryonic/germ cell development, such as antigens, migration, invasion, and immune escape. It was documented that embryonic/germ cell genes play crucial roles in tumor behaviors, e.g. tumor initiation and metastasis. Of note, embryonic/germ cell-like tumor cells at different developmental stages including PGC and oocyte to the early embryo-like stage were identified in diverse tumor types by our group. These embryonic/germ cell-like cancer cells resemble the natural embryonic/germ cells in morphology, gene expression, the capability of teratoma formation, and the ability to undergo the process of oocyte maturation and parthenogenesis. These embryonic/germ cell-like cancer cells are derived from somatic cells and contribute to tumor formation, metastasis, and drug resistance, establishing asexual meiotic embryonic life cycle. p53 inhibits the reactivation of embryonic/germ cell state in somatic cells and oocyte-like cell maturation. Based on earlier and our recent studies, we propose a novel model to complete the gametogenesis-related hypothesis of tumors, which can be applied to certain somatic tumors. That is, tumors tend to establish a somatic asexual meiotic embryonic cycle through the activation of somatic female gametogenesis and parthenogenesis in somatic tumor cells during the tumor progression, thus passing on corresponding embryonic/germ cell traits leading to the malignant behaviors and enhancing the cells' independence. This concept may be instrumental to better understand the nature and evolution of tumors. We rationalize that targeting the key events of somatic pregnancy is likely a better therapeutic strategy for cancer treatment than directly targeting cell mitotic proliferation, especially for those tumors with p53 inactivation.
Collapse
Affiliation(s)
- Chunfang Liu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
| | - Asad Moten
- Medical Sciences Division, University of Oxford, Oxford OX3 9DU, UK
| | - Zhan Ma
- Department of Laboratory Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
6
|
Martin ET, Blatt P, Nguyen E, Lahr R, Selvam S, Yoon HAM, Pocchiari T, Emtenani S, Siekhaus DE, Berman A, Fuchs G, Rangan P. A translation control module coordinates germline stem cell differentiation with ribosome biogenesis during Drosophila oogenesis. Dev Cell 2022; 57:883-900.e10. [PMID: 35413237 PMCID: PMC9011129 DOI: 10.1016/j.devcel.2022.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 01/11/2022] [Accepted: 03/10/2022] [Indexed: 01/26/2023]
Abstract
Ribosomal defects perturb stem cell differentiation, and this is the cause of ribosomopathies. How ribosome levels control stem cell differentiation is not fully known. Here, we discover that three DExD/H-box proteins govern ribosome biogenesis (RiBi) and Drosophila oogenesis. Loss of these DExD/H-box proteins, which we name Aramis, Athos, and Porthos, aberrantly stabilizes p53, arrests the cell cycle, and stalls germline stem cell (GSC) differentiation. Aramis controls cell-cycle progression by regulating translation of mRNAs that contain a terminal oligo pyrimidine (TOP) motif in their 5' UTRs. We find that TOP motifs confer sensitivity to ribosome levels that are mediated by La-related protein (Larp). One such TOP-containing mRNA codes for novel nucleolar protein 1 (Non1), a conserved p53 destabilizing protein. Upon a sufficient ribosome concentration, Non1 is expressed, and it promotes GSC cell-cycle progression via p53 degradation. Thus, a previously unappreciated TOP motif in Drosophila responds to reduced RiBi to co-regulate the translation of ribosomal proteins and a p53 repressor, coupling RiBi to GSC differentiation.
Collapse
Affiliation(s)
- Elliot T Martin
- Department of Biological Sciences/RNA Institute, University at Albany, SUNY, Albany, NY 12202, USA
| | - Patrick Blatt
- Department of Biological Sciences/RNA Institute, University at Albany, SUNY, Albany, NY 12202, USA
| | - Elaine Nguyen
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Roni Lahr
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Sangeetha Selvam
- Department of Biological Sciences/RNA Institute, University at Albany, SUNY, Albany, NY 12202, USA
| | - Hyun Ah M Yoon
- Department of Biological Sciences/RNA Institute, University at Albany, SUNY, Albany, NY 12202, USA; Albany Medical College, Albany, NY 12208, USA
| | - Tyler Pocchiari
- Department of Biological Sciences/RNA Institute, University at Albany, SUNY, Albany, NY 12202, USA; SUNY Upstate Medical University, Syracuse, NY 13210-2375, USA
| | - Shamsi Emtenani
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Daria E Siekhaus
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Andrea Berman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Gabriele Fuchs
- Department of Biological Sciences/RNA Institute, University at Albany, SUNY, Albany, NY 12202, USA.
| | - Prashanth Rangan
- Department of Biological Sciences/RNA Institute, University at Albany, SUNY, Albany, NY 12202, USA.
| |
Collapse
|
7
|
Chakravarti A, Thirimanne HN, Brown S, Calvi BR. Drosophila p53 isoforms have overlapping and distinct functions in germline genome integrity and oocyte quality control. eLife 2022; 11:61389. [PMID: 35023826 PMCID: PMC8758136 DOI: 10.7554/elife.61389] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/13/2021] [Indexed: 12/13/2022] Open
Abstract
p53 gene family members in humans and other organisms encode a large number of protein isoforms whose functions are largely undefined. Using Drosophila as a model, we find that a p53B isoform is expressed predominantly in the germline where it colocalizes with p53A into subnuclear bodies. It is only p53A, however, that mediates the apoptotic response to ionizing radiation in the germline and soma. In contrast, p53A and p53B are both required for the normal repair of meiotic DNA breaks, an activity that is more crucial when meiotic recombination is defective. We find that in oocytes with persistent DNA breaks p53A is also required to activate a meiotic pachytene checkpoint. Our findings indicate that Drosophila p53 isoforms have DNA lesion and cell type-specific functions, with parallels to the functions of mammalian p53 family members in the genotoxic stress response and oocyte quality control.
Collapse
Affiliation(s)
| | | | - Savanna Brown
- Department of Biology, Indiana University, Bloomington, United States
| | - Brian R Calvi
- Department of Biology, Indiana University, Bloomington, United States
| |
Collapse
|
8
|
McDonald JI, Diab N, Arthofer E, Hadley M, Kanholm T, Rentia U, Gomez S, Yu A, Grundy EE, Cox O, Topper MJ, Xing X, Strissel PL, Strick R, Wang T, Baylin SB, Chiappinelli KB. Epigenetic Therapies in Ovarian Cancer Alter Repetitive Element Expression in a TP53-Dependent Manner. Cancer Res 2021; 81:5176-5189. [PMID: 34433584 PMCID: PMC8530980 DOI: 10.1158/0008-5472.can-20-4243] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/15/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022]
Abstract
Epithelial ovarian carcinomas are particularly deadly due to intratumoral heterogeneity, resistance to standard-of-care therapies, and poor response to alternative treatments such as immunotherapy. Targeting the ovarian carcinoma epigenome with DNA methyltransferase inhibitors (DNMTi) or histone deacetylase inhibitors (HDACi) increases immune signaling and recruits CD8+ T cells and natural killer cells to fight ovarian carcinoma in murine models. This increased immune activity is caused by increased transcription of repetitive elements (RE) that form double-stranded RNA (dsRNA) and trigger an IFN response. To understand which REs are affected by epigenetic therapies in ovarian carcinoma, we assessed the effect of DNMTi and HDACi on ovarian carcinoma cell lines and patient samples. Subfamily-level (TEtranscripts) and individual locus-level (Telescope) analysis of REs showed that DNMTi treatment upregulated more REs than HDACi treatment. Upregulated REs were predominantly LTR and SINE subfamilies, and SINEs exhibited the greatest loss of DNA methylation upon DNMTi treatment. Cell lines with TP53 mutations exhibited significantly fewer upregulated REs with epigenetic therapy than wild-type TP53 cell lines. This observation was validated using isogenic cell lines; the TP53-mutant cell line had significantly higher baseline expression of REs but upregulated fewer upon epigenetic treatment. In addition, p53 activation increased expression of REs in wild-type but not mutant cell lines. These data give a comprehensive, genome-wide picture of RE chromatin and transcription-related changes in ovarian carcinoma after epigenetic treatment and implicate p53 in RE transcriptional regulation. SIGNIFICANCE: This study identifies the repetitive element targets of epigenetic therapies in ovarian carcinoma and indicates a role for p53 in this process.
Collapse
Affiliation(s)
- James I McDonald
- The George Washington University Cancer Center (GWCC), Washington, D.C
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC
| | - Noor Diab
- The George Washington University Cancer Center (GWCC), Washington, D.C
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC
| | - Elisa Arthofer
- The George Washington University Cancer Center (GWCC), Washington, D.C
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC
| | - Melissa Hadley
- The George Washington University Cancer Center (GWCC), Washington, D.C
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC
| | - Tomas Kanholm
- The George Washington University Cancer Center (GWCC), Washington, D.C
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC
- The Institute for Biomedical Sciences at the George Washington University, Washington, DC
| | - Uzma Rentia
- The George Washington University Cancer Center (GWCC), Washington, D.C
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC
| | - Stephanie Gomez
- The George Washington University Cancer Center (GWCC), Washington, D.C
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC
- The Institute for Biomedical Sciences at the George Washington University, Washington, DC
| | - Angela Yu
- The George Washington University Cancer Center (GWCC), Washington, D.C
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC
| | - Erin E Grundy
- The George Washington University Cancer Center (GWCC), Washington, D.C
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC
- The Institute for Biomedical Sciences at the George Washington University, Washington, DC
| | - Olivia Cox
- The George Washington University Cancer Center (GWCC), Washington, D.C
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC
| | - Michael J Topper
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Xiaoyun Xing
- The Edison Family Center for Genome Sciences and Systems Biology, Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Pamela L Strissel
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Reiner Strick
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ting Wang
- The Edison Family Center for Genome Sciences and Systems Biology, Department of Genetics, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Stephen B Baylin
- Department of Oncology, The Johns Hopkins School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Katherine B Chiappinelli
- The George Washington University Cancer Center (GWCC), Washington, D.C.
- Department of Microbiology, Immunology & Tropical Medicine, The George Washington University, Washington, DC
| |
Collapse
|
9
|
Yang Y, Kong R, Goh FG, Somers WG, Hime GR, Li Z, Cai Y. dRTEL1 is essential for the maintenance of Drosophila male germline stem cells. PLoS Genet 2021; 17:e1009834. [PMID: 34644293 PMCID: PMC8513875 DOI: 10.1371/journal.pgen.1009834] [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: 04/09/2021] [Accepted: 09/23/2021] [Indexed: 11/19/2022] Open
Abstract
Stem cells have the potential to maintain undifferentiated state and differentiate into specialized cell types. Despite numerous progress has been achieved in understanding stem cell self-renewal and differentiation, many fundamental questions remain unanswered. In this study, we identify dRTEL1, the Drosophila homolog of Regulator of Telomere Elongation Helicase 1, as a novel regulator of male germline stem cells (GSCs). Our genome-wide transcriptome analysis and ChIP-Seq results suggest that dRTEL1 affects a set of candidate genes required for GSC maintenance, likely independent of its role in DNA repair. Furthermore, dRTEL1 prevents DNA damage-induced checkpoint activation in GSCs. Finally, dRTEL1 functions to sustain Stat92E protein levels, the key player in GSC maintenance. Together, our findings reveal an intrinsic role of the DNA helicase dRTEL1 in maintaining male GSC and provide insight into the function of dRTEL1.
Collapse
Affiliation(s)
- Ying Yang
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | - Ruiyan Kong
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Feng Guang Goh
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore
| | - W. Gregory Somers
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Australia
| | - Gary R. Hime
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Australia
| | - Zhouhua Li
- College of Life Sciences, Capital Normal University, Beijing, China
| | - Yu Cai
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
10
|
Kar FM, Hochwagen A. Phospho-Regulation of Meiotic Prophase. Front Cell Dev Biol 2021; 9:667073. [PMID: 33928091 PMCID: PMC8076904 DOI: 10.3389/fcell.2021.667073] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Germ cells undergoing meiosis rely on an intricate network of surveillance mechanisms that govern the production of euploid gametes for successful sexual reproduction. These surveillance mechanisms are particularly crucial during meiotic prophase, when cells execute a highly orchestrated program of chromosome morphogenesis and recombination, which must be integrated with the meiotic cell division machinery to ensure the safe execution of meiosis. Dynamic protein phosphorylation, controlled by kinases and phosphatases, has emerged as one of the main signaling routes for providing readout and regulation of chromosomal and cellular behavior throughout meiotic prophase. In this review, we discuss common principles and provide detailed examples of how these phosphorylation events are employed to ensure faithful passage of chromosomes from one generation to the next.
Collapse
Affiliation(s)
- Funda M Kar
- Department of Biology, New York University, New York, NY, United States
| | - Andreas Hochwagen
- Department of Biology, New York University, New York, NY, United States
| |
Collapse
|
11
|
Gebel J, Tuppi M, Sänger N, Schumacher B, Dötsch V. DNA Damaged Induced Cell Death in Oocytes. Molecules 2020; 25:molecules25235714. [PMID: 33287328 PMCID: PMC7730327 DOI: 10.3390/molecules25235714] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022] Open
Abstract
The production of haploid gametes through meiosis is central to the principle of sexual reproduction. The genetic diversity is further enhanced by exchange of genetic material between homologous chromosomes by the crossover mechanism. This mechanism not only requires correct pairing of homologous chromosomes but also efficient repair of the induced DNA double-strand breaks. Oocytes have evolved a unique quality control system that eliminates cells if chromosomes do not correctly align or if DNA repair is not possible. Central to this monitoring system that is conserved from nematodes and fruit fly to humans is the p53 protein family, and in vertebrates in particular p63. In mammals, oocytes are stored for a long time in the prophase of meiosis I which, in humans, can last more than 50 years. During the entire time of this arrest phase, the DNA damage checkpoint remains active. The treatment of female cancer patients with DNA damaging irradiation or chemotherapeutics activates this checkpoint and results in elimination of the oocyte pool causing premature menopause and infertility. Here, we review the molecular mechanisms of this quality control system and discuss potential therapeutic intervention for the preservation of the oocyte pool during chemotherapy.
Collapse
Affiliation(s)
- Jakob Gebel
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany; (J.G.); (M.T.)
| | - Marcel Tuppi
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany; (J.G.); (M.T.)
| | - Nicole Sänger
- Department for Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Venusberg-Campus 1, 53217 Bonn, Germany;
| | - Björn Schumacher
- Institute for Genome Stability in Aging and Disease, Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD) Research Center, and Center for Molecular Medicine, University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany;
| | - Volker Dötsch
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany; (J.G.); (M.T.)
- Correspondence: ; Tel.: +49-69-798-29631
| |
Collapse
|
12
|
Fat Body p53 Regulates Systemic Insulin Signaling and Autophagy under Nutrient Stress via Drosophila Upd2 Repression. Cell Rep 2020; 33:108321. [DOI: 10.1016/j.celrep.2020.108321] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/05/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
|
13
|
Hemmer LW, Dias GB, Smith B, Van Vaerenberghe K, Howard A, Bergman CM, Blumenstiel JP. Hybrid dysgenesis in Drosophila virilis results in clusters of mitotic recombination and loss-of-heterozygosity but leaves meiotic recombination unaltered. Mob DNA 2020; 11:10. [PMID: 32082426 PMCID: PMC7023781 DOI: 10.1186/s13100-020-0205-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/28/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Transposable elements (TEs) are endogenous mutagens and their harmful effects are especially evident in syndromes of hybrid dysgenesis. In Drosophila virilis, hybrid dysgenesis is a syndrome of incomplete gonadal atrophy that occurs when males with multiple active TE families fertilize females that lack active copies of the same families. This has been demonstrated to cause the transposition of paternally inherited TE families, with gonadal atrophy driven by the death of germline stem cells. Because there are abundant, active TEs in the male inducer genome, that are not present in the female reactive genome, the D. virilis syndrome serves as an excellent model for understanding the effects of hybridization between individuals with asymmetric TE profiles. RESULTS Using the D. virilis syndrome of hybrid dysgenesis as a model, we sought to determine how the landscape of germline recombination is affected by parental TE asymmetry. Using a genotyping-by-sequencing approach, we generated a high-resolution genetic map of D. virilis and show that recombination rate and TE density are negatively correlated in this species. We then contrast recombination events in the germline of dysgenic versus non-dysgenic F1 females to show that the landscape of meiotic recombination is hardly perturbed during hybrid dysgenesis. In contrast, hybrid dysgenesis in the female germline increases transmission of chromosomes with mitotic recombination. Using a de novo PacBio assembly of the D. virilis inducer genome we show that clusters of mitotic recombination events in dysgenic females are associated with genomic regions with transposons implicated in hybrid dysgenesis. CONCLUSIONS Overall, we conclude that increased mitotic recombination is likely the result of early TE activation in dysgenic progeny, but a stable landscape of meiotic recombination indicates that either transposition is ameliorated in the adult female germline or that regulation of meiotic recombination is robust to ongoing transposition. These results indicate that the effects of parental TE asymmetry on recombination are likely sensitive to the timing of transposition.
Collapse
Affiliation(s)
- Lucas W. Hemmer
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS 66045 USA
- Present Address: Department of Biology, University of Rochester, Rochester, NY 14627 USA
| | - Guilherme B. Dias
- Department of Genetics and Institute of Bioinformatics, University of Georgia, Athens, GA 30602 USA
| | - Brittny Smith
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS 66045 USA
| | - Kelley Van Vaerenberghe
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS 66045 USA
| | - Ashley Howard
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS 66045 USA
| | - Casey M. Bergman
- Department of Genetics and Institute of Bioinformatics, University of Georgia, Athens, GA 30602 USA
| | - Justin P. Blumenstiel
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS 66045 USA
| |
Collapse
|
14
|
Xu C, Shah MA, Mipam T, Wu S, Yi C, Luo H, Yuan M, Chai Z, Zhao W, Cai X. Bovid microRNAs involved in the process of spermatogonia differentiation into spermatocytes. Int J Biol Sci 2020; 16:239-250. [PMID: 31929752 PMCID: PMC6949159 DOI: 10.7150/ijbs.38232] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 09/28/2019] [Indexed: 12/17/2022] Open
Abstract
The male infertility of cattleyak resulted from spermatogenic arrest has greatly restricted the effective utilization of the heterosis from crossbreeding of cattle and yak. Based on our previous studies, the significant divergences of the transcriptomic and proteomic sequencing between yak and cattleyak prompt us to investigate the critical roles of microRNAs in post-transcriptional regulation of gene expression during spermatogenesis. TUNEL-POD analysis presented sharply decreased spermatogenic cell types and the increased apoptotic spermatogonia in cattleyak. The STA-PUT velocity sedimentation was employed to obtain spermatogonia and spermatocytes from cattle, yak and cattleyak and these spermatogenic cells were verified by the morphological and phenotypic identification. MicroRNA microarray showed that 27 differentially expressed miRNAs were simultaneously identified both in cattleyak vs cattle and in cattleyak vs yak comparisons. Further analysis revealed that the down-regulation of bta-let-7 families, bta-miR-125 and bta-miR-23a might impair the RA-induced differentiation of spermatogonia. Target gene analysis for differentially expressed miRNAs revealed that miRNAs targeted major players involved in vesicle-mediated transport, regulation of protein kinase activity and Pathways in cancer. In addition, spermatogonia transfection analysis revealed that the down-regulation of bta-miR-449a in the cattleyak might block the transition of male germ cells from the mitotic cycle to the meiotic program. The present study provided valuable information for future elucidating the regulatory roles of miRNAs involved in spermatogenic arrest of cattleyak.
Collapse
Affiliation(s)
- Chuanfei Xu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610041, Sichuan, China.,School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Mujahid Ali Shah
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - TserangDonko Mipam
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610041, Sichuan, China
| | - Shixin Wu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Chuanping Yi
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Hui Luo
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Meng Yuan
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Zhixin Chai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610041, Sichuan, China
| | - Wangsheng Zhao
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| | - Xin Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610041, Sichuan, China.,School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, Sichuan, China
| |
Collapse
|
15
|
Bhargava V, Goldstein CD, Russell L, Xu L, Ahmed M, Li W, Casey A, Servage K, Kollipara R, Picciarelli Z, Kittler R, Yatsenko A, Carmell M, Orth K, Amatruda JF, Yanowitz JL, Buszczak M. GCNA Preserves Genome Integrity and Fertility Across Species. Dev Cell 2019; 52:38-52.e10. [PMID: 31839537 DOI: 10.1016/j.devcel.2019.11.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 08/07/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022]
Abstract
The propagation of species depends on the ability of germ cells to protect their genome from numerous exogenous and endogenous threats. While these cells employ ubiquitous repair pathways, specialized mechanisms that ensure high-fidelity replication, chromosome segregation, and repair of germ cell genomes remain incompletely understood. We identified Germ Cell Nuclear Acidic Peptidase (GCNA) as a conserved regulator of genome stability in flies, worms, zebrafish, and human germ cell tumors. GCNA contains an acidic intrinsically disordered region (IDR) and a protease-like SprT domain. In addition to chromosomal instability and replication stress, Gcna mutants accumulate DNA-protein crosslinks (DPCs). GCNA acts in parallel with the SprT domain protein Spartan. Structural analysis reveals that while the SprT domain is needed to limit DNA damage, the IDR imparts significant function. This work shows that GCNA protects germ cells from various sources of damage, providing insights into conserved mechanisms that promote genome integrity across generations.
Collapse
Affiliation(s)
- Varsha Bhargava
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Courtney D Goldstein
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Logan Russell
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, PA 15213, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Murtaza Ahmed
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wei Li
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, PA 15213, USA; Tsinghua University MD Program, School of Medicine, Tsinghua University, Haidian District, Beijing 100084, PR China
| | - Amanda Casey
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kelly Servage
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, 6000 Harry Hines Boulevard NA5.120F, Dallas, TX 75235, USA
| | - Rahul Kollipara
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zachary Picciarelli
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, PA 15213, USA
| | - Ralf Kittler
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alexander Yatsenko
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, PA 15213, USA
| | - Michelle Carmell
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Biological Sciences, Wellesley College, Wellesley, MA 02481, USA
| | - Kim Orth
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, 6000 Harry Hines Boulevard NA5.120F, Dallas, TX 75235, USA
| | - James F Amatruda
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Judith L Yanowitz
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, PA 15213, USA.
| | - Michael Buszczak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
16
|
Wei Y, Bettedi L, Ting CY, Kim K, Zhang Y, Cai J, Lilly MA. The GATOR complex regulates an essential response to meiotic double-stranded breaks in Drosophila. eLife 2019; 8:e42149. [PMID: 31650955 PMCID: PMC6834368 DOI: 10.7554/elife.42149] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/13/2019] [Indexed: 01/18/2023] Open
Abstract
The TORC1 regulator GATOR1/SEACIT controls meiotic entry and early meiotic events in yeast. However, how metabolic pathways influence meiotic progression in metazoans remains poorly understood. Here we examine the role of the TORC1 regulators GATOR1 and GATOR2 in the response to meiotic double-stranded breaks (DSB) during Drosophila oogenesis. We find that in mutants of the GATOR2 component mio, meiotic DSBs trigger the constitutive downregulation of TORC1 activity and a permanent arrest in oocyte growth. Conversely, in GATOR1 mutants, high TORC1 activity results in the delayed repair of meiotic DSBs and the hyperactivation of p53. Unexpectedly, we found that GATOR1 inhibits retrotransposon expression in the presence of meiotic DSBs in a pathway that functions in parallel to p53. Thus, our studies have revealed a link between oocyte metabolism, the repair of meiotic DSBs and retrotransposon expression.
Collapse
Affiliation(s)
- Youheng Wei
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
- College of Bioscience and BiotechnologyYangzhou UniversityYangzhouChina
| | - Lucia Bettedi
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Chun-Yuan Ting
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Kuikwon Kim
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Yingbiao Zhang
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Jiadong Cai
- College of Bioscience and BiotechnologyYangzhou UniversityYangzhouChina
| | - Mary A Lilly
- Cell Biology and Neurobiology BranchNational Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
17
|
Kurtz P, Jones AE, Tiwari B, Link N, Wylie A, Tracy C, Krämer H, Abrams JM. Drosophila p53 directs nonapoptotic programs in postmitotic tissue. Mol Biol Cell 2019; 30:1339-1351. [PMID: 30892991 PMCID: PMC6724604 DOI: 10.1091/mbc.e18-12-0791] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 12/26/2022] Open
Abstract
TP53 is the most frequently mutated gene in human cancers, and despite intensive research efforts, genome-scale studies of p53 function in whole animal models are rare. The need for such in vivo studies is underscored by recent challenges to established paradigms, indicating that unappreciated p53 functions contribute to cancer prevention. Here we leveraged the Drosophila system to interrogate p53 function in a postmitotic context. In the developing embryo, p53 robustly activates important apoptotic genes in response to radiation-induced DNA damage. We recently showed that a p53 enhancer (p53RErpr) near the cell death gene reaper forms chromatin contacts and enables p53 target activation across long genomic distances. Interestingly, we found that this canonical p53 apoptotic program fails to activate in adult heads. Moreover, this failure to exhibit apoptotic responses was not associated with altered chromatin contacts. Instead, we determined that p53 does not occupy the p53RErpr enhancer in this postmitotic tissue as it does in embryos. Through comparative RNA-seq and chromatin immunoprecipitation-seq studies of developing and postmitotic tissues, we further determined that p53 regulates distinct transcriptional programs in adult heads, including DNA repair, metabolism, and proteolysis genes. Strikingly, in the postmitotic context, p53-binding landscapes were poorly correlated with nearby transcriptional effects, raising the possibility that p53 enhancers could be generally acting through long distances.
Collapse
Affiliation(s)
- Paula Kurtz
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Amanda E. Jones
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Bhavana Tiwari
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Nichole Link
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute, Houston, TX 77030
| | - Annika Wylie
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Charles Tracy
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Helmut Krämer
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - John M. Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
18
|
Genotypic and Phenotypic Variables Affect Meiotic Cell Cycle Progression, Tumor Ploidy, and Cancer-Associated Mortality in a brca2-Mutant Zebrafish Model. JOURNAL OF ONCOLOGY 2019; 2019:9218251. [PMID: 30930946 PMCID: PMC6413366 DOI: 10.1155/2019/9218251] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/10/2019] [Accepted: 01/28/2019] [Indexed: 11/17/2022]
Abstract
Successful cell replication requires both cell cycle completion and accurate chromosomal segregation. The tumor suppressor BRCA2 is positioned to influence both of these outcomes, and thereby influence genomic integrity, during meiotic and mitotic cell cycles. Accordingly, mutations in BRCA2 induce chromosomal abnormalities and disrupt cell cycle progression in both germ cells and somatic cells. Despite these findings, aneuploidy is not more prevalent in BRCA2-associated versus non-BRCA2-associated human cancers. More puzzlingly, diploidy in BRCA2-associated cancers is a negative prognostic factor, unlike non-BRCA2-associated cancers and many other human cancers. We used a brca2-mutant/tp53-mutant cancer-prone zebrafish model to explore the impact of BRCA2 mutation on cell cycle progression, ploidy, and cancer-associated mortality by performing DNA content/cell cycle analysis on zebrafish germ cells, somatic cells, and cancer cells. First, we determined that combined brca2/tp53 mutations uniquely disrupt meiotic progression. Second, we determined that sex significantly influences ploidy outcome in zebrafish cancers. Third, we determined that brca2 mutation and female sex each significantly reduce survival time in cancer-bearing zebrafish. Finally, we provide evidence to support a link between BRCA2 mutation, tumor diploidy, and poor survival outcome. These outcomes underscore the utility of this model for studying BRCA2-associated genomic aberrations in normal and cancer cells.
Collapse
|
19
|
Robin M, Issa AR, Santos CC, Napoletano F, Petitgas C, Chatelain G, Ruby M, Walter L, Birman S, Domingos PM, Calvi BR, Mollereau B. Drosophila p53 integrates the antagonism between autophagy and apoptosis in response to stress. Autophagy 2018; 15:771-784. [PMID: 30563404 DOI: 10.1080/15548627.2018.1558001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The tumor suppressor TP53/p53 is a known regulator of apoptosis and macroautophagy/autophagy. However, the molecular mechanism by which TP53 regulates 2 apparently incompatible processes remains unknown. We found that Drosophila lacking p53 displayed impaired autophagic flux, higher caspase activation and mortality in response to oxidative stress compared with wild-type flies. Moreover, autophagy and apoptosis were differentially regulated by the p53 (p53B) and ΔNp53 (p53A) isoforms: while the former induced autophagy in differentiated neurons, which protected against cell death, the latter inhibited autophagy by activating the caspases Dronc, Drice, and Dcp-1. Our results demonstrate that the differential use of p53 isoforms combined with the antagonism between apoptosis and autophagy ensures the generation of an appropriate p53 biological response to stress.
Collapse
Affiliation(s)
- Marion Robin
- a Université de Lyon, ENSL, UCBL, CNRS, LBMC, UMS 3444 Biosciences Lyon Gerland , Lyon , France
| | - Abdul Raouf Issa
- b Genes Circuits Rhythms and Neuropathology , Brain Plasticity Unit, CNRS, ESPCI Paris, Labex Memolife, PSL Research University , Paris , France.,e Department of Life Sciences , University of Trieste c/o CIB National Laboratory , Area Science Park , Trieste , Italy
| | - Cristiana C Santos
- c Instituto de Tecnologia Química e Biológica , Universidade Nova de Lisboa , Oeiras , Portugal
| | - Francesco Napoletano
- a Université de Lyon, ENSL, UCBL, CNRS, LBMC, UMS 3444 Biosciences Lyon Gerland , Lyon , France.,e Department of Life Sciences , University of Trieste c/o CIB National Laboratory , Area Science Park , Trieste , Italy
| | - Céline Petitgas
- b Genes Circuits Rhythms and Neuropathology , Brain Plasticity Unit, CNRS, ESPCI Paris, Labex Memolife, PSL Research University , Paris , France
| | - Gilles Chatelain
- a Université de Lyon, ENSL, UCBL, CNRS, LBMC, UMS 3444 Biosciences Lyon Gerland , Lyon , France
| | - Mathilde Ruby
- a Université de Lyon, ENSL, UCBL, CNRS, LBMC, UMS 3444 Biosciences Lyon Gerland , Lyon , France
| | - Ludivine Walter
- a Université de Lyon, ENSL, UCBL, CNRS, LBMC, UMS 3444 Biosciences Lyon Gerland , Lyon , France
| | - Serge Birman
- b Genes Circuits Rhythms and Neuropathology , Brain Plasticity Unit, CNRS, ESPCI Paris, Labex Memolife, PSL Research University , Paris , France
| | - Pedro M Domingos
- c Instituto de Tecnologia Química e Biológica , Universidade Nova de Lisboa , Oeiras , Portugal
| | - Brian R Calvi
- d Department of Biology , Indiana University , Bloomington , IN , USA
| | - Bertrand Mollereau
- a Université de Lyon, ENSL, UCBL, CNRS, LBMC, UMS 3444 Biosciences Lyon Gerland , Lyon , France
| |
Collapse
|
20
|
McCarthy A, Deiulio A, Martin ET, Upadhyay M, Rangan P. Tip60 complex promotes expression of a differentiation factor to regulate germline differentiation in female Drosophila. Mol Biol Cell 2018; 29:2933-2945. [PMID: 30230973 PMCID: PMC6329907 DOI: 10.1091/mbc.e18-06-0385] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/06/2018] [Accepted: 09/13/2018] [Indexed: 01/23/2023] Open
Abstract
Germline stem cells (GSCs) self-renew and differentiate to sustain a continuous production of gametes. In the female Drosophila germ line, two differentiation factors, bag of marbles ( bam) and benign gonial cell neoplasm ( bgcn), work in concert in the stem cell daughter to promote the generation of eggs. In GSCs, bam transcription is repressed by signaling from the niche and is activated in stem cell daughters. In contrast, bgcn is transcribed in both the GSCs and stem cell daughters, but little is known about how bgcn is transcriptionally modulated. Here we find that the conserved protein Nipped-A acts through the Tat interactive protein 60-kDa (Tip60) histone acetyl transferase complex in the germ line to promote GSC daughter differentiation. We find that Nipped-A is required for efficient exit from the gap phase 2 (G2) of cell cycle of the GSC daughter and for expression of a differentiation factor, bgcn. Loss of Nipped-A results in accumulation of GSC daughters . Forced expression of bgcn in Nipped-A germline-depleted ovaries rescues this differentiation defect. Together, our results indicate that Tip60 complex coordinates cell cycle progression and expression of bgcn to help drive GSC daughters toward a differentiation program.
Collapse
Affiliation(s)
- Alicia McCarthy
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY 12222
| | - Aron Deiulio
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY 12222
| | - Elliot Todd Martin
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY 12222
| | - Maitreyi Upadhyay
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY 12222
| | - Prashanth Rangan
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY 12222
| |
Collapse
|
21
|
Yasin M, Mazdak R, Mino I. Aflatoxin B1 impairs spermatogenesis: An experimental study for crosslink between oxidative stress and mitochondria-dependent apoptosis. ENVIRONMENTAL TOXICOLOGY 2018; 33:1204-1213. [PMID: 30126036 DOI: 10.1002/tox.22627] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 07/02/2018] [Accepted: 07/16/2018] [Indexed: 06/08/2023]
Abstract
The present experimental study was carried out to investigate the crosslink between aflatoxin B1 (AFB1)-induced oxidative stress and mitochondria-dependent apoptosis in testicles. For this purpose, 24 mature male Swiss albino mice were randomly divided into control and test groups. The AFB1 was dissolved in corn oil and ethanol (95:5, v/v) vehicle. The animals in test group subdivided into three groups, which received the AFB1 at a daily dose of 20 μg/kg body weight, through intraperitoneal (i.p.) route, for 7, 14, and 21 days. The mice in the control group received the vehicle alone for 21 days. The expression of Bcl-2, Bax, p53, and caspase-3 at both mRNA and protein levels were analyzed by using reverse transcription PCR (RT-PCR) and immunohistochemistry, respectively. Moreover, the mitochondrial content of germinal epithelium, tubular differentiation (TDI), and spermiogenesis (SPI) indices was analyzed. Finally, the apoptosis was assessed by using TUNEL staining. Observations revealed that the AFB1 remarkably (P < .05) reduced Bcl-2 expression at both mRNA and protein levels. Up-regulated Bax, caspase-3, and p53 expression were revealed in AFB1-received animals, which developed time-dependently. Histological examinations exhibited a significant reduction in TDI and SPI indices. Finally, the AFB1-induced apoptosis index increased time-dependently. In conclusion, the AFB1 adversely affects the spermatogenesis via inducing oxidative stress, diminishing cellular mitochondrial content and enhancing pro-apoptotic Bax, caspase-3, and p53 expression. All these impairments result in mitochondria-dependent apoptosis.
Collapse
Affiliation(s)
- Malekzadeh Yasin
- Department of Biology, Faculty of Basic Science, Urmia University, Urmia, Iran
| | - Razi Mazdak
- Department of Basic Science, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Ilkhanipour Mino
- Department of Biology, Faculty of Basic Science, Urmia University, Urmia, Iran
| |
Collapse
|
22
|
Liu C, Cai Z, Jin G, Peng D, Pan BS, Zhang X, Han F, Xu X, Lin HK. Abnormal gametogenesis induced by p53 deficiency promotes tumor progression and drug resistance. Cell Discov 2018; 4:54. [PMID: 30302273 PMCID: PMC6167385 DOI: 10.1038/s41421-018-0054-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/08/2018] [Accepted: 07/12/2018] [Indexed: 01/15/2023] Open
Abstract
The century-old embryonal/gametogenesis hypothesis of tumors could link diverse tumors' malignant features together likely representing the real "stemness" of tumors. However, the genetic evidence to validate abnormal gametogenesis in tumors remains lacking. Here we show that p53 deficiency elicits abnormal gametogenesis from primordial germ cell-like stage to late oocyte-like stage and subsequent parthenogenetic activation. The similar upregulation of abnormal gametogenesis by p53 deficiency is observed both in p53-/- mouse model and cultured cancer cells. Notably, germ cell-like cells isolated from distinct tumors from p53-/- mice and cancer cell lines display potent tumorigenicity potential. Abnormal oogenesis induced by p53 deficiency and then spontaneous parthenogenetic activation endow tumors with imitated embryonic development, life cycle, and therapeutic resistance. Our study establishes the genetic evidence to support embryonal/gametogenesis theory of tumors and reveals a pivotal role of p53 in restricting abnormal gametogenesis that may represent a novel aspect for p53's tumor suppression.
Collapse
Affiliation(s)
- Chunfang Liu
- 1Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040 China
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Zhen Cai
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Guoxiang Jin
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Danni Peng
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Bo-Syong Pan
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Xian Zhang
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Fei Han
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Xiaohong Xu
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Hui-Kuan Lin
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
- 4Graduate Institute of Basic Medical Science, China Medical University, Taichung, 404 Taiwan
- 5Department of Biotechnology, Asia University, Taichung, 41354 Taiwan
| |
Collapse
|
23
|
Female Meiosis: Synapsis, Recombination, and Segregation in Drosophila melanogaster. Genetics 2018; 208:875-908. [PMID: 29487146 PMCID: PMC5844340 DOI: 10.1534/genetics.117.300081] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/18/2017] [Indexed: 12/11/2022] Open
Abstract
A century of genetic studies of the meiotic process in Drosophila melanogaster females has been greatly augmented by both modern molecular biology and major advances in cytology. These approaches, and the findings they have allowed, are the subject of this review. Specifically, these efforts have revealed that meiotic pairing in Drosophila females is not an extension of somatic pairing, but rather occurs by a poorly understood process during premeiotic mitoses. This process of meiotic pairing requires the function of several components of the synaptonemal complex (SC). When fully assembled, the SC also plays a critical role in maintaining homolog synapsis and in facilitating the maturation of double-strand breaks (DSBs) into mature crossover (CO) events. Considerable progress has been made in elucidating not only the structure, function, and assembly of the SC, but also the proteins that facilitate the formation and repair of DSBs into both COs and noncrossovers (NCOs). The events that control the decision to mature a DSB as either a CO or an NCO, as well as determining which of the two CO pathways (class I or class II) might be employed, are also being characterized by genetic and genomic approaches. These advances allow a reconsideration of meiotic phenomena such as interference and the centromere effect, which were previously described only by genetic studies. In delineating the mechanisms by which the oocyte controls the number and position of COs, it becomes possible to understand the role of CO position in ensuring the proper orientation of homologs on the first meiotic spindle. Studies of bivalent orientation have occurred in the context of numerous investigations into the assembly, structure, and function of the first meiotic spindle. Additionally, studies have examined the mechanisms ensuring the segregation of chromosomes that have failed to undergo crossing over.
Collapse
|
24
|
Bayer FE, Zimmermann M, Preiss A, Nagel AC. Overexpression of the Drosophila ATR homologous checkpoint kinase Mei-41 induces a G2/M checkpoint in Drosophila imaginal tissue. Hereditas 2018; 155:27. [PMID: 30202398 PMCID: PMC6125995 DOI: 10.1186/s41065-018-0066-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 08/28/2018] [Indexed: 11/25/2022] Open
Abstract
Background DNA damage generally results in the activation of ATM/ATR kinases and the downstream checkpoint kinases Chk1/Chk2. In Drosophila melanogaster, the ATR homologue meiotic 41 (mei-41) is pivotal to DNA damage repair and cell cycle checkpoint signalling. Although various mei-41 mutant alleles have been analyzed in the past, no gain-of-function allele is yet available. To fill this gap, we have generated transgenic flies allowing temporal and tissue-specific induction of mei-41. Results Overexpression of mei-41 in wing and eye anlagen affects proliferation and a G2/M checkpoint even in the absence of genomic stress. Similar consequences were observed following the overexpression of the downstream kinase Grapes (Grp) but not of Loki (Lok), encoding the respective Drosophila Chk1 and Chk2 homologues, in agreement with their previously reported activities. Moreover, we show that irradiation induced cell cycle arrest was prolonged in the presence of ectopic mei-41 expression. Similar to irradiation stress, mei-41 triggered the occurrence of a slower migrating form of Grp, implying specific phosphorylation of Grp in response to either signal. Using a p53R-GFP biosensor, we further show that overexpression of mei-41 was sufficient to elicit a robust p53 activation in vivo. Conclusion We conclude that overexpression of the Drosophila ATR homologue mei-41 elicits an effectual DNA damage response irrespective of irradiation. Electronic supplementary material The online version of this article (10.1186/s41065-018-0066-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fabienne E Bayer
- Universität Hohenheim, Institut für Genetik, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Mirjam Zimmermann
- Universität Hohenheim, Institut für Genetik, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Anette Preiss
- Universität Hohenheim, Institut für Genetik, Garbenstr. 30, 70599 Stuttgart, Germany
| | - Anja C Nagel
- Universität Hohenheim, Institut für Genetik, Garbenstr. 30, 70599 Stuttgart, Germany
| |
Collapse
|
25
|
Transposons, p53 and Genome Security. Trends Genet 2018; 34:846-855. [PMID: 30195581 DOI: 10.1016/j.tig.2018.08.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/31/2018] [Accepted: 08/07/2018] [Indexed: 12/16/2022]
Abstract
p53, the most commonly mutated tumor suppressor, is a transcription factor known to regulate proliferation, senescence, and apoptosis. Compelling studies have found that p53 may prevent oncogenesis through effectors that are unrelated to these canonical processes and recent findings have uncovered ancient roles for p53 in the containment of mobile elements. Together, these developments raise the possibility that some p53-driven cancers could result from unrestrained transposons. Here, we explore evidence linking conserved features of p53 biology to the control of transposons. We also show how p53-deficient cells can be exploited to probe the behavior of transposons and illustrate how unrestrained transposons incited by p53 loss might contribute to human malignancies.
Collapse
|
26
|
Contreras EG, Sierralta J, Glavic A. p53 is required for brain growth but is dispensable for resistance to nutrient restriction during Drosophila larval development. PLoS One 2018; 13:e0194344. [PMID: 29621246 PMCID: PMC5886404 DOI: 10.1371/journal.pone.0194344] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/01/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Animal growth is influenced by the genetic background and the environmental circumstances. How genes promote growth and coordinate adaptation to nutrient availability is still an open question. p53 is a transcription factor that commands the cellular response to different types of stresses. In adult Drosophila melanogaster, p53 regulates the metabolic adaptation to nutrient restriction that supports fly viability. Furthermore, the larval brain is protected from nutrient restriction in a phenomenon called 'brain sparing'. Therefore, we hypothesised that p53 may regulate brain growth and show a protective role over brain development under nutrient restriction. RESULTS Here, we studied the function of p53 during brain growth in normal conditions and in animals subjected to developmental nutrient restriction. We showed that p53 loss of function reduced animal growth and larval brain size. Endogenous p53 was expressed in larval neural stem cells, but its levels and activity were not affected by nutritional stress. Interestingly, p53 knockdown only in neural stem cells was sufficient to decrease larval brain growth. Finally, we showed that in p53 mutant larvae under nutrient restriction, the energy storage levels were not altered, and these larvae generated adults with brains of similar size than wild-type animals. CONCLUSIONS Using genetic approaches, we demonstrate that p53 is required for proper growth of the larval brain. This developmental role of p53 does not have an impact on animal resistance to nutritional stress since brain growth in p53 mutants under nutrient restriction is similar to control animals.
Collapse
Affiliation(s)
- Esteban G. Contreras
- Biomedical Neuroscience Institute and Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Independencia Santiago-Chile
- Center for Genome Regulation, Department of Biology, Faculty of Science, Universidad of Chile, Las Palmeras Nuñoa, Santiago-Chile
| | - Jimena Sierralta
- Biomedical Neuroscience Institute and Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Independencia Santiago-Chile
- * E-mail: (AG); (JS)
| | - Alvaro Glavic
- Center for Genome Regulation, Department of Biology, Faculty of Science, Universidad of Chile, Las Palmeras Nuñoa, Santiago-Chile
- * E-mail: (AG); (JS)
| |
Collapse
|
27
|
Testa E, Nardozi D, Antinozzi C, Faieta M, Di Cecca S, Caggiano C, Fukuda T, Bonanno E, Zhenkun L, Maldonado A, Roig I, Di Giacomo M, Barchi M. H2AFX and MDC1 promote maintenance of genomic integrity in male germ cells. J Cell Sci 2018; 131:jcs.214411. [PMID: 29437857 DOI: 10.1242/jcs.214411] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 01/31/2018] [Indexed: 12/18/2022] Open
Abstract
In somatic cells, H2afx and Mdc1 are close functional partners in DNA repair and damage response. However, it is not known whether they are also involved in the maintenance of genome integrity in meiosis. By analyzing chromosome dynamics in H2afx-/- spermatocytes, we found that the synapsis of autosomes and X-Y chromosomes was impaired in a fraction of cells. Such defects correlated with an abnormal recombination profile. Conversely, Mdc1 was dispensable for the synapsis of the autosomes and played only a minor role in X-Y synapsis, compared with the action of H2afx This suggested that those genes have non-overlapping functions in chromosome synapsis. However, we observed that both genes play a similar role in the assembly of MLH3 onto chromosomes, a key step in crossover formation. Moreover, we show that H2afx and Mdc1 cooperate in promoting the activation of the recombination-dependent checkpoint, a mechanism that restrains the differentiation of cells with unrepaired DSBs. This occurs by a mechanism that involves P53. Overall, our data show that, in male germ cells, H2afx and Mdc1 promote the maintenance of genome integrity.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Erika Testa
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Daniela Nardozi
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Cristina Antinozzi
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Monica Faieta
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Stefano Di Cecca
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Cinzia Caggiano
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Tomoyuki Fukuda
- Department of Cellular Physiology, Niigata University Graduate School of Medical and Dental Sciences, 951-8510 Niigata, Japan.,Graduate School of Biological Sciences, Nara Institute of Science and Technology, 630-0192 Nara, Japan
| | - Elena Bonanno
- Department of Experimental Medicine and Surgery, Section of Pathological Anatomy, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Lou Zhenkun
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905 USA
| | - Andros Maldonado
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Ignasi Roig
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | | | - Marco Barchi
- Department of Biomedicine and Prevention, Section of Anatomy, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
28
|
Clémot M, Molla-Herman A, Mathieu J, Huynh JR, Dostatni N. The replicative histone chaperone CAF-1 is essential for the maintenance of identity and genome integrity in adult stem cells. Development 2018; 145:dev.161190. [DOI: 10.1242/dev.161190] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 07/20/2018] [Indexed: 12/14/2022]
Abstract
Chromatin packaging and modifications are important to define the identity of stem cells. How chromatin properties are retained over multiple cycles of stem cell replication, while generating differentiating progeny at the same time, remains a challenging question. The chromatin assembly factor CAF-1 is a conserved histone chaperone, which assembles histones H3 and H4 onto newly synthesized DNA during replication and repair. Here, we investigated the role of CAF-1 in the maintenance of germline stem cells (GSCs) in Drosophila ovaries. We depleted P180, the large subunit of CAF-1, in germ cells and found that it was required in GSCs to maintain their identity. In the absence of P180, GSCs still harbor stem cell properties but concomitantly express markers of differentiation. In addition, P180-depleted germ cells exhibit elevated levels of DNA damage and de-repression of the transposable I-element. These DNA damages activate p53- and Chk2-dependent checkpoints pathways, leading to cell death and female sterility. Altogether, our work demonstrates that chromatin dynamics mediated by CAF-1 play an important role in both the regulation of stem cell identity and genome integrity.
Collapse
Affiliation(s)
- Marie Clémot
- Institut Curie, PSL Research University, CNRS, Sorbonne Université, Nuclear Dynamics, Paris, France
| | - Anahi Molla-Herman
- Institut Curie, PSL Research University, CNRS, Inserm, Sorbonne Université, Genetics and Developmental Biology, Paris, France
| | - Juliette Mathieu
- Institut Curie, PSL Research University, CNRS, Inserm, Sorbonne Université, Genetics and Developmental Biology, Paris, France
| | - Jean-René Huynh
- Institut Curie, PSL Research University, CNRS, Inserm, Sorbonne Université, Genetics and Developmental Biology, Paris, France
| | - Nathalie Dostatni
- Institut Curie, PSL Research University, CNRS, Sorbonne Université, Nuclear Dynamics, Paris, France
| |
Collapse
|
29
|
p53 is required for female germline stem cell maintenance in P-element hybrid dysgenesis. Dev Biol 2017; 434:215-220. [PMID: 29294306 DOI: 10.1016/j.ydbio.2017.12.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/01/2017] [Accepted: 12/28/2017] [Indexed: 02/07/2023]
Abstract
Hybrid dysgenesis is a sterility syndrome resulting from the mobilization of certain transposable elements in the Drosophila germline. Particularly extreme is the hybrid dysgenesis syndrome caused by P-element DNA transposons, in which dysgenic female ovaries often contain few or no germline cells. Those offspring that are produced from dysgenic germlines exhibit high rates of de novo mutation and recombination, implicating transposition-associated DNA damage as the cause of germline loss. However, how this loss occurs, in terms of the particular cellular response that is triggered (cell cycle arrest, senescence, or cell death) remains poorly understood. We demonstrate that two components of the DNA damage response, Checkpoint kinase 2 and its downstream target p53, determine the frequency of ovarian atrophy that is associated with P-element hybrid dysgenesis. We further show that p53 is strongly induced in the germline stem cells (GSCs) of dysgenic females, and is required for their maintenance. Our observations support the critical role for p53 in conferring tolerance of transposable element activity in stem cells.
Collapse
|
30
|
p53 and cyclin G cooperate in mediating genome stability in somatic cells of Drosophila. Sci Rep 2017; 7:17890. [PMID: 29263364 PMCID: PMC5738409 DOI: 10.1038/s41598-017-17973-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/04/2017] [Indexed: 11/16/2022] Open
Abstract
One of the key players in genome surveillance is the tumour suppressor p53 mediating the adaptive response to a multitude of stress signals. Here we identify Cyclin G (CycG) as co-factor of p53-mediated genome stability. CycG has been shown before to be involved in double-strand break repair during meiosis. Moreover, it is also important for mediating DNA damage response in somatic tissue. Here we find it in protein complexes together with p53, and show that the two proteins interact physically in vitro and in vivo in response to ionizing irradiation. In contrast to mammals, Drosophila Cyclin G is no transcriptional target of p53. Genetic interaction data reveal that p53 activity during DNA damage response requires the presence of CycG. Morphological defects caused by overexpression of p53 are ameliorated in cycG null mutants. Moreover, using a p53 biosensor we show that p53 activity is impeded in cycG mutants. As both p53 and CycG are likewise required for DNA damage repair and longevity we propose that CycG plays a positive role in mediating p53 function in genome surveillance of Drosophila.
Collapse
|
31
|
Napoletano F, Gibert B, Yacobi-Sharon K, Vincent S, Favrot C, Mehlen P, Girard V, Teil M, Chatelain G, Walter L, Arama E, Mollereau B. p53-dependent programmed necrosis controls germ cell homeostasis during spermatogenesis. PLoS Genet 2017; 13:e1007024. [PMID: 28945745 PMCID: PMC5629030 DOI: 10.1371/journal.pgen.1007024] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 10/05/2017] [Accepted: 09/15/2017] [Indexed: 01/02/2023] Open
Abstract
The importance of regulated necrosis in pathologies such as cerebral stroke and myocardial infarction is now fully recognized. However, the physiological relevance of regulated necrosis remains unclear. Here, we report a conserved role for p53 in regulating necrosis in Drosophila and mammalian spermatogenesis. We found that Drosophila p53 is required for the programmed necrosis that occurs spontaneously in mitotic germ cells during spermatogenesis. This form of necrosis involved an atypical function of the initiator caspase Dronc/Caspase 9, independent of its catalytic activity. Prevention of p53-dependent necrosis resulted in testicular hyperplasia, which was reversed by restoring necrosis in spermatogonia. In mouse testes, p53 was required for heat-induced germ cell necrosis, indicating that regulation of necrosis is a primordial function of p53 conserved from invertebrates to vertebrates. Drosophila and mouse spermatogenesis will thus be useful models to identify inducers of necrosis to treat cancers that are refractory to apoptosis.
Collapse
Affiliation(s)
- Francesco Napoletano
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Benjamin Gibert
- Apoptosis, Cancer and Development Laboratory- Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Centre de Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Keren Yacobi-Sharon
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Stéphane Vincent
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Clémentine Favrot
- Apoptosis, Cancer and Development Laboratory- Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Centre de Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Patrick Mehlen
- Apoptosis, Cancer and Development Laboratory- Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Centre de Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Victor Girard
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Margaux Teil
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Gilles Chatelain
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Ludivine Walter
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Eli Arama
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Bertrand Mollereau
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
- * E-mail:
| |
Collapse
|
32
|
Dai MS, Hall SJ, Vantangoli Policelli MM, Boekelheide K, Spade DJ. Spontaneous testicular atrophy occurs despite normal spermatogonial proliferation in a Tp53 knockout rat. Andrology 2017; 5:1141-1152. [PMID: 28834365 DOI: 10.1111/andr.12409] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 06/12/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022]
Abstract
The tumor suppressor protein p53 (TP53) has many functions in cell cycle regulation, apoptosis, and DNA damage repair and is also involved in spermatogenesis in the mouse. To evaluate the role of p53 in spermatogenesis in the rat, we characterized testis biology in adult males of a novel p53 knockout rat (SD-Tp53tm1sage ). p53 knockout rats exhibited variable levels of testicular atrophy, including significantly decreased testis weights, atrophic seminiferous tubules, decreased seminiferous tubule diameter, and elevated spermatocyte TUNEL labeling rates, indicating a dysfunction in spermatogenesis. Phosphorylated histone H2AX protein levels and distribution were similar in the non-atrophic seminiferous tubules of both genotypes, showing evidence of pre-synaptic DNA double-strand breaks in leptotene and zygotene spermatocytes, preceding cell death in p53 knockout rat testes. Quantification of the spermatogonial stem cell (SSC) proliferation rate with bromodeoxyuridine (BrdU) labeling, in addition to staining with the undifferentiated type A spermatogonial marker GDNF family receptor alpha-1 (GFRA1), indicated that the undifferentiated spermatogonial population was normal in p53 knockout rats. Following exposure to 0.5 or 5 Gy X-ray, p53 knockout rats exhibited no germ cell apoptotic response beyond their unirradiated phenotype, while germ cell death in wild-type rat testes was elevated to a level similar to the unexposed p53 knockout rats. This study indicates that seminiferous tubule atrophy occurs following spontaneous, elevated levels of spermatocyte death in the p53 knockout rat. This phenomenon is variable across individual rats. These results indicate a critical role for p53 in rat germ cell survival and spermatogenesis.
Collapse
Affiliation(s)
- Matthew S Dai
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Susan J Hall
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | | | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Daniel J Spade
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
33
|
Marcet-Ortega M, Pacheco S, Martínez-Marchal A, Castillo H, Flores E, Jasin M, Keeney S, Roig I. p53 and TAp63 participate in the recombination-dependent pachytene arrest in mouse spermatocytes. PLoS Genet 2017; 13:e1006845. [PMID: 28617799 PMCID: PMC5491309 DOI: 10.1371/journal.pgen.1006845] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 06/29/2017] [Accepted: 06/01/2017] [Indexed: 01/09/2023] Open
Abstract
To protect germ cells from genomic instability, surveillance mechanisms ensure meiosis occurs properly. In mammals, spermatocytes that display recombination defects experience a so-called recombination-dependent arrest at the pachytene stage, which relies on the MRE11 complex—ATM—CHK2 pathway responding to unrepaired DNA double-strand breaks (DSBs). Here, we asked if p53 family members—targets of ATM and CHK2—participate in this arrest. We bred double-mutant mice combining a mutation of a member of the p53 family (p53, TAp63, or p73) with a Trip13 mutation. Trip13 deficiency triggers a recombination-dependent response that arrests spermatocytes in pachynema before they have incorporated the testis-specific histone variant H1t into their chromatin. We find that deficiency for either p53 or TAp63, but not p73, allowed spermatocytes to progress further into meiotic prophase despite the presence of numerous unrepaired DSBs. Even so, the double mutant spermatocytes apoptosed at late pachynema because of sex body deficiency; thus p53 and TAp63 are dispensable for arrest caused by sex body defects. These data affirm that recombination-dependent and sex body-deficient arrests occur via genetically separable mechanisms. Meiosis is a specialized cell division that generates haploid gametes by halving chromosome content through two consecutive rounds of chromosome segregation. At the onset of the first meiotic division, SPO11 protein introduces double-strand breaks (DSBs) throughout the genome. These DSBs are repaired through homologous recombination, which promotes pairing and synapsis of the homologous chromosomes. Some DSBs will become repaired as crossovers, providing a physical connection between the homologous chromosomes which promotes correct chromosome segregation. In fact, recombination defects can lead to formation of aneuploid gametes, one of the major causes of miscarriages and chromosome abnormalities in humans. To protect germ cells from genomic instability and to produce balanced gametes, surveillance mechanisms ensure that meiosis occurs properly. It is known that in the presence of unrepaired DSBs a control mechanism promotes a spermatogenic block at the pachytene stage. Here we describe that, downstream MRE11-ATM-CHK2 pathway, p53 and TAp63 are the effectors responsible for activating recombination-dependent arrest in mouse spermatocytes.
Collapse
Affiliation(s)
- Marina Marcet-Ortega
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Cytology and Histology Unit, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Sarai Pacheco
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Cytology and Histology Unit, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Ana Martínez-Marchal
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Cytology and Histology Unit, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Helena Castillo
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Cytology and Histology Unit, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Elsa Flores
- Department of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Maria Jasin
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Scott Keeney
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Ignasi Roig
- Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Cytology and Histology Unit, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- * E-mail:
| |
Collapse
|
34
|
Moshari S, Nejati V, Najafi G, razi M. Nanomicelle curcumin-induced DNA fragmentation in testicular tissue; Correlation between mitochondria dependent apoptosis and failed PCNA-related hemostasis. Acta Histochem 2017; 119:372-381. [PMID: 28385400 DOI: 10.1016/j.acthis.2017.03.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/25/2017] [Accepted: 03/25/2017] [Indexed: 10/19/2022]
Abstract
Current study was done to assess possible anti-proliferative effect of nanomicelle curcumin (NMCM) against germ cells in testicular tissue. For this purpose, 24 mature male Wistar rats were divided into control and test groups. The animals in test groups received 7.5mg/kg, 15mg/kg and 30mg/kg of NMC (NO=6 rats in each group). Following 48days, the expression of Bcl-2, Bax, caspase-3, P53 and proliferating cell nuclear antigen (PCNA) were evaluated by using reverse transcription-PCR and immunohistochemistry. Histological changes, tubular differentiation index (TDI), tissue cellularity and serum level of testosterone were analyzed. Finally, the DNA laddering test was used to assess the DNA fragmentation as hallmark for apoptosis. The NMCM significantly (P<0.05) diminished the Bcl-2, p53 and PCNA and enhanced the Bax and caspase-3 mRNA levels. The NMCM significantly (P<0.05) elevated the percentage of Bax and caspase-3-positive tubules and remarkably reduced the percentage of tubules with positive reaction for Bcl-2, p53 and PCNA. The NCMN-received animals exhibited remarkable (P<0.05) reduction in cell population, TDI ratio and serum level of testosterone. Severe DNA fragmentation was observed in 30mg/kg NMCM-received group. In conclusion, the NMCM by reducing the testicular endocrine status, down-regulating Bcl-2 expression and by enhancing the Bax and caspase-3 expression initiates the intrinsic apoptosis pathway. On the other hand, inhibited expression of p53 and PCNA (at dose level of 30mg/kg) suppresses the p53 and PCNA-related hemostasis/preservative reactions. All these alterations adversely affect the spermatogenesis.
Collapse
|
35
|
D’Brot A, Kurtz P, Regan E, Jakubowski B, Abrams JM. A platform for interrogating cancer-associated p53 alleles. Oncogene 2017; 36:286-291. [PMID: 26996664 PMCID: PMC5031501 DOI: 10.1038/onc.2016.48] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 01/06/2016] [Accepted: 01/19/2016] [Indexed: 12/13/2022]
Abstract
p53 is the most frequently mutated gene in human cancer. Compelling evidence argues that full transformation involves loss of growth suppression encoded by wild-type p53 together with poorly understood oncogenic activity encoded by missense mutations. Furthermore, distinguishing disease alleles from natural polymorphisms is an important clinical challenge. To interrogate the genetic activity of human p53 variants, we leveraged the Drosophila model as an in vivo platform. We engineered strains that replace the fly p53 gene with human alleles, producing a collection of stocks that are, in effect, 'humanized' for p53 variants. Like the fly counterpart, human p53 transcriptionally activated a biosensor and induced apoptosis after DNA damage. However, all humanized strains representing common alleles found in cancer patients failed to complement in these assays. Surprisingly, stimulus-dependent activation of hp53 occurred without stabilization, demonstrating that these two processes can be uncoupled. Like its fly counterpart, hp53 formed prominent nuclear foci in germline cells but cancer-associated p53 variants did not. Moreover, these same mutant alleles disrupted hp53 foci and inhibited biosensor activity, suggesting that these properties are functionally linked. Together these findings establish a functional platform for interrogating human p53 alleles and suggest that simple phenotypes could be used to stratify disease variants.
Collapse
Affiliation(s)
- Alejandro D’Brot
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Paula Kurtz
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Erin Regan
- Department of Physical Therapy, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - John M Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
36
|
Hu MW, Meng TG, Jiang ZZ, Dong MZ, Schatten H, Xu X, Wang ZB, Sun QY. Protein Phosphatase 6 Protects Prophase I-Arrested Oocytes by Safeguarding Genomic Integrity. PLoS Genet 2016; 12:e1006513. [PMID: 27930667 PMCID: PMC5179128 DOI: 10.1371/journal.pgen.1006513] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 12/22/2016] [Accepted: 11/29/2016] [Indexed: 12/21/2022] Open
Abstract
Mammalian oocytes are arrested at prophase of the first meiotic division in the primordial follicle pool for months, even years, after birth depending on species, and only a limited number of oocytes resume meiosis, complete maturation, and ovulate with each reproductive cycle. We recently reported that protein phosphatase 6 (PP6), a member of the PP2A-like subfamily, which accounts for cellular serine/threonine phosphatase activity, functions in completing the second meiosis. Here, we generated mutant mice with a specific deletion of Ppp6c in oocytes from the primordial follicle stage by crossing Ppp6cF/F mice with Gdf9-Cre mice and found that Ppp6cF/F; GCre+ mice are infertile. Depletion of PP6c caused folliculogenesis defects and germ cell loss independent of the traditional AKT/mTOR pathway, but due to persistent phosphorylation of H2AX (a marker of double strand breaks), increased susceptibility to DNA damage and defective DNA repair, which led to massive oocyte elimination and eventually premature ovarian failure (POF). Our findings uncover an important role for PP6 as an indispensable guardian of genomic integrity of the lengthy prophase I oocyte arrest, maintenance of primordial follicle pool, and thus female fertility. Formation of haploid gametes from diploid germ cells requires a specialized reductive cell division known as meiosis. In contrast to male meiosis that takes place continuously, a unique feature of female meiosis in mammals is the long arrest in meiosis I, which lasts up to 50 years in humans. Because the size of the germ cell pool determines the reproductive lifespan of females, it is important to discover mechanisms preserving the germ cell pool during the lengthy meiotic arrest. In this study, we examined the physiological role of a member of the PP2A-like serine/threonine phosphatase subfamily, protein phosphatase 6, in mouse oocytes during ovarian follicular development. This is the first study linking PP6 to the maintenance of the female germ cell pool and fertility. We find PP6 is an indispensable protector of arrested oocytes by safeguarding genomic integrity during their dormancy in the mouse ovary.
Collapse
Affiliation(s)
- Meng-Wen Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Tie-Gang Meng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zong-Zhe Jiang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ming-Zhe Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States of America
| | - Xingzhi Xu
- Beijing Key Laboratory of DNA Damage Response and College of Life Sciences, Capital Normal University, Beijing, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qing-Yuan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
37
|
Affiliation(s)
- Abigail-Rachele Mateo
- a Department of Molecular Genetics , University of Toronto , Toronto , Ontario , Canada.,b Developmental and Stem Cell Biology Program, Hospital for Sick Children , Toronto , Ontario , Canada
| | - W Brent Derry
- a Department of Molecular Genetics , University of Toronto , Toronto , Ontario , Canada.,b Developmental and Stem Cell Biology Program, Hospital for Sick Children , Toronto , Ontario , Canada
| |
Collapse
|
38
|
Abstract
Throughout the animal kingdom, p53 genes function to restrain mobile elements and recent observations indicate that transposons become derepressed in human cancers. Together, these emerging lines of evidence suggest that cancers driven by p53 mutations could represent "transpospoathies," i.e. disease states linked to eruptions of mobile elements. The transposopathy hypothesis predicts that p53 acts through conserved mechanisms to contain transposon movement, and in this way, prevents tumor formation. How transposon eruptions provoke neoplasias is not well understood but, from a broader perspective, this hypothesis also provides an attractive framework to explore unrestrained mobile elements as inciters of late-onset idiopathic disease. Also see the video abstract here.
Collapse
Affiliation(s)
- Annika Wylie
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amanda E Jones
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John M Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
39
|
Ma X, Han Y, Song X, Do T, Yang Z, Ni J, Xie T. DNA damage-induced Lok/CHK2 activation compromises germline stem cell self-renewal and lineage differentiation. Development 2016; 143:4312-4323. [PMID: 27729408 DOI: 10.1242/dev.141069] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 09/20/2016] [Indexed: 12/21/2022]
Abstract
Stem cells in adult tissues are constantly exposed to genotoxic stress and also accumulate DNA damage with age. However, it remains largely unknown how DNA damage affects both stem cell self-renewal and differentiation. In this study, we show that DNA damage retards germline stem cell (GSC) self-renewal and progeny differentiation in a Lok kinase-dependent manner in the Drosophila ovary. Both heatshock-inducible endonuclease I-CreI expression and X-ray irradiation can efficiently introduce double-strand breaks in GSCs and their progeny, resulting in a rapid GSC loss and a GSC progeny differentiation defect. Surprisingly, the elimination of Lok or its kinase activity can almost fully rescue the GSC loss and the progeny differentiation defect caused by DNA damage induced by I-CreI or X-ray. In addition, the reduction in bone morphogenetic protein signaling and Shotgun expression only makes a limited contribution to DNA damage-induced GSC loss. Finally, DNA damage also decreases the expression of the master differentiation factor Bam in a Lok-dependent manner, which helps explain the GSC progeny differentiation defect. Therefore, this study demonstrates, for the first time in vivo, that Lok kinase activation is required for the DNA damage-mediated disruption of adult stem cell self-renewal and lineage differentiation, and might also offer novel insight into how DNA damage causes tissue aging and cancer formation.
Collapse
Affiliation(s)
- Xing Ma
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA.,Department of Cell Biology and Anatomy, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Yingying Han
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Xiaoqing Song
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Trieu Do
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Zhihao Yang
- Tsinghua University School of Medicine, Beijing 100084, China
| | - Jianquan Ni
- Tsinghua University School of Medicine, Beijing 100084, China
| | - Ting Xie
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA .,Department of Cell Biology and Anatomy, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
40
|
Marjault HB, Allemand I. Consequences of irradiation on adult spermatogenesis: Between infertility and hereditary risk. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 770:340-348. [DOI: 10.1016/j.mrrev.2016.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 12/31/2022]
|
41
|
Xiong M, Ferder IC, Ohguchi Y, Wang N. Quantitative analysis of male germline stem cell differentiation reveals a role for the p53-mTORC1 pathway in spermatogonial maintenance. Cell Cycle 2016; 14:2905-13. [PMID: 26177380 DOI: 10.1080/15384101.2015.1069928] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
p53 protects cells from DNA damage by inducing cell-cycle arrest upon encountering genomic stress. Among other pathways, p53 elicits such an effect by inhibiting mammalian target of rapamycin complex 1 (mTORC1), the master regulator of cell proliferation and growth. Although recent studies have indicated roles for both p53 and mTORC1 in stem cell maintenance, it remains unclear whether the p53-mTORC1 pathway is conserved to mediate this process under normal physiological conditions. Spermatogenesis is a classic stem cell-dependent process in which undifferentiated spermatogonia undergo self-renewal and differentiation to maintain the lifelong production of spermatozoa. To better understand this process, we have developed a novel flow cytometry (FACS)-based approach that isolates spermatogonia at consecutive differentiation stages. By using this as a tool, we show that genetic loss of p53 augments mTORC1 activity during early spermatogonial differentiation. Functionally, loss of p53 drives spermatogonia out of the undifferentiated state and causes a consistent expansion of early differentiating spermatogonia until the stage of preleptotene (premeiotic) spermatocyte. The frequency of early meiotic spermatocytes is, however, dramatically decreased. Thus, these data suggest that p53-mTORC1 pathway plays a critical role in maintaining the homeostasis of early spermatogonial differentiation. Moreover, our FACS approach could be a valuable tool in understanding spermatogonial differentiation.
Collapse
Affiliation(s)
- Mulin Xiong
- a Vincent Center for Reproductive Biology; Vincent Department of Obstetrics and Gynecology; Massachusetts General Hospital; Harvard Medical School ; Boston , MA USA
| | - Ianina C Ferder
- a Vincent Center for Reproductive Biology; Vincent Department of Obstetrics and Gynecology; Massachusetts General Hospital; Harvard Medical School ; Boston , MA USA
| | - Yasuyo Ohguchi
- a Vincent Center for Reproductive Biology; Vincent Department of Obstetrics and Gynecology; Massachusetts General Hospital; Harvard Medical School ; Boston , MA USA
| | - Ning Wang
- a Vincent Center for Reproductive Biology; Vincent Department of Obstetrics and Gynecology; Massachusetts General Hospital; Harvard Medical School ; Boston , MA USA
| |
Collapse
|
42
|
Hou S, Xian L, Shi P, Li C, Lin Z, Gao X. The Magea gene cluster regulates male germ cell apoptosis without affecting the fertility in mice. Sci Rep 2016; 6:26735. [PMID: 27226137 PMCID: PMC4880894 DOI: 10.1038/srep26735] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 05/06/2016] [Indexed: 11/21/2022] Open
Abstract
While apoptosis is essential for male germ cell development, improper activation of apoptosis in the testis can affect spermatogenesis and cause reproduction defects. Members of the MAGE-A (melanoma antigen family A) gene family are frequently clustered in mammalian genomes and are exclusively expressed in the testes of normal animals but abnormally activated in a wide variety of cancers. We investigated the potential roles of these genes in spermatogenesis by generating a mouse model with a 210-kb genomic deletion encompassing six members of the Magea gene cluster (Magea1, Magea2, Magea3, Magea5, Magea6 and Magea8). Male mice carrying the deletion displayed smaller testes from 2 months old with a marked increase in apoptotic germ cells in the first wave of spermatogenesis. Furthermore, we found that Magea genes prevented stress-induced spermatogenic apoptosis after N-ethyl-N-nitrosourea (ENU) treatment during the adult stage. Mechanistically, deletion of the Magea gene cluster resulted in a dramatic increase in apoptotic germ cells, predominantly spermatocytes, with activation of p53 and induction of Bax in the testes. These observations demonstrate that the Magea genes are crucial in maintaining normal testicular size and protecting germ cells from excessive apoptosis under genotoxic stress.
Collapse
Affiliation(s)
- Siyuan Hou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Li Xian
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Peiliang Shi
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Chaojun Li
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Zhaoyu Lin
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| | - Xiang Gao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, China
| |
Collapse
|
43
|
Levine AJ, Ting DT, Greenbaum BD. P53 and the defenses against genome instability caused by transposons and repetitive elements. Bioessays 2016; 38:508-13. [PMID: 27172878 PMCID: PMC5031199 DOI: 10.1002/bies.201600031] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The recent publication by Wylie et al. is reviewed, demonstrating that the p53 protein regulates the movement of transposons. While this work presents genetic evidence for a piRNA‐mediated p53 interaction with transposons in Drosophila and zebrafish, it is herein placed in the context of a decade or so of additional work that demonstrated a role for p53 in regulating transposons and other repetitive elements. The line of thought in those studies began with the observation that transposons damage DNA and p53 regulates DNA damage. The presence of transposon movement can increase the rate of evolution in the germ line and alter genes involved in signal transduction pathways. Transposition can also play an important role in cancers where the p53 gene function is often mutated. This is particularly interesting as recent work has shown that de‐repression of repetitive elements in cancer has important consequences for the immune system and tumor microenvironment.
Collapse
Affiliation(s)
- Arnold J Levine
- Institute for Advanced Study, School of Natural Sciences, The Simons Center for Systems Biology, Princeton, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - David T Ting
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA.,Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Benjamin D Greenbaum
- Department of Medicine, Hematology, and Medical Oncology, and Pathology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
44
|
The p53-like Protein CEP-1 Is Required for Meiotic Fidelity in C. elegans. Curr Biol 2016; 26:1148-58. [PMID: 27151662 DOI: 10.1016/j.cub.2016.03.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 02/05/2016] [Accepted: 03/14/2016] [Indexed: 11/21/2022]
Abstract
The passage of genetic information during meiosis requires exceptionally high fidelity to prevent birth defects and infertility. Accurate chromosome segregation during the first meiotic division relies on the formation of crossovers between homologous chromosomes and a series of precisely controlled steps to exchange genetic information. Many studies have hinted at a role for p53 in meiosis, but how it functions in this process is poorly understood. Here, we have identified a cooperative role for the p53-like protein CEP-1 and the meiotic protein HIM-5 in maintaining genome stability in the C. elegans germline. Loss of cep-1 and him-5 results in synthetic lethality that is dependent on the upstream DNA damage checkpoint but independent of the downstream core apoptotic pathway. We show that this synthetic lethality is the result of defective crossover formation due to reduced SPO-11-dependent double-strand breaks. Using cep-1 separation-of-function alleles, we show that cep-1 and him-5 also suppress inappropriate activation of the nonhomologous end joining (NHEJ) pathway. This work reveals an ancestral function for the p53 family in ensuring the fidelity of meiosis and establishes CEP-1 as a critical determinant of repair pathway choice.
Collapse
|
45
|
Wylie A, Jones AE, D'Brot A, Lu WJ, Kurtz P, Moran JV, Rakheja D, Chen KS, Hammer RE, Comerford SA, Amatruda JF, Abrams JM. p53 genes function to restrain mobile elements. Genes Dev 2015; 30:64-77. [PMID: 26701264 PMCID: PMC4701979 DOI: 10.1101/gad.266098.115] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 11/23/2015] [Indexed: 11/24/2022]
Abstract
Wylie et al. show that p53 restricts retrotransposon activity and genetically interacts with components of the piRNA pathway. In gene complementation studies, normal human p53 alleles restrained these mobile elements, but mutant p53 alleles from cancer patients could not. Consistent with these observations, they also found patterns of unrestrained retrotransposons in p53-driven mouse and human cancers. Throughout the animal kingdom, p53 genes govern stress response networks by specifying adaptive transcriptional responses. The human member of this gene family is mutated in most cancers, but precisely how p53 functions to mediate tumor suppression is not well understood. Using Drosophila and zebrafish models, we show that p53 restricts retrotransposon activity and genetically interacts with components of the piRNA (piwi-interacting RNA) pathway. Furthermore, transposon eruptions occurring in the p53− germline were incited by meiotic recombination, and transcripts produced from these mobile elements accumulated in the germ plasm. In gene complementation studies, normal human p53 alleles suppressed transposons, but mutant p53 alleles from cancer patients could not. Consistent with these observations, we also found patterns of unrestrained retrotransposons in p53-driven mouse and human cancers. Furthermore, p53 status correlated with repressive chromatin marks in the 5′ sequence of a synthetic LINE-1 element. Together, these observations indicate that ancestral functions of p53 operate through conserved mechanisms to contain retrotransposons. Since human p53 mutants are disabled for this activity, our findings raise the possibility that p53 mitigates oncogenic disease in part by restricting transposon mobility.
Collapse
Affiliation(s)
- Annika Wylie
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Amanda E Jones
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Alejandro D'Brot
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Wan-Jin Lu
- Stanford University Medical Center, Stanford, California 94305, USA
| | - Paula Kurtz
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - John V Moran
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, Michigan 48019, USA; Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48019, USA; Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, Michigan 48019, USA
| | - Dinesh Rakheja
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Kenneth S Chen
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Robert E Hammer
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Sarah A Comerford
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - James F Amatruda
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - John M Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
46
|
Zhang B, Rotelli M, Dixon M, Calvi BR. The function of Drosophila p53 isoforms in apoptosis. Cell Death Differ 2015; 22:2058-67. [PMID: 25882045 PMCID: PMC4816103 DOI: 10.1038/cdd.2015.40] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 12/20/2022] Open
Abstract
The p53 protein is a major mediator of the cellular response to genotoxic stress and is a crucial suppressor of tumor formation. In a variety of organisms, p53 and its paralogs, p63 and p73, each encode multiple protein isoforms through alternative splicing, promoters, and translation start sites. The function of these isoforms in development and disease are still being defined. Here, we evaluate the apoptotic potential of multiple isoforms of the single p53 gene in the genetic model Drosophila melanogaster. Most previous studies have focused on the p53A isoform, but it has been recently shown that a larger p53B isoform can induce apoptosis when overexpressed. It has remained unclear, however, whether one or both isoforms are required for the apoptotic response to genotoxic stress. We show that p53B is a much more potent inducer of apoptosis than p53A when overexpressed. Overexpression of two newly identified short isoforms perturbed development and inhibited the apoptotic response to ionizing radiation. Analysis of physiological protein expression indicated that p53A is the most abundant isoform, and that both p53A and p53B can form a complex and co-localize to sub-nuclear compartments. In contrast to the overexpression results, new isoform-specific loss-of-function mutants indicated that it is the shorter p53A isoform, not full-length p53B, that is the primary mediator of pro-apoptotic gene transcription and apoptosis after ionizing radiation. Together, our data show that it is the shorter p53A isoform that mediates the apoptotic response to DNA damage, and further suggest that p53B and shorter isoforms have specialized functions.
Collapse
Affiliation(s)
- B Zhang
- Department of Biology, Indiana University, Bloomington, IN, USA
| | - M Rotelli
- Department of Biology, Indiana University, Bloomington, IN, USA
| | - M Dixon
- Department of Biology, Indiana University, Bloomington, IN, USA
| | - B R Calvi
- Department of Biology, Indiana University, Bloomington, IN, USA
| |
Collapse
|
47
|
Aneuploidy causes premature differentiation of neural and intestinal stem cells. Nat Commun 2015; 6:8894. [PMID: 26573328 PMCID: PMC4660207 DOI: 10.1038/ncomms9894] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 10/14/2015] [Indexed: 12/31/2022] Open
Abstract
Aneuploidy is associated with a variety of diseases such as cancer and microcephaly. Although many studies have addressed the consequences of a non-euploid genome in cells, little is known about their overall consequences in tissue and organism development. Here we use two different mutant conditions to address the consequences of aneuploidy during tissue development and homeostasis in Drosophila. We show that aneuploidy causes brain size reduction due to a decrease in the number of proliferative neural stem cells (NSCs), but not through apoptosis. Instead, aneuploid NSCs present an extended G1 phase, which leads to cell cycle exit and premature differentiation. Moreover, we show that this response to aneuploidy is also present in adult intestinal stem cells but not in the wing disc. Our work highlights a neural and intestine stem cell-specific response to aneuploidy, which prevents their proliferation and expansion. It is unclear why certain tissues are more susceptible to the consequences of aneuploidy. Here, in Drosophila, Gogendeau et al. identify aneuploidy as the cause of lengthened G1 and premature differentiation in both neural and adult intestinal stem cells, which prevents cells with abnormal genomes from cycling.
Collapse
|
48
|
Abstract
The canonical role of p53 in preserving genome integrity and limiting carcinogenesis has been well established. In the presence of acute DNA-damage, oncogene deregulation and other forms of cellular stress, p53 orchestrates a myriad of pleiotropic processes to repair cellular damages and maintain homeostasis. Beside these well-studied functions of p53, recent studies in Drosophila have unraveled intriguing roles of Dmp53 in promoting cell division in apoptosis-induced proliferation, enhancing fitness and proliferation of the winner cell in cell competition and coordinating growth at the organ and organismal level in the presence of stress. In this review, we describe these new functions of Dmp53 and discuss their relevance in the context of carcinogenesis.
Collapse
Affiliation(s)
- Bertrand Mollereau
- Laboratory of Molecular Biology of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, UMS 3444 Biosciences Lyon Gerland, University of Lyon, Lyon, France,
| | | |
Collapse
|
49
|
Subramanian VV, Hochwagen A. The meiotic checkpoint network: step-by-step through meiotic prophase. Cold Spring Harb Perspect Biol 2014; 6:a016675. [PMID: 25274702 DOI: 10.1101/cshperspect.a016675] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The generation of haploid gametes by meiosis is a highly conserved process for sexually reproducing organisms that, in almost all cases, involves the extensive breakage of chromosomes. These chromosome breaks occur during meiotic prophase and are essential for meiotic recombination as well as the subsequent segregation of homologous chromosomes. However, their formation and repair must be carefully monitored and choreographed with nuclear dynamics and the cell division program to avoid the creation of aberrant chromosomes and defective gametes. It is becoming increasingly clear that an intricate checkpoint-signaling network related to the canonical DNA damage response is deeply interwoven with the meiotic program and preserves order during meiotic prophase. This meiotic checkpoint network (MCN) creates a wide range of dependent relationships controlling chromosome movement, chromosome pairing, chromatin structure, and double-strand break (DSB) repair. In this review, we summarize our current understanding of the MCN. We discuss commonalities and differences in different experimental systems, with a particular emphasis on the emerging design principles that control and limit cross talk between signals to ultimately ensure the faithful inheritance of chromosomes by the next generation.
Collapse
Affiliation(s)
| | - Andreas Hochwagen
- Department of Biology, New York University, New York, New York 10003
| |
Collapse
|
50
|
Tollini LA, Jin A, Park J, Zhang Y. Regulation of p53 by Mdm2 E3 ligase function is dispensable in embryogenesis and development, but essential in response to DNA damage. Cancer Cell 2014; 26:235-47. [PMID: 25117711 PMCID: PMC4369778 DOI: 10.1016/j.ccr.2014.06.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 03/14/2014] [Accepted: 06/12/2014] [Indexed: 12/21/2022]
Abstract
Mdm2 E3 ubiquitin ligase-mediated p53 degradation is generally accepted as the major mechanism for p53 regulation; nevertheless, the in vivo significance of this function has not been unequivocally established. Here, we have generated an Mdm2(Y487A) knockin mouse; Mdm2(Y487A) mutation inactivates Mdm2 E3 ligase function without affecting its ability to bind its homolog MdmX. Unexpectedly, Mdm2(Y487A/Y487A) mice were viable and developed normally into adulthood. While disruption of Mdm2 E3 ligase function resulted in p53 accumulation, p53 transcriptional activity remained low; however, exposure to sublethal stress resulted in hyperactive p53 and p53-dependent mortality in Mdm2(Y487A/Y487A) mice. These findings reveal a potentially dispensable nature for Mdm2 E3 ligase function in p53 regulation, providing insight that may affect how this pathway is targeted therapeutically.
Collapse
Affiliation(s)
- Laura A Tollini
- Department of Radiation Oncology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7512, USA; Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7512, USA; Curriculum in Genetics and Molecular Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7512, USA
| | - Aiwen Jin
- Department of Radiation Oncology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7512, USA; Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7512, USA
| | - Jikyoung Park
- Department of Radiation Oncology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7512, USA; Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7512, USA
| | - Yanping Zhang
- Department of Radiation Oncology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7512, USA; Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7512, USA; Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7512, USA; Curriculum in Genetics and Molecular Biology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7512, USA; Laboratory of Biological Cancer Therapy, Cancer Institute, Xuzhou Medical College, Xuzhou 221002, China.
| |
Collapse
|