1
|
Koch LB, Marston AL. The functional organisation of the centromere and kinetochore during meiosis. Curr Opin Cell Biol 2025; 94:102486. [PMID: 40015116 PMCID: PMC7617577 DOI: 10.1016/j.ceb.2025.102486] [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: 10/03/2024] [Revised: 02/04/2025] [Accepted: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Meiosis generates gametes through a specialised cell cycle that reduces the genome by half. Homologous chromosomes are segregated in meiosis I and sister chromatids are segregated in meiosis II. Centromeres and kinetochores play central roles in instructing this specialised chromosome segregation pattern. Accordingly, kinetochores acquire meiosis-specific modifications. Here we contextualise recent highlights in our understanding of how centromeres and kinetochores direct the sorting of chromosomes into gametes via meiosis.
Collapse
Affiliation(s)
- Lori B Koch
- Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, United Kingdom
| | - Adele L Marston
- Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, United Kingdom.
| |
Collapse
|
2
|
Langeoire A, Kem-Seng A, Cladière D, Wassmann K, Buffin E. Prolonged metaphase II arrest weakens Aurora B/C-dependent error correction in mouse oocytes. Curr Biol 2025; 35:2019-2031.e4. [PMID: 40215962 DOI: 10.1016/j.cub.2025.03.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 02/10/2025] [Accepted: 03/14/2025] [Indexed: 05/08/2025]
Abstract
Chromosome segregation during meiosis is highly error-prone in mammalian oocytes. The mechanisms controlling chromosome attachments and the spindle assembly checkpoint (SAC) have been extensively studied in meiosis I, but our knowledge of these mechanisms during meiosis II is rather limited. Although mammalian oocytes arrest in metaphase II for an extended period awaiting fertilization, some misattached chromosomes may persist. This suggests that the mechanism correcting misattachments is not fully functional during the arrest. In this study, we investigated whether low inter-kinetochore tension, which characterizes incorrect attachments, can be detected by Aurora B/C-dependent error correction in meiosis II. We found that low tension, induced by low dose of STLC in early metaphase II, does indeed mediate microtubule detachment by Aurora B/C and, consequently, anaphase II delay through SAC activation. Surprisingly, we also found that, during prolonged metaphase II arrest, Aurora B/C activity is no longer sufficient to detach low-tension attachments, correlating with high accumulation of PP2A at kinetochores. As a result, the SAC is not activated, and sister chromatids segregate in anaphase II without delay even in the presence of low tension. Hence, during the prolonged metaphase II arrest to await fertilization, oocytes become unable to discriminate between correct and incorrect attachments and may allow errors to persist.
Collapse
Affiliation(s)
- Antoine Langeoire
- Université Paris Cité, CNRS, Institut Jacques Monod, 15 rue Hélène Brion, 75013 Paris, France; Sorbonne Université, Institut de Biologie Paris Seine, 9 quai St. Bernard, 75252 Paris, France; CNRS UMR7622, Developmental Biology Lab, Sorbonne Université, 9 quai St. Bernard, 75252 Paris, France
| | - Alison Kem-Seng
- Sorbonne Université, Institut de Biologie Paris Seine, 9 quai St. Bernard, 75252 Paris, France; CNRS UMR7622, Developmental Biology Lab, Sorbonne Université, 9 quai St. Bernard, 75252 Paris, France
| | - Damien Cladière
- Université Paris Cité, CNRS, Institut Jacques Monod, 15 rue Hélène Brion, 75013 Paris, France; Sorbonne Université, Institut de Biologie Paris Seine, 9 quai St. Bernard, 75252 Paris, France; CNRS UMR7622, Developmental Biology Lab, Sorbonne Université, 9 quai St. Bernard, 75252 Paris, France
| | - Katja Wassmann
- Université Paris Cité, CNRS, Institut Jacques Monod, 15 rue Hélène Brion, 75013 Paris, France; Sorbonne Université, Institut de Biologie Paris Seine, 9 quai St. Bernard, 75252 Paris, France; CNRS UMR7622, Developmental Biology Lab, Sorbonne Université, 9 quai St. Bernard, 75252 Paris, France.
| | - Eulalie Buffin
- Université Paris Cité, CNRS, Institut Jacques Monod, 15 rue Hélène Brion, 75013 Paris, France; Sorbonne Université, Institut de Biologie Paris Seine, 9 quai St. Bernard, 75252 Paris, France; CNRS UMR7622, Developmental Biology Lab, Sorbonne Université, 9 quai St. Bernard, 75252 Paris, France.
| |
Collapse
|
3
|
Pan B, Bruno M, Macfarlan TS, Akera T. Meiosis-specific distal cohesion site decoupled from the kinetochore. Nat Commun 2025; 16:2116. [PMID: 40032846 PMCID: PMC11876576 DOI: 10.1038/s41467-025-57438-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
Primary constriction of the M-phase chromosome serves as a marker for the kinetochore position. Underlying this observation is the concept that the kinetochore is spatially linked with the pericentromere where sister-chromatids are cohered. Here, we find an unconventional chromatid-cohesion pattern in Peromyscus oocytes, with sister chromatids cohered at a chromosome end, spatially separated from the kinetochore. This distal locus enriches cohesin protectors specifically during meiosis, and chromosomes with this additional cohesion site exhibit enhanced cohesin protection at anaphase I compared to those without it, implying an adaptive evolution to ensure cohesion during meiosis. The distal locus corresponds to an additional centromeric satellite block, located far from the satellite block building the kinetochore. Analyses on three Peromyscus species reveal that the internal satellite consistently assembles the kinetochore in mitosis and meiosis, whereas the distal satellite selectively enriches cohesin protectors in meiosis to promote cohesion. Our study demonstrates that cohesion regulation is flexible, controlling chromosome segregation in a cell-type dependent manner.
Collapse
Affiliation(s)
- Bo Pan
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Melania Bruno
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Todd S Macfarlan
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Takashi Akera
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
4
|
Wang D, Liu X, Chen B, Shang Y, Wan T, Zhang S, Liu H, Shi Y, Chen X, Sun H. Down-regulation of miR-138-5p in PP2A KO mice promoted apoptosis of spermatocytes. Mol Biol Rep 2024; 51:1147. [PMID: 39535579 DOI: 10.1007/s11033-024-10096-5] [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: 03/13/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Protein phosphatase 2 A (PP2A) is known to have a pivotal and diverse functions in various physiological processes. In the previous study, we utilized the cre-loxp system to generate germ cell-specific knockout mice for the PP2A catalytic subunit alpha subunit (Ppp2cacKO). METHODS AND RESULTS Using high-throughput miRNA sequencing of testis tissues and real‑time PCR, we have identified a notable decrease in the expression of miR-138-5p in the testes of Ppp2cacKO mice. Our findings indicate that miR-138-5p plays a role in the regulation of apoptosis and proliferation of GC2 cells. Furthermore, bioinformatics analyses suggested that miR-138- 5p may target the transcriptional repressor Trps1. Consistent with these predictions, we observed a significant upregulation of Trps1 in the testes of Ppp2cacKO mice. Through transfection experiments, we have validated the negative regulation of Trps1 expression by miR-138-5p in GC2 cells. CONCLUSION Our study indicates that the down-regulation of miR-138-5p in PP2A KO mice, which targets Trps1 to promote spermatocyte apoptosis.
Collapse
Affiliation(s)
- Danni Wang
- Center of Reproduction, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University,Changzhou Medical Center, Nanjing Medical University, No. 68 Gehu Road, Jiangsu, Changzhou, 213003, China
| | - Xing Liu
- Center of Reproduction, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University,Changzhou Medical Center, Nanjing Medical University, No. 68 Gehu Road, Jiangsu, Changzhou, 213003, China
| | - Bingyan Chen
- Center of Reproduction, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University,Changzhou Medical Center, Nanjing Medical University, No. 68 Gehu Road, Jiangsu, Changzhou, 213003, China
| | - Yuwei Shang
- Center of Reproduction, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University,Changzhou Medical Center, Nanjing Medical University, No. 68 Gehu Road, Jiangsu, Changzhou, 213003, China
| | - Ting Wan
- Changzhou Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu Province, 213003, China
| | - Shu Zhang
- Center of Reproduction, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University,Changzhou Medical Center, Nanjing Medical University, No. 68 Gehu Road, Jiangsu, Changzhou, 213003, China
| | - Huijun Liu
- Center of Reproduction, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University,Changzhou Medical Center, Nanjing Medical University, No. 68 Gehu Road, Jiangsu, Changzhou, 213003, China
| | - Yichao Shi
- Center of Reproduction, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University,Changzhou Medical Center, Nanjing Medical University, No. 68 Gehu Road, Jiangsu, Changzhou, 213003, China
| | - Xia Chen
- Center of Reproduction, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University,Changzhou Medical Center, Nanjing Medical University, No. 68 Gehu Road, Jiangsu, Changzhou, 213003, China
| | - Huiting Sun
- Center of Reproduction, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University,Changzhou Medical Center, Nanjing Medical University, No. 68 Gehu Road, Jiangsu, Changzhou, 213003, China.
| |
Collapse
|
5
|
Mihalas BP, Pieper GH, Aboelenain M, Munro L, Srsen V, Currie CE, Kelly DA, Hartshorne GM, Telfer EE, McAinsh AD, Anderson RA, Marston AL. Age-dependent loss of cohesion protection in human oocytes. Curr Biol 2024; 34:117-131.e5. [PMID: 38134935 PMCID: PMC7617652 DOI: 10.1016/j.cub.2023.11.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 11/05/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023]
Abstract
Aneuploid human eggs (oocytes) are a major cause of infertility, miscarriage, and chromosomal disorders. Such aneuploidies increase greatly as women age, with defective linkages between sister chromatids (cohesion) in meiosis as a common cause. We found that loss of a specific pool of the cohesin protector protein, shugoshin 2 (SGO2), may contribute to this phenomenon. Our data indicate that SGO2 preserves sister chromatid cohesion in meiosis by protecting a "cohesin bridge" between sister chromatids. In human oocytes, SGO2 localizes to both sub-centromere cups and the pericentromeric bridge, which spans the sister chromatid junction. SGO2 normally colocalizes with cohesin; however, in meiosis II oocytes from older women, SGO2 is frequently lost from the pericentromeric bridge and sister chromatid cohesion is weakened. MPS1 and BUB1 kinase activities maintain SGO2 at sub-centromeres and the pericentromeric bridge. Removal of SGO2 throughout meiosis I by MPS1 inhibition reduces cohesion protection, increasing the incidence of single chromatids at meiosis II. Therefore, SGO2 deficiency in human oocytes can exacerbate the effects of maternal age by rendering residual cohesin at pericentromeres vulnerable to loss in anaphase I. Our data show that impaired SGO2 localization weakens cohesion integrity and may contribute to the increased incidence of aneuploidy observed in human oocytes with advanced maternal age.
Collapse
Affiliation(s)
- Bettina P Mihalas
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Gerard H Pieper
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Mansour Aboelenain
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK; Theriogenology department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Lucy Munro
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Vlastimil Srsen
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Cerys E Currie
- Centre for Mechanochemical Cell Biology & Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK
| | - David A Kelly
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Geraldine M Hartshorne
- Centre for Mechanochemical Cell Biology & Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK; University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
| | - Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Andrew D McAinsh
- Centre for Mechanochemical Cell Biology & Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK
| | - Richard A Anderson
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Adele L Marston
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK.
| |
Collapse
|
6
|
Wassmann K. Separase Control and Cohesin Cleavage in Oocytes: Should I Stay or Should I Go? Cells 2022; 11:3399. [PMID: 36359795 PMCID: PMC9656630 DOI: 10.3390/cells11213399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 10/19/2023] Open
Abstract
The key to gametogenesis is the proper execution of a specialized form of cell division named meiosis. Prior to the meiotic divisions, the recombination of maternal and paternal chromosomes creates new genetic combinations necessary for fitness and adaptation to an ever-changing environment. Two rounds of chromosome segregation -meiosis I and II- have to take place without intermediate S-phase and lead to the creation of haploid gametes harboring only half of the genetic material. Importantly, the segregation patterns of the two divisions are fundamentally different and require adaptation of the mitotic cell cycle machinery to the specificities of meiosis. Separase, the enzyme that cleaves Rec8, a subunit of the cohesin complex constituting the physical connection between sister chromatids, has to be activated twice: once in meiosis I and immediately afterwards, in meiosis II. Rec8 is cleaved on chromosome arms in meiosis I and in the centromere region in meiosis II. This step-wise cohesin removal is essential to generate gametes of the correct ploidy and thus, embryo viability. Hence, separase control and Rec8 cleavage must be perfectly controlled in time and space. Focusing on mammalian oocytes, this review lays out what we know and what we still ignore about this fascinating mechanism.
Collapse
Affiliation(s)
- Katja Wassmann
- Institut Jacques Monod, Université Paris Cité, CNRS, 75013 Paris, France
| |
Collapse
|
7
|
Ogushi S, Rattani A, Godwin J, Metson J, Schermelleh L, Nasmyth K. Loss of sister kinetochore co-orientation and peri-centromeric cohesin protection after meiosis I depends on cleavage of centromeric REC8. Dev Cell 2021; 56:3100-3114.e4. [PMID: 34758289 PMCID: PMC8629431 DOI: 10.1016/j.devcel.2021.10.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/28/2021] [Accepted: 10/18/2021] [Indexed: 11/27/2022]
Abstract
Protection of peri-centromeric (periCEN) REC8 cohesin from Separase and sister kinetochore (KT) attachment to microtubules emanating from the same spindle pole (co-orientation) ensures that sister chromatids remain associated after meiosis I. Both features are lost during meiosis II, resulting in sister chromatid disjunction and the production of haploid gametes. By transferring spindle-chromosome complexes (SCCs) between meiosis I and II in mouse oocytes, we discovered that both sister KT co-orientation and periCEN cohesin protection depend on the SCC, and not the cytoplasm. Moreover, the catalytic activity of Separase at meiosis I is necessary not only for converting KTs from a co- to a bi-oriented state but also for deprotection of periCEN cohesion, and cleavage of REC8 may be the key event. Crucially, selective cleavage of REC8 in the vicinity of KTs is sufficient to destroy co-orientation in univalent chromosomes, albeit not in bivalents where resolution of chiasmata may also be required.
Collapse
Affiliation(s)
- Sugako Ogushi
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; The Hakubi Center for Advanced Research, Kyoto University, Kyoto 606-8501, Japan.
| | - Ahmed Rattani
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Jonathan Godwin
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Jean Metson
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | | | - Kim Nasmyth
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK.
| |
Collapse
|
8
|
Mengoli V, Jonak K, Lyzak O, Lamb M, Lister LM, Lodge C, Rojas J, Zagoriy I, Herbert M, Zachariae W. Deprotection of centromeric cohesin at meiosis II requires APC/C activity but not kinetochore tension. EMBO J 2021; 40:e106812. [PMID: 33644894 PMCID: PMC8013787 DOI: 10.15252/embj.2020106812] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 01/03/2023] Open
Abstract
Genome haploidization involves sequential loss of cohesin from chromosome arms and centromeres during two meiotic divisions. At centromeres, cohesin's Rec8 subunit is protected from separase cleavage at meiosis I and then deprotected to allow its cleavage at meiosis II. Protection of centromeric cohesin by shugoshin-PP2A seems evolutionarily conserved. However, deprotection has been proposed to rely on spindle forces separating the Rec8 protector from cohesin at metaphase II in mammalian oocytes and on APC/C-dependent destruction of the protector at anaphase II in yeast. Here, we have activated APC/C in the absence of sister kinetochore biorientation at meiosis II in yeast and mouse oocytes, and find that bipolar spindle forces are dispensable for sister centromere separation in both systems. Furthermore, we show that at least in yeast, protection of Rec8 by shugoshin and inhibition of separase by securin are both required for the stability of centromeric cohesin at metaphase II. Our data imply that related mechanisms preserve the integrity of dyad chromosomes during the short metaphase II of yeast and the prolonged metaphase II arrest of mammalian oocytes.
Collapse
Affiliation(s)
- Valentina Mengoli
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
- Present address:
Institute for Research in BiomedicineUniversità della Svizzera ItalianaBellinzonaSwitzerland
| | - Katarzyna Jonak
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
| | - Oleksii Lyzak
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
| | - Mahdi Lamb
- Biosciences InstituteCentre for LifeTimes SquareNewcastle UniversityNewcastle upon TyneUK
| | - Lisa M Lister
- Biosciences InstituteCentre for LifeTimes SquareNewcastle UniversityNewcastle upon TyneUK
| | - Chris Lodge
- Biosciences InstituteCentre for LifeTimes SquareNewcastle UniversityNewcastle upon TyneUK
| | - Julie Rojas
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
| | - Ievgeniia Zagoriy
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
- Present address:
EMBL HeidelbergHeidelbergGermany
| | - Mary Herbert
- Biosciences InstituteCentre for LifeTimes SquareNewcastle UniversityNewcastle upon TyneUK
| | - Wolfgang Zachariae
- Laboratory of Chromosome BiologyMax Planck Institute of BiochemistryMartinsriedGermany
| |
Collapse
|
9
|
Gryaznova Y, Keating L, Touati SA, Cladière D, El Yakoubi W, Buffin E, Wassmann K. Kinetochore individualization in meiosis I is required for centromeric cohesin removal in meiosis II. EMBO J 2021; 40:e106797. [PMID: 33644892 PMCID: PMC8013791 DOI: 10.15252/embj.2020106797] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
Partitioning of the genome in meiosis occurs through two highly specialized cell divisions, named meiosis I and meiosis II. Step-wise cohesin removal is required for chromosome segregation in meiosis I, and sister chromatid segregation in meiosis II. In meiosis I, mono-oriented sister kinetochores appear as fused together when examined by high-resolution confocal microscopy, whereas they are clearly separated in meiosis II, when attachments are bipolar. It has been proposed that bipolar tension applied by the spindle is responsible for the physical separation of sister kinetochores, removal of cohesin protection, and chromatid separation in meiosis II. We show here that this is not the case, and initial separation of sister kinetochores occurs already in anaphase I independently of bipolar spindle forces applied on sister kinetochores, in mouse oocytes. This kinetochore individualization depends on separase cleavage activity. Crucially, without kinetochore individualization in meiosis I, bivalents when present in meiosis II oocytes separate into chromosomes and not sister chromatids. This shows that whether centromeric cohesin is removed or not is determined by the kinetochore structure prior to meiosis II.
Collapse
Affiliation(s)
- Yulia Gryaznova
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Leonor Keating
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Sandra A Touati
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Damien Cladière
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Warif El Yakoubi
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
- Present address:
Cell and Developmental Biology CenterNational Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMDUSA
| | - Eulalie Buffin
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| | - Katja Wassmann
- Institut de Biologie Paris SeineSorbonne UniversitéParisFrance
- CNRS UMR7622 Developmental Biology LabSorbonne UniversitéParisFrance
| |
Collapse
|
10
|
Li J, Zhang HY, Wang F, Sun QY, Qian WP. The Cyclin B2/CDK1 Complex Conservatively Inhibits Separase Activity in Oocyte Meiosis II. Front Cell Dev Biol 2021; 9:648053. [PMID: 33777955 PMCID: PMC7993350 DOI: 10.3389/fcell.2021.648053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/09/2021] [Indexed: 11/15/2022] Open
Abstract
Recently, we have reported that the cyclin B2/CDK1 complex regulates homologous chromosome segregation through inhibiting separase activity in oocyte meiosis I, which further elucidates the compensation of cyclin B2 on cyclin B1’s function in meiosis I. However, whether cyclin B2/CDK1 complex also negatively regulates separase activity during oocyte meiosis II remains unknown. In the present study, we investigated the function of cyclin B2 in meiosis II of oocyte. We found that stable cyclin B2 expression impeded segregation of sister chromatids after oocyte parthenogenetic activation. Consistently, stable cyclin B2 inhibited separase activation, while introduction of non-phosphorylatable separase mutant rescued chromatid separation in the stable cyclin B2-expressed oocytes. Therefore, the cyclin B2/CDK1 complex conservatively regulates separase activity via inhibitory phosphorylation of separase in both meiosis I and meiosis II of mouse oocyte.
Collapse
Affiliation(s)
- Jian Li
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Hong-Yong Zhang
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Feng Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Wei-Ping Qian
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.,Guangdong Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen PKU-HKUST Medical Center, Shenzhen, China
| |
Collapse
|
11
|
Functioning mechanisms of Shugoshin-1 in centromeric cohesion during mitosis. Essays Biochem 2021; 64:289-297. [PMID: 32451529 DOI: 10.1042/ebc20190077] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022]
Abstract
Proper regulation of centromeric cohesion is required for faithful chromosome segregation that prevents chromosomal instability. Extensive studies have identified and established the conserved protein Shugoshin (Sgo1/2) as an essential protector for centromeric cohesion. In this review, we summarize the current understanding of how Shugoshin-1 (Sgo1) protects centromeric cohesion at the molecular level. Targeting of Sgo1 to inner centromeres is required for its proper function of cohesion protection. We therefore discuss about the molecular mechanisms that install Sgo1 onto inner centromeres. At metaphase-to-anaphase transition, Sgo1 at inner centromeres needs to be disabled for the subsequent sister-chromatid segregation. A few recent studies suggest interesting models to explain how it is achieved. These models are discussed as well.
Collapse
|
12
|
Goguet-Rubio P, Amin P, Awal S, Vigneron S, Charrasse S, Mechali F, Labbé JC, Lorca T, Castro A. PP2A-B55 Holoenzyme Regulation and Cancer. Biomolecules 2020; 10:biom10111586. [PMID: 33266510 PMCID: PMC7700614 DOI: 10.3390/biom10111586] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 01/05/2023] Open
Abstract
Protein phosphorylation is a post-translational modification essential for the control of the activity of most enzymes in the cell. This protein modification results from a fine-tuned balance between kinases and phosphatases. PP2A is one of the major serine/threonine phosphatases that is involved in the control of a myriad of different signaling cascades. This enzyme, often misregulated in cancer, is considered a tumor suppressor. In this review, we will focus on PP2A-B55, a particular holoenzyme of the family of the PP2A phosphatases whose specific role in cancer development and progression has only recently been highlighted. The discovery of the Greatwall (Gwl)/Arpp19-ENSA cascade, a new pathway specifically controlling PP2A-B55 activity, has been shown to be frequently altered in cancer. Herein, we will review the current knowledge about the mechanisms controlling the formation and the regulation of the activity of this phosphatase and its misregulation in cancer.
Collapse
|
13
|
Ma JY, Li S, Chen LN, Schatten H, Ou XH, Sun QY. Why is oocyte aneuploidy increased with maternal aging? J Genet Genomics 2020; 47:659-671. [PMID: 33184002 DOI: 10.1016/j.jgg.2020.04.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/19/2022]
Abstract
One of the main causes of pregnancy failure and fetus abortion is oocyte aneuploidy, which is increased with maternal aging. Numerous possible causes of oocyte aneuploidy in aged women have been proposed, including cross-over formation defect, cohesin loss, spindle deformation, spindle assembly checkpoint malfunction, microtubule-kinetochore attachment failure, kinetochore mis-orientation, mitochondria dysfunction-induced increases in reactive oxygen species, protein over-acetylation, and DNA damage. However, it still needs to be answered if these aneuploidization factors have inherent relations, and how to prevent chromosome aneuploidy in aged oocytes. Epidemiologically, oocyte aneuploidy has been found to be weakly associated with higher homocysteine concentrations, obesity, ionizing radiation and even seasonality. In this review, we summarize the research progress and present an integrated view of oocyte aneuploidization.
Collapse
Affiliation(s)
- Jun-Yu Ma
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Sen Li
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Lei-Ning Chen
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Xiang-Hong Ou
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
14
|
Wang H, Qiu P, Zhu S, Zhang M, Li Y, Zhang M, Wang X, Shang J, Qu B, Liu J, Zhao Q. SET nuclear proto-oncogene gene expression is associated with microsatellite instability in human colorectal cancer identified by co-expression analysis. Dig Liver Dis 2020; 52:339-346. [PMID: 31495599 DOI: 10.1016/j.dld.2019.07.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUNDS AND AIMS Microsatellite instability (MSI) is one of the promising biomarkers in human colorectal cancers (CRCs), and it is influenced by an intricate gene interaction network. Hence, we aimed to identify and validate hub genes associated with MSI CRC and to illustrate its underlying mechanisms. METHODS Weighted gene co-expression network analysis (WGCNA) was used to investigate potential regulatory targets and relationships between key modules and hub genes associated with MSI CRC. RESULTS In the red module (r = 0.83), SET nuclear proto-oncogene (SET) was selected due to its high intra-modular connectivity and module membership. In the test sets, SET expression was downregulated in MSI CRCs compared to that in microsatellite stability (MSS) CRCs. SET expression level had a good performance in stratifying patients into MSI or MSS CRCs (area under the curve = 0.953). Moreover, the BRAF V600E mutation was highly associated with SET expression, and MSI/HLA- samples showed lower levels of SET mRNA expression than MSS/HLA- samples. Finally, gene set enrichment analysis (GSEA) indicated that patients in the SET low expression group were enriched in base excision repair. CONCLUSION SET was identified and validated as a novel potential biomarker in MSI CRCs, and SET probably acts through regulating the base excision repair pathway.
Collapse
Affiliation(s)
- Haizhou Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Peishan Qiu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Shuyun Zhu
- Department of Gastroenterology, The Central Hospital of Enshi Autonomous Prefecture, Enshi, China
| | - Mengna Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Yizhang Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Meng Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Xiaobing Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Jian Shang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Bing Qu
- Department of Gastroenterology, The Central Hospital of Enshi Autonomous Prefecture, Enshi, China.
| | - Jing Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China.
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China; Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China.
| |
Collapse
|
15
|
Keating L, Touati SA, Wassmann K. A PP2A-B56-Centered View on Metaphase-to-Anaphase Transition in Mouse Oocyte Meiosis I. Cells 2020; 9:E390. [PMID: 32046180 PMCID: PMC7072534 DOI: 10.3390/cells9020390] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/12/2022] Open
Abstract
Meiosis is required to reduce to haploid the diploid genome content of a cell, generating gametes-oocytes and sperm-with the correct number of chromosomes. To achieve this goal, two specialized cell divisions without intermediate S-phase are executed in a time-controlled manner. In mammalian female meiosis, these divisions are error-prone. Human oocytes have an exceptionally high error rate that further increases with age, with significant consequences for human fertility. To understand why errors in chromosome segregation occur at such high rates in oocytes, it is essential to understand the molecular players at work controlling these divisions. In this review, we look at the interplay of kinase and phosphatase activities at the transition from metaphase-to-anaphase for correct segregation of chromosomes. We focus on the activity of PP2A-B56, a key phosphatase for anaphase onset in both mitosis and meiosis. We start by introducing multiple roles PP2A-B56 occupies for progression through mitosis, before laying out whether or not the same principles may apply to the first meiotic division in oocytes, and describing the known meiosis-specific roles of PP2A-B56 and discrepancies with mitotic cell cycle regulation.
Collapse
Affiliation(s)
- Leonor Keating
- Mammalian Oocyte Meiosis (MOM) UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France; (L.K.); (S.A.T.)
- CNRS UMR7622 Developmental Biology Lab, Sorbonne Université, 75005 Paris, France
| | - Sandra A. Touati
- Mammalian Oocyte Meiosis (MOM) UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France; (L.K.); (S.A.T.)
- CNRS UMR7622 Developmental Biology Lab, Sorbonne Université, 75005 Paris, France
| | - Katja Wassmann
- Mammalian Oocyte Meiosis (MOM) UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, 75005 Paris, France; (L.K.); (S.A.T.)
- CNRS UMR7622 Developmental Biology Lab, Sorbonne Université, 75005 Paris, France
| |
Collapse
|
16
|
Schmitz ML, Higgins JMG, Seibert M. Priming chromatin for segregation: functional roles of mitotic histone modifications. Cell Cycle 2020; 19:625-641. [PMID: 31992120 DOI: 10.1080/15384101.2020.1719585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Posttranslational modifications (PTMs) of histone proteins are important for various cellular processes including regulation of gene expression and chromatin structure, DNA damage response and chromosome segregation. Here we comprehensively review mitotic histone PTMs, in particular phosphorylations, and discuss their interplay and functions in the control of dynamic protein-protein interactions as well as their contribution to centromere and chromosome structure and function during cell division. Histone phosphorylations can create binding sites for mitotic regulators such as the chromosomal passenger complex, which is required for correction of erroneous spindle attachments and chromosome bi-orientation. Other histone PTMs can alter the structural properties of nucleosomes and the accessibility of chromatin. Epigenetic marks such as lysine methylations are maintained during mitosis and may also be important for mitotic transcription as well as bookmarking of transcriptional states to ensure the transmission of gene expression programs through cell division. Additionally, histone phosphorylation can dissociate readers of methylated histones without losing epigenetic information. Through all of these processes, mitotic histone PTMs play a functional role in priming the chromatin for faithful chromosome segregation and preventing genetic instability, one of the characteristic hallmarks of cancer cells.
Collapse
Affiliation(s)
- M Lienhard Schmitz
- Institute of Biochemistry, Medical Faculty, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| | - Jonathan M G Higgins
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Markus Seibert
- Institute of Biochemistry, Medical Faculty, Member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
17
|
A new regulatory mechanism of protein phosphatase 2A activity via SET in acute myeloid leukemia. Blood Cancer J 2020; 10:3. [PMID: 31913266 PMCID: PMC6949222 DOI: 10.1038/s41408-019-0270-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 12/04/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy. Although novel emerging drugs are available, the overall prognosis remains poor and new therapeutic approaches are required. PP2A phosphatase is a key regulator of cell homeostasis and is recurrently inactivated in AML. The anticancer activity of several PP2A-activating drugs (e.g., FTY720) depends on their interaction with the SET oncoprotein, an endogenous PP2A inhibitor that is overexpressed in 30% of AML cases. Elucidation of SET regulatory mechanisms may therefore provide novel targeted therapies for SET-overexpressing AMLs. Here, we show that upregulation of protein kinase p38β is a common event in AML. We provide evidence that p38β potentiates SET-mediated PP2A inactivation by two mechanisms: facilitating SET cytoplasmic translocation through CK2 phosphorylation, and directly binding to and stabilizing the SET protein. We demonstrate the importance of this new regulatory mechanism in primary AML cells from patients and in zebrafish xenograft models. Accordingly, combination of the CK2 inhibitor CX-4945, which retains SET in the nucleus, and FTY720, which disrupts the SET-PP2A binding in the cytoplasm, significantly reduces the viability and migration of AML cells. In conclusion, we show that the p38β/CK2/SET axis represents a new potential therapeutic pathway in AML patients with SET-dependent PP2A inactivation.
Collapse
|
18
|
Asai Y, Fukuchi K, Tanno Y, Koitabashi-Kiyozuka S, Kiyozuka T, Noda Y, Matsumura R, Koizumi T, Watanabe A, Nagata K, Watanabe Y, Terada Y. Aurora B kinase activity is regulated by SET/TAF1 on Sgo2 at the inner centromere. J Cell Biol 2019; 218:3223-3236. [PMID: 31527146 PMCID: PMC6781429 DOI: 10.1083/jcb.201811060] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/19/2019] [Accepted: 07/09/2019] [Indexed: 01/23/2023] Open
Abstract
Phosphorylation of kinetochore proteins destabilizes improper kinetochore–microtubule attachments. Asai et al. find that SET/TAF1, an inhibitor of the PP2A phosphatase, binds shugoshin 2 and corrects erroneous kinetochore–microtubule attachment by maintaining Aurora B kinase activity. Therefore, SET has a key role in establishing chromosome bi-orientation by balancing Aurora B and PP2A activity. The accurate regulation of phosphorylation at the kinetochore is essential for establishing chromosome bi-orientation. Phosphorylation of kinetochore proteins by the Aurora B kinase destabilizes improper kinetochore–microtubule attachments, whereas the phosphatase PP2A has a counteracting role. Imbalanced phosphoregulation leads to error-prone chromosome segregation and aneuploidy, a hallmark of cancer cells. However, little is known about the molecular events that control the balance of phosphorylation at the kinetochore. Here, we show that localization of SET/TAF1, an oncogene product, to centromeres maintains Aurora B kinase activity by inhibiting PP2A, thereby correcting erroneous kinetochore–microtubule attachment. SET localizes at the inner centromere by interacting directly with shugoshin 2, with SET levels declining at increased distances between kinetochore pairs, leading to establishment of chromosome bi-orientation. Moreover, SET overexpression induces chromosomal instability by disrupting kinetochore–microtubule attachment. Thus, our findings reveal the novel role of SET in fine-tuning the phosphorylation level at the kinetochore by balancing the activities of Aurora B and PP2A.
Collapse
Affiliation(s)
- Yuichiro Asai
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Koh Fukuchi
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Yuji Tanno
- Bioscience Department, Veritas Corporation, Tokyo, Japan
| | - Saki Koitabashi-Kiyozuka
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Tatsuyuki Kiyozuka
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Yuko Noda
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Rieko Matsumura
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Tetsuo Koizumi
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Atsushi Watanabe
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Kyosuke Nagata
- Department of Infection Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | | | - Yasuhiko Terada
- Department of Chemistry and Biochemistry, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| |
Collapse
|
19
|
Qu Q, Zhang Q, Yang L, Chen Y, Liu H. SET binding to Sgo1 inhibits Sgo1-cohesin interactions and promotes chromosome segregation. J Cell Biol 2019; 218:2514-2528. [PMID: 31227592 PMCID: PMC6683731 DOI: 10.1083/jcb.201810096] [Citation(s) in RCA: 4] [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: 10/18/2018] [Revised: 03/29/2019] [Accepted: 05/15/2019] [Indexed: 12/28/2022] Open
Abstract
At anaphase onset, Sgo1 function of cohesion protection must be disabled to allow timely chromosome segregation, but how this is achieved is not fully understood. Here, we show that SET, a known PP2A inhibitor, directly binds to a domain in Sgo1 in close proximity to the cohesin-binding motif. The Sgo1-cohesin binding can be disrupted by SET in a dose-dependent manner in vitro as well as by SET overexpression in cells, suggesting that SET is also an inhibitor to the Sgo1-cohesin binding. Furthermore, the SET binding-deficient Sgo1 mutant fully supports centromeric cohesion protection but delays chromosome segregation, suggesting that the SET-Sgo1 binding is required for timely chromosome segregation. Moreover, overexpression of SET WT, not the Sgo1 binding-deficient mutant, exacerbates the occurrence of cohesion fatigue in MG132-arrested cells. Conversely, SET depletion delays it. Thus, we propose that a major function of SET during mitosis is to disrupt the Sgo1-cohesin interaction, thereby promoting centromeric cohesion de-protection and timely chromosome segregation at anaphase onset.
Collapse
Affiliation(s)
- Qianhui Qu
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Qian Zhang
- Department of Biochemistry and Molecular Biology and Tulane Aging Center, Tulane University Health Science Center, New Orleans, LA
| | - Lu Yang
- Department of Biochemistry and Molecular Biology and Tulane Aging Center, Tulane University Health Science Center, New Orleans, LA
| | - Yujue Chen
- Department of Biochemistry and Molecular Biology and Tulane Aging Center, Tulane University Health Science Center, New Orleans, LA
| | - Hong Liu
- Department of Biochemistry and Molecular Biology and Tulane Aging Center, Tulane University Health Science Center, New Orleans, LA
| |
Collapse
|
20
|
Zhong J, Ren X, Chen Z, Zhang H, Zhou L, Yuan J, Li P, Chen X, Liu W, Wu D, Yang X, Liu J. miR-21-5p promotes lung adenocarcinoma progression partially through targeting SET/TAF-Iα. Life Sci 2019; 231:116539. [PMID: 31176779 DOI: 10.1016/j.lfs.2019.06.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/29/2019] [Accepted: 06/05/2019] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Although SET(I2PP2A) and miRNAs are reported to play a pivotal role in lung cancer, the underlying mechanisms have remained obscure. To address this issue, we investigated how miRNAs and SET participate in the progression of lung cancer. METHODS miRNAs that target SET were predicted from multiple miRNA databases. Three human NSCLC cell lines and two normal lung cell lines were used to evaluate aberrant miRNA and SET expressions. A dual luciferase reporter assay system was employed to verify the interaction between miRNA and SET. Stable miRNA knockdown and SET overexpression in A549 cells were achieved through lentivirus transfection; the corresponding influences on lung cancer progression were also examined. RESULTS In this study, A549 was the sole cell line to lack SET/TAF-Iα expression, which was inversely correlated with the up-regulation of miR-21-5p. SET was subsequently revealed as the direct target site of miR-21-5p in A549 cells. The stable miR-21-5p knockdown and SET/TAF-Iα overexpression were shown to markedly enhance the expression of SET/TAF-Iα and to inhibit the migration, invasion, proliferation as well as the in vivo tumorigenicity of A549 cells. CONCLUSION We suggest that SET/TAF-Iα might be a tumor suppressing factor regulated by miR-21-5p in lung adenocarcinoma. This might provide a target for lung adenocarcinoma therapy.
Collapse
Affiliation(s)
- Jiacheng Zhong
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Xiaohu Ren
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Zhihong Chen
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Hang Zhang
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Li Zhou
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Jianhui Yuan
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Ping Li
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Xiao Chen
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Wei Liu
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Desheng Wu
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Xifei Yang
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Jianjun Liu
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China.
| |
Collapse
|
21
|
Gheldof A, Mackay DJG, Cheong Y, Verpoest W. Genetic diagnosis of subfertility: the impact of meiosis and maternal effects. J Med Genet 2019; 56:271-282. [PMID: 30728173 PMCID: PMC6581078 DOI: 10.1136/jmedgenet-2018-105513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 12/24/2018] [Accepted: 12/27/2018] [Indexed: 02/06/2023]
Abstract
During reproductive age, approximately one in seven couples are confronted with fertility problems. While the aetiology is diverse, including infections, metabolic diseases, hormonal imbalances and iatrogenic effects, it is becoming increasingly clear that genetic factors have a significant contribution. Due to the complex nature of infertility that often hints at a multifactorial cause, the search for potentially causal gene mutations in idiopathic infertile couples has remained difficult. Idiopathic infertility patients with a suspicion of an underlying genetic cause can be expected to have mutations in genes that do not readily affect general health but are only essential in certain processes connected to fertility. In this review, we specifically focus on genes involved in meiosis and maternal-effect processes, which are of critical importance for reproduction and initial embryonic development. We give an overview of genes that have already been linked to infertility in human, as well as good candidates which have been described in other organisms. Finally, we propose a phenotypic range in which we expect an optimal diagnostic yield of a meiotic/maternal-effect gene panel.
Collapse
Affiliation(s)
- Alexander Gheldof
- Center for Medical Genetics, Universitair Ziekenhuis Brussel, Brussels, Belgium
- Reproduction and Genetics Department, Vrije Universiteit Brussel, Brussels, Belgium
| | - Deborah J G Mackay
- Faculty of Medicine, University of Southampton, Southampton University Hospital, Southampton, UK
| | - Ying Cheong
- Complete Fertility, Human Development of Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Willem Verpoest
- Reproduction and Genetics Department, Vrije Universiteit Brussel, Brussels, Belgium
- Center for Reproductive Medicine, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
22
|
Vallardi G, Allan LA, Crozier L, Saurin AT. Division of labour between PP2A-B56 isoforms at the centromere and kinetochore. eLife 2019; 8:e42619. [PMID: 30829571 PMCID: PMC6398977 DOI: 10.7554/elife.42619] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/03/2019] [Indexed: 11/13/2022] Open
Abstract
PP2A-B56 is a serine/threonine phosphatase complex that regulates several major mitotic processes, including sister chromatid cohesion, kinetochore-microtubule attachment and the spindle assembly checkpoint. We show here that these key functions are divided between different B56 isoforms that localise to either the centromere or kinetochore. The centromeric isoforms rely on a specific interaction with Sgo2, whereas the kinetochore isoforms bind preferentially to BubR1 and other proteins containing an LxxIxE motif. In addition to these selective binding partners, Sgo1 helps to anchor PP2A-B56 at both locations: it collaborates with BubR1 to maintain B56 at the kinetochore and it helps to preserve the Sgo2/B56 complex at the centromere. A series of chimaeras were generated to map the critical region in B56 down to a small C-terminal loop that regulates the key interactions and defines B56 localisation. Together, this study describes how different PP2A-B56 complexes utilise isoform-specific interactions to control distinct processes during mitosis.
Collapse
Affiliation(s)
- Giulia Vallardi
- Division of Cellular Medicine, School of MedicineUniversity of DundeeDundeeUnited Kingdom
| | - Lindsey A Allan
- Division of Cellular Medicine, School of MedicineUniversity of DundeeDundeeUnited Kingdom
| | - Lisa Crozier
- Division of Cellular Medicine, School of MedicineUniversity of DundeeDundeeUnited Kingdom
| | - Adrian T Saurin
- Division of Cellular Medicine, School of MedicineUniversity of DundeeDundeeUnited Kingdom
| |
Collapse
|
23
|
Seibert M, Krüger M, Watson NA, Sen O, Daum JR, Slotman JA, Braun T, Houtsmuller AB, Gorbsky GJ, Jacob R, Kracht M, Higgins JMG, Schmitz ML. CDK1-mediated phosphorylation at H2B serine 6 is required for mitotic chromosome segregation. J Cell Biol 2019; 218:1164-1181. [PMID: 30765437 PMCID: PMC6446833 DOI: 10.1083/jcb.201806057] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/17/2018] [Accepted: 01/17/2019] [Indexed: 12/11/2022] Open
Abstract
Faithful mitotic chromosome segregation is required for the maintenance of genomic stability. We discovered the phosphorylation of histone H2B at serine 6 (H2B S6ph) as a new chromatin modification site and found that this modification occurs during the early mitotic phases at inner centromeres and pericentromeric heterochromatin. This modification is directly mediated by cyclin B1-associated CDK1, and indirectly by Aurora B, and is antagonized by PP1-mediated dephosphorylation. H2B S6ph impairs chromatin binding of the histone chaperone SET (I2PP2A), which is important for mitotic fidelity. Injection of phosphorylation-specific H2B S6 antibodies in mitotic cells caused anaphase defects with impaired chromosome segregation and incomplete cytokinesis. As H2B S6ph is important for faithful chromosome separation, this modification may contribute to the prevention chromosomal instability and aneuploidy which frequently occur in cancer cells.
Collapse
Affiliation(s)
- Markus Seibert
- Institute of Biochemistry, Justus-Liebig-University, Member of the German Center for Lung Research, Giessen, Germany
| | - Marcus Krüger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Nikolaus A Watson
- Cell Division Biology Research Group, Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, England, UK
| | - Onur Sen
- Cell Division Biology Research Group, Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, England, UK
| | - John R Daum
- Cell Cycle and Cancer Biology Research Program, Oklahoma Medical Research Foundation, and Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Johan A Slotman
- Department of Pathology, Josephine Nefkens Institute, Erasmus Optical Imaging Centre, Erasmus MC, Rotterdam, Netherlands
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Adriaan B Houtsmuller
- Department of Pathology, Josephine Nefkens Institute, Erasmus Optical Imaging Centre, Erasmus MC, Rotterdam, Netherlands
| | - Gary J Gorbsky
- Cell Cycle and Cancer Biology Research Program, Oklahoma Medical Research Foundation, and Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Ralf Jacob
- Department of Cell Biology and Cell Pathology, Philipps University of Marburg, Marburg, Germany
| | - Michael Kracht
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University, Member of the German Center for Lung Research, Giessen, Germany
| | - Jonathan M G Higgins
- Cell Division Biology Research Group, Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, England, UK
| | - M Lienhard Schmitz
- Institute of Biochemistry, Justus-Liebig-University, Member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
24
|
Spangenberg V, Arakelyan M, Galoyan E, Pankin M, Petrosyan R, Stepanyan I, Grishaeva T, Danielyan F, Kolomiets O. Extraordinary centromeres: differences in the meiotic chromosomes of two rock lizards species Darevskia portschinskii and Darevskia raddei. PeerJ 2019; 7:e6360. [PMID: 30723630 PMCID: PMC6359900 DOI: 10.7717/peerj.6360] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/28/2018] [Indexed: 01/27/2023] Open
Abstract
According to the synthesis of 30 years of multidisciplinary studies, parthenogenetic species of rock lizards of genus Darevskia were formed as a result of different combination patterns of interspecific hybridization of the four bisexual parental species: Darevskia raddei, D. mixta, D. valentini, and D. portschinskii. In particular, D. portschinskii and D. raddei are considered as the parental species for the parthenogenetic species D. rostombekowi. Here for the first time, we present the result of comparative immunocytochemical study of primary spermatocyte nuclei spreads from the leptotene to diplotene stages of meiotic prophase I in two species: D. portschinskii and D. raddei. We observed similar chromosome lengths for both synaptonemal complex (SC) karyotypes as well as a similar number of crossing over sites. However, unexpected differences in the number and distribution of anti-centromere antibody (ACA) foci were detected in the SC structure of bivalents of the two species. In all examined D. portschinskii spermatocyte nuclei, one immunostained centromere focus was detected per SC bivalent. In contrast, in almost every studied D. raddei nuclei we identified three to nine SCs with additional immunostained ACA foci per SC bivalent. Thus, the obtained results allow us to identify species-specific karyotype features, previously not been detected using conventional mitotic chromosome analysis. Presumably the additional centromere foci are result of epigenetic chromatin modifications. We assume that this characteristic of the D. raddei karyotype could represent useful marker for the future studies of parthenogenetic species hybrid karyotypes related to D. raddei.
Collapse
Affiliation(s)
| | - Marine Arakelyan
- Department of Zoology, Yerevan State University, Yerevan, Armenia
| | - Eduard Galoyan
- Zoological Museum, Moscow State University, Moscow, Russia
| | - Mark Pankin
- Vavilov Institute of General Genetics, Moscow, Russian Federation
| | | | - Ilona Stepanyan
- Scientific Center of Zoology and Hydroecology, Yerevan, Armenia
| | | | - Felix Danielyan
- Department of Zoology, Yerevan State University, Yerevan, Armenia
| | - Oxana Kolomiets
- Vavilov Institute of General Genetics, Moscow, Russian Federation
| |
Collapse
|
25
|
Nilsson J. Protein phosphatases in the regulation of mitosis. J Cell Biol 2018; 218:395-409. [PMID: 30446607 PMCID: PMC6363451 DOI: 10.1083/jcb.201809138] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 12/15/2022] Open
Abstract
The accurate segregation of genetic material to daughter cells during mitosis depends on the precise coordination and regulation of hundreds of proteins by dynamic phosphorylation. Mitotic kinases are major regulators of protein function, but equally important are protein phosphatases that balance their actions, their coordinated activity being essential for accurate chromosome segregation. Phosphoprotein phosphatases (PPPs) that dephosphorylate phosphoserine and phosphothreonine residues are increasingly understood as essential regulators of mitosis. In contrast to kinases, the lack of a pronounced peptide-binding cleft on the catalytic subunit of PPPs suggests that these enzymes are unlikely to be specific. However, recent exciting insights into how mitotic PPPs recognize specific substrates have revealed that they are as specific as kinases. Furthermore, the activities of PPPs are tightly controlled at many levels to ensure that they are active only at the proper time and place. Here, I will discuss substrate selection and regulation of mitotic PPPs focusing mainly on animal cells and explore how these actions control mitosis, as well as important unanswered questions.
Collapse
Affiliation(s)
- Jakob Nilsson
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Avet C, Denoyelle C, L'Hôte D, Petit F, Guigon CJ, Cohen-Tannoudji J, Simon V. GnRH regulates the expression of its receptor accessory protein SET in pituitary gonadotropes. PLoS One 2018; 13:e0201494. [PMID: 30052687 PMCID: PMC6063425 DOI: 10.1371/journal.pone.0201494] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/16/2018] [Indexed: 01/26/2023] Open
Abstract
Reproductive function is under the control of the neurohormone GnRH, which activates a G-protein-coupled receptor (GnRHR) expressed in pituitary gonadotrope cells. GnRHR activates a complex signaling network to regulate synthesis and secretion of the two gonadotropin hormones, luteinizing hormone and follicle-stimulating hormone, both regulating gametogenesis and steroidogenesis in gonads. Recently, in an attempt to identify the mechanisms underlying GnRHR signaling plasticity, we identified the first interacting partner of GnRHR, the proto-oncogene SET. We showed that SET binds to intracellular domains of GnRHR to enhance its coupling to cAMP pathway in αT3-1 gonadotrope cells. Here, we demonstrate that SET protein is rapidly regulated by GnRH, which increases SET phosphorylation state and decreases dose-dependently SET protein level. Our results highlight a post-translational regulation of SET protein involving the proteasome pathway. We determined that SET phosphorylation upon GnRH stimulation is mediated by PKC and that PKC mediates GnRH-induced SET down-regulation. Phosphorylation on serine 9 targets SET for degradation into the proteasome. Furthermore, a non-phosphorylatable SET mutant on serine 9 is resistant to GnRH-induced down-regulation. Altogether, these data suggest that GnRH-induced SET phosphorylation on serine 9 mediates SET protein down-regulation through the proteasome pathway. Noteworthy, SET down-regulation was also observed in response to pulsatile GnRH stimulation in LβT2 gonadotrope cells as well as in vivo in prepubertal female mice supporting its physiological relevance. In conclusion, this study highlights a regulation of SET protein by the neurohormone GnRH and identifies some of the mechanisms involved.
Collapse
Affiliation(s)
- Charlotte Avet
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS UMR 8251, INSERM U1133, Biologie Fonctionnelle et Adaptative, Physiologie de l'axe gonadotrope, Paris, France
| | - Chantal Denoyelle
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS UMR 8251, INSERM U1133, Biologie Fonctionnelle et Adaptative, Physiologie de l'axe gonadotrope, Paris, France
| | - David L'Hôte
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS UMR 8251, INSERM U1133, Biologie Fonctionnelle et Adaptative, Physiologie de l'axe gonadotrope, Paris, France
| | - Florence Petit
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS UMR 8251, INSERM U1133, Biologie Fonctionnelle et Adaptative, Physiologie de l'axe gonadotrope, Paris, France
| | - Céline J Guigon
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS UMR 8251, INSERM U1133, Biologie Fonctionnelle et Adaptative, Physiologie de l'axe gonadotrope, Paris, France
| | - Joëlle Cohen-Tannoudji
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS UMR 8251, INSERM U1133, Biologie Fonctionnelle et Adaptative, Physiologie de l'axe gonadotrope, Paris, France
| | - Violaine Simon
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS UMR 8251, INSERM U1133, Biologie Fonctionnelle et Adaptative, Physiologie de l'axe gonadotrope, Paris, France
| |
Collapse
|
27
|
Regulation of the meiotic divisions of mammalian oocytes and eggs. Biochem Soc Trans 2018; 46:797-806. [PMID: 29934303 PMCID: PMC6103459 DOI: 10.1042/bst20170493] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 12/28/2022]
Abstract
Initiated by luteinizing hormone and finalized by the fertilizing sperm, the mammalian oocyte completes its two meiotic divisions. The first division occurs in the mature Graafian follicle during the hours preceding ovulation and culminates in an extreme asymmetric cell division and the segregation of the two pairs of homologous chromosomes. The newly created mature egg rearrests at metaphase of the second meiotic division prior to ovulation and only completes meiosis following a Ca2+ signal initiated by the sperm at gamete fusion. Here, we review the cellular events that govern the passage of the oocyte through meiosis I with a focus on the role of the spindle assembly checkpoint in regulating its timing. In meiosis II, we examine how the egg achieves its arrest and how the fertilization Ca2+ signal allows the initiation of embryo development.
Collapse
|
28
|
Wolf PG, Cuba Ramos A, Kenzel J, Neumann B, Stemmann O. Studying meiotic cohesin in somatic cells reveals that Rec8-containing cohesin requires Stag3 to function and is regulated by Wapl and sororin. J Cell Sci 2018; 131:jcs212100. [PMID: 29724914 DOI: 10.1242/jcs.212100] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 04/26/2018] [Indexed: 01/21/2023] Open
Abstract
The DNA-embracing, ring-shaped multiprotein complex cohesin mediates sister chromatid cohesion and is stepwise displaced in mitosis by Wapl and separase (also known as ESPL1) to facilitate anaphase. Proper regulation of chromosome cohesion throughout meiosis is critical for preventing formation of aneuploid gametes, which are associated with trisomies and infertility in humans. Studying cohesion in meiocytes is complicated by their difficult experimental amenability and the absence of cohesin turnover. Here, we use cultured somatic cells to unravel fundamental aspects of meiotic cohesin. When expressed in Hek293 cells, the kleisin Rec8 displays no affinity for the peripheral cohesin subunits Stag1 or Stag2 and remains cytoplasmic. However, co-expression of Stag3 is sufficient for Rec8 to enter the nucleus, load onto chromatin, and functionally replace its mitotic counterpart Scc1 (also known as RAD21) during sister chromatid cohesion and dissolution. Rec8-Stag3 cohesin physically interacts with Pds5, Wapl and sororin (also known as CDCA5). Importantly, Rec8-Stag3 cohesin is shown to be susceptible to Wapl-dependent ring opening and sororin-mediated protection. These findings exemplify that our model system is suitable to rapidly generate testable predictions for important unresolved issues of meiotic cohesion regulation.
Collapse
Affiliation(s)
- Peter G Wolf
- Chair of Genetics, University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Alexander Cuba Ramos
- Chair of Genetics, University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Julia Kenzel
- Chair of Genetics, University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Brigitte Neumann
- Chair of Genetics, University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Olaf Stemmann
- Chair of Genetics, University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| |
Collapse
|
29
|
Do Gametes Woo? Evidence for Their Nonrandom Union at Fertilization. Genetics 2018; 207:369-387. [PMID: 28978771 DOI: 10.1534/genetics.117.300109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/10/2017] [Indexed: 12/18/2022] Open
Abstract
A fundamental tenet of inheritance in sexually reproducing organisms such as humans and laboratory mice is that gametes combine randomly at fertilization, thereby ensuring a balanced and statistically predictable representation of inherited variants in each generation. This principle is encapsulated in Mendel's First Law. But exceptions are known. With transmission ratio distortion, particular alleles are preferentially transmitted to offspring. Preferential transmission usually occurs in one sex but not both, and is not known to require interactions between gametes at fertilization. A reanalysis of our published work in mice and of data in other published reports revealed instances where any of 12 mutant genes biases fertilization, with either too many or too few heterozygotes and homozygotes, depending on the mutant gene and on dietary conditions. Although such deviations are usually attributed to embryonic lethality of the underrepresented genotypes, the evidence is more consistent with genetically-determined preferences for specific combinations of egg and sperm at fertilization that result in genotype bias without embryo loss. This unexpected discovery of genetically-biased fertilization could yield insights about the molecular and cellular interactions between sperm and egg at fertilization, with implications for our understanding of inheritance, reproduction, population genetics, and medical genetics.
Collapse
|
30
|
Mishra PK, Thapa KS, Chen P, Wang S, Hazbun TR, Basrai MA. Budding yeast CENP-A Cse4 interacts with the N-terminus of Sgo1 and regulates its association with centromeric chromatin. Cell Cycle 2018; 17:11-23. [PMID: 28980861 DOI: 10.1080/15384101.2017.1380129] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Shugoshin is an evolutionarily conserved protein, which is involved in tension sensing on mitotic chromosomes, kinetochore biorientation, and protection of centromeric (CEN) cohesin for faithful chromosome segregation. Interaction of the C-terminus of Sgo1 with phosphorylated histone H2A regulates its association with CEN and pericentromeric (peri-CEN) chromatin, whereas mutations in histone H3 selectively compromise the association of Sgo1 with peri-CEN but not CEN chromatin. Given that histone H3 is absent from CEN and is replaced by a histone H3 variant CENP-ACse4, we investigated if CENP-ACse4 interacts with Sgo1 and promotes its association with the CEN chromatin. In this study, we found that Sgo1 interacts with CENP-ACse4 in vivo and in vitro. The N-terminus coiled-coil domain of Sgo1 without the C-terminus (sgo1-NT) is sufficient for its interaction with CENP-ACse4, association with CEN but not the peri-CEN, and this CEN association is cell cycle dependent with maximum enrichment in mitosis. In agreement with the role of CENP-ACse4 in CEN maintenance of Sgo1, depletion of CENP-ACse4 results in the loss of Sgo1 and sgo1-NT from the CEN chromatin. The N-terminus of Sgo1 is required for genome stability as a mutant lacking the N-terminus (sgo1-CT) exhibits increased chromosome missegregation when compared to a sgo1-NT mutant. In summary, our results define a novel role for the N-terminus of Sgo1 in CENP-ACse4 mediated recruitment of Sgo1 to CEN chromatin for faithful chromosome segregation.
Collapse
Affiliation(s)
- Prashant K Mishra
- a Genetics Branch , National Cancer Institute , National Institutes of Health , Bethesda , MD , USA
| | - Kriti S Thapa
- b Purdue University , Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University Center for Cancer Research (PUCCR) , West Lafayette , IN , USA
| | - Panyue Chen
- b Purdue University , Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University Center for Cancer Research (PUCCR) , West Lafayette , IN , USA
| | - Suyu Wang
- b Purdue University , Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University Center for Cancer Research (PUCCR) , West Lafayette , IN , USA
| | - Tony R Hazbun
- b Purdue University , Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University Center for Cancer Research (PUCCR) , West Lafayette , IN , USA
| | - Munira A Basrai
- a Genetics Branch , National Cancer Institute , National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
31
|
Drosophila protein phosphatases 2A B' Wdb and Wrd regulate meiotic centromere localization and function of the MEI-S332 Shugoshin. Proc Natl Acad Sci U S A 2017; 114:12988-12993. [PMID: 29158400 PMCID: PMC5724294 DOI: 10.1073/pnas.1718450114] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Proper segregation of chromosomes in meiosis is essential to prevent miscarriages and birth defects. This requires that sister chromatids maintain cohesion at the centromere as cohesion is released on the chromatid arms when the homologs segregate at anaphase I. The Shugoshin proteins preserve centromere cohesion by protecting the cohesin complex from cleavage, and this has been shown in yeasts to be mediated by recruitment of the protein phosphatase 2A B' (PP2A B'). In metazoans, delineation of the role of PP2A B' in meiosis has been hindered by its myriad of other essential roles. The Drosophila Shugoshin MEI-S332 can bind directly to both of the B' regulatory subunits of PP2A, Wdb and Wrd, in yeast two-hybrid experiments. Exploiting experimental advantages of Drosophila spermatogenesis, we found that the Wdb subunit localizes first along chromosomes in meiosis I, becoming restricted to the centromere region as MEI-S332 binds. Wdb and MEI-S332 show colocalization at the centromere region until release of sister-chromatid cohesion at the metaphase II/anaphase II transition. MEI-S332 is necessary for Wdb localization, but, additionally, both Wdb and Wrd are required for MEI-S332 localization. Thus, rather than MEI-S332 being hierarchical to PP2A B', these proteins reciprocally ensure centromere localization of the complex. We analyzed functional relationships between MEI-S332 and the two forms of PP2A by quantifying meiotic chromosome segregation defects in double or triple mutants. These studies revealed that both Wdb and Wrd contribute to MEI-S332's ability to ensure accurate segregation of sister chromatids, but, as in centromere localization, they do not act solely downstream of MEI-S332.
Collapse
|
32
|
El Yakoubi W, Buffin E, Cladière D, Gryaznova Y, Berenguer I, Touati SA, Gómez R, Suja JA, van Deursen JM, Wassmann K. Mps1 kinase-dependent Sgo2 centromere localisation mediates cohesin protection in mouse oocyte meiosis I. Nat Commun 2017; 8:694. [PMID: 28947820 PMCID: PMC5612927 DOI: 10.1038/s41467-017-00774-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 07/27/2017] [Indexed: 01/10/2023] Open
Abstract
A key feature of meiosis is the step-wise removal of cohesin, the protein complex holding sister chromatids together, first from arms in meiosis I and then from the centromere region in meiosis II. Centromeric cohesin is protected by Sgo2 from Separase-mediated cleavage, in order to maintain sister chromatids together until their separation in meiosis II. Failures in step-wise cohesin removal result in aneuploid gametes, preventing the generation of healthy embryos. Here, we report that kinase activities of Bub1 and Mps1 are required for Sgo2 localisation to the centromere region. Mps1 inhibitor-treated oocytes are defective in centromeric cohesin protection, whereas oocytes devoid of Bub1 kinase activity, which cannot phosphorylate H2A at T121, are not perturbed in cohesin protection as long as Mps1 is functional. Mps1 and Bub1 kinase activities localise Sgo2 in meiosis I preferentially to the centromere and pericentromere respectively, indicating that Sgo2 at the centromere is required for protection.In meiosis I centromeric cohesin is protected by Sgo2 from Separase-mediated cleavage ensuring that sister chromatids are kept together until their separation in meiosis II. Here the authors demonstrate that Bub1 and Mps1 kinase activities are required for Sgo2 localisation to the centromere region.
Collapse
Affiliation(s)
- Warif El Yakoubi
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
| | - Eulalie Buffin
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
| | - Damien Cladière
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
| | - Yulia Gryaznova
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
| | - Inés Berenguer
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
- Unidad de Biología Celular, Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Sandra A Touati
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France
- Chromosome Segregation Laboratory, Lincoln's Inn Fields Laboratory, The Francis Crick Institute, London, WC2A 3LY, UK
| | - Rocío Gómez
- Unidad de Biología Celular, Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - José A Suja
- Unidad de Biología Celular, Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Jan M van Deursen
- Department of Pediatric and Adolescent Medicine and Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Katja Wassmann
- Sorbonne Universités, UPMC Univ. Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75005, France.
- CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75005, France.
| |
Collapse
|
33
|
Chu N, Gui Y, Qiu X, Zhang N, Li L, Li D, Tang W, Gober HJ, Zhang B, Wang L. The effect of DHEA on apoptosis and cohesin levels in oocytes in aged mice. Biosci Trends 2017; 11:427-438. [PMID: 28717062 DOI: 10.5582/bst.2017.01108] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Female fertility declines with age as the number of ovarian follicles decreases and aneuploidy increases. Degradation of the cohesin complex might be responsible for age-related aneuploidy. Dehydroepiandrosterone (DHEA) can improve the ovarian reserve and reduce the rate of aneuploidy, but the relationship between DHEA and cohesin levels in oocytes is still unknown. The aim of the current study was to evaluate the effect of the supplement DHEA on ovarian function, including the number of follicles and cohesin levels in oocytes. C57BL/6J mice at 3 weeks, 6 weeks, 12 weeks, 6 months, and 10 months of age were used to obtain a systematic view into follicle apoptosis and cohesin levels in oocytes. Nine-month-old C57BL/6J mice were administered saline (n = 5), 17β-estradiol (100 µg/kg per day, n = 5), or DHEA (5mg/Kg per day, n = 5). After 4 weeks, aged mice were weighed and sacrificed, and ovarian tissue samples were prepared. Anti-VASA staining and HE staining were used to count the number of follicles. Anti-γH2AX staining and TUNEL were used to measure follicle apoptosis and immunofluorescent staining was used to detect the levels of three oocyte cohesin subunits: REC8, SMC1β, and SMC3. Administration of the supplements 17β-estradiol and DHEA to aged mice increased the number of primordial and primary follicles and decreased the age-related apoptosis of follicles. Levels of the cohesin subunits REC8 and SMC1β declined with age, but DHEA and 17β-estradiol tended to delay that decline. The supplement DHEA increased the number of primordial and primary follicles in aged mice by inhibiting follicle apoptosis and tended to delay the decrease in cohesin levels in oocytes.
Collapse
Affiliation(s)
- Nan Chu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| | - Yuyan Gui
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Xuemin Qiu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Na Zhang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Lisha Li
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Dajin Li
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- The Academy of Integrative Medicine of Fudan University
| | - Wei Tang
- Department of Surgery, Graduate School of Medicine, The University of Tokyo
| | | | - Bin Zhang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| | - Ling Wang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Fudan University Shanghai Medical College
- The Academy of Integrative Medicine of Fudan University
| |
Collapse
|
34
|
Abstract
Genome haploidization involves two meiotic divisions following a single round of DNA replication. In this issue of Developmental Cell, Argüello-Miranda et al. (2017) show that production and packaging of the single-copy genome into gametes during the second meiotic division is coordinated by a conserved casein kinase 1.
Collapse
Affiliation(s)
- Mary Herbert
- Newcastle Fertility Centre, Institute of Genetic Medicine, Newcastle University, Centre for Life, Times Square, Newcastle upon Tyne NE1 4EP, UK.
| | - Attila Toth
- Institute of Physiological Chemistry, Medical Faculty of TU Dresden, Fiedlerstrasse 42, 01307 Dresden, Germany.
| |
Collapse
|
35
|
Krishnan S, Smits AH, Vermeulen M, Reinberg D. Phospho-H1 Decorates the Inter-chromatid Axis and Is Evicted along with Shugoshin by SET during Mitosis. Mol Cell 2017; 67:579-593.e6. [PMID: 28781233 PMCID: PMC5562512 DOI: 10.1016/j.molcel.2017.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/26/2017] [Accepted: 07/06/2017] [Indexed: 12/22/2022]
Abstract
Precise control of sister chromatid separation during mitosis is pivotal to maintaining genomic integrity. Yet, the regulatory mechanisms involved are not well understood. Remarkably, we discovered that linker histone H1 phosphorylated at S/T18 decorated the inter-chromatid axial DNA on mitotic chromosomes. Sister chromatid resolution during mitosis required the eviction of such H1S/T18ph by the chaperone SET, with this process being independent of and most likely downstream of arm-cohesin dissociation. SET also directed the disassembly of Shugoshins in a polo-like kinase 1-augmented manner, aiding centromere resolution. SET ablation compromised mitotic fidelity as evidenced by unresolved sister chromatids with marked accumulation of H1S/T18ph and centromeric Shugoshin. Thus, chaperone-assisted eviction of linker histones and Shugoshins is a fundamental step in mammalian mitotic progression. Our findings also elucidate the functional implications of the decades-old observation of mitotic linker histone phosphorylation, serving as a paradigm to explore the role of linker histones in bio-signaling processes.
Collapse
Affiliation(s)
- Swathi Krishnan
- Howard Hughes Medical Institute, New York University Langone School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Langone School of Medicine, New York, NY 10016, USA
| | - Arne H Smits
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, 6525 GA Nijmegen, the Netherlands
| | - Danny Reinberg
- Howard Hughes Medical Institute, New York University Langone School of Medicine, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, New York University Langone School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
36
|
Jonak K, Zagoriy I, Oz T, Graf P, Rojas J, Mengoli V, Zachariae W. APC/C-Cdc20 mediates deprotection of centromeric cohesin at meiosis II in yeast. Cell Cycle 2017; 16:1145-1152. [PMID: 28514186 DOI: 10.1080/15384101.2017.1320628] [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] [Indexed: 10/19/2022] Open
Abstract
Cells undergoing meiosis produce haploid gametes through one round of DNA replication followed by 2 rounds of chromosome segregation. This requires that cohesin complexes, which establish sister chromatid cohesion during S phase, are removed in a stepwise manner. At meiosis I, the separase protease triggers the segregation of homologous chromosomes by cleaving cohesin's Rec8 subunit on chromosome arms. Cohesin persists at centromeres because the PP2A phosphatase, recruited by the shugoshin protein, dephosphorylates Rec8 and thereby protects it from cleavage. While chromatids disjoin upon cleavage of centromeric Rec8 at meiosis II, it was unclear how and when centromeric Rec8 is liberated from its protector PP2A. One proposal is that bipolar spindle forces separate PP2A from Rec8 as cells enter metaphase II. We show here that sister centromere biorientation is not sufficient to "deprotect" Rec8 at meiosis II in yeast. Instead, our data suggest that the ubiquitin-ligase APC/CCdc20 removes PP2A from centromeres by targeting for degradation the shugoshin Sgo1 and the kinase Mps1. This implies that Rec8 remains protected until entry into anaphase II when it is phosphorylated concurrently with the activation of separase. Here, we provide further support for this model and speculate on its relevance to mammalian oocytes.
Collapse
Affiliation(s)
- Katarzyna Jonak
- a Laboratory of Chromosome Biology , Max Planck Institute of Biochemistry , Martinsried , Germany
| | - Ievgeniia Zagoriy
- a Laboratory of Chromosome Biology , Max Planck Institute of Biochemistry , Martinsried , Germany
| | - Tugce Oz
- a Laboratory of Chromosome Biology , Max Planck Institute of Biochemistry , Martinsried , Germany
| | - Peter Graf
- a Laboratory of Chromosome Biology , Max Planck Institute of Biochemistry , Martinsried , Germany
| | - Julie Rojas
- a Laboratory of Chromosome Biology , Max Planck Institute of Biochemistry , Martinsried , Germany
| | - Valentina Mengoli
- a Laboratory of Chromosome Biology , Max Planck Institute of Biochemistry , Martinsried , Germany
| | - Wolfgang Zachariae
- a Laboratory of Chromosome Biology , Max Planck Institute of Biochemistry , Martinsried , Germany
| |
Collapse
|
37
|
Zhang N, Pati D. Biology and insights into the role of cohesin protease separase in human malignancies. Biol Rev Camb Philos Soc 2017; 92:2070-2083. [PMID: 28177203 DOI: 10.1111/brv.12321] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/05/2017] [Accepted: 01/12/2017] [Indexed: 12/11/2022]
Abstract
Separase, an enzyme that resolves sister chromatid cohesion during the metaphase-to-anaphase transition, plays a pivotal role in chromosomal segregation and cell division. Separase protein, encoded by the extra spindle pole bodies like 1 (ESPL1) gene, is overexpressed in numerous human cancers including breast, bone, brain, and prostate. Separase is oncogenic, and its overexpression is sufficient to induce mammary tumours in mice. Either acute or chronic overexpression of separase in mouse mammary glands leads to aneuploidy and tumorigenesis, and inhibition of separase enzymatic activity decreases the growth of human breast tumour xenografts in mice. This review focuses on the biology of and insights into the molecular mechanisms of separase as an oncogene, and its significance and implications for human cancers.
Collapse
Affiliation(s)
- Nenggang Zhang
- Departments of Pediatrics and Molecular and Cellular Biology, Texas Children's Cancer Center, Baylor College of Medicine, 1102 Bates St., FC1220, Houston, TX 77030, U.S.A
| | - Debananda Pati
- Departments of Pediatrics and Molecular and Cellular Biology, Texas Children's Cancer Center, Baylor College of Medicine, 1102 Bates St., FC1220, Houston, TX 77030, U.S.A
| |
Collapse
|
38
|
Argüello-Miranda O, Zagoriy I, Mengoli V, Rojas J, Jonak K, Oz T, Graf P, Zachariae W. Casein Kinase 1 Coordinates Cohesin Cleavage, Gametogenesis, and Exit from M Phase in Meiosis II. Dev Cell 2017; 40:37-52. [PMID: 28017619 DOI: 10.1016/j.devcel.2016.11.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/15/2016] [Accepted: 11/22/2016] [Indexed: 01/08/2023]
Abstract
Meiosis consists of DNA replication followed by two consecutive nuclear divisions and gametogenesis or spore formation. While meiosis I has been studied extensively, less is known about the regulation of meiosis II. Here we show that Hrr25, the conserved casein kinase 1δ of budding yeast, links three mutually independent key processes of meiosis II. First, Hrr25 induces nuclear division by priming centromeric cohesin for cleavage by separase. Hrr25 simultaneously phosphorylates Rec8, the cleavable subunit of cohesin, and removes from centromeres the cohesin protector composed of shugoshin and the phosphatase PP2A. Second, Hrr25 initiates the sporulation program by inducing the synthesis of membranes that engulf the emerging nuclei at anaphase II. Third, Hrr25 mediates exit from meiosis II by activating pathways that trigger the destruction of M-phase-promoting kinases. Thus, Hrr25 synchronizes formation of the single-copy genome with gamete differentiation and termination of meiosis.
Collapse
Affiliation(s)
- Orlando Argüello-Miranda
- Laboratory of Chromosome Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Ievgeniia Zagoriy
- Laboratory of Chromosome Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Valentina Mengoli
- Laboratory of Chromosome Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Julie Rojas
- Laboratory of Chromosome Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Katarzyna Jonak
- Laboratory of Chromosome Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Tugce Oz
- Laboratory of Chromosome Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Peter Graf
- Laboratory of Chromosome Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Wolfgang Zachariae
- Laboratory of Chromosome Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany.
| |
Collapse
|
39
|
El Yakoubi W, Wassmann K. Meiotic Divisions: No Place for Gender Equality. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1002:1-17. [PMID: 28600780 DOI: 10.1007/978-3-319-57127-0_1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In multicellular organisms the fusion of two gametes with a haploid set of chromosomes leads to the formation of the zygote, the first cell of the embryo. Accurate execution of the meiotic cell division to generate a female and a male gamete is required for the generation of healthy offspring harboring the correct number of chromosomes. Unfortunately, meiosis is error prone. This has severe consequences for fertility and under certain circumstances, health of the offspring. In humans, female meiosis is extremely error prone. In this chapter we will compare male and female meiosis in humans to illustrate why and at which frequency errors occur, and describe how this affects pregnancy outcome and health of the individual. We will first introduce key notions of cell division in meiosis and how they differ from mitosis, followed by a detailed description of the events that are prone to errors during the meiotic divisions.
Collapse
Affiliation(s)
- Warif El Yakoubi
- Sorbonne Universités, UPMC Univ Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75252, France.,CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75252, France
| | - Katja Wassmann
- Sorbonne Universités, UPMC Univ Paris 06, Institut de Biologie Paris Seine (IBPS), UMR7622, Paris, 75252, France. .,CNRS, IBPS, UMR7622 Developmental Biology Lab, Paris, 75252, France.
| |
Collapse
|
40
|
Zhou W, Gao J, Ma J, Cao L, Zhang C, Zhu Y, Dong A, Shen WH. Distinct roles of the histone chaperones NAP1 and NRP and the chromatin-remodeling factor INO80 in somatic homologous recombination in Arabidopsis thaliana. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2016; 88:397-410. [PMID: 27352805 DOI: 10.1111/tpj.13256] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 06/24/2016] [Indexed: 05/10/2023]
Abstract
Homologous recombination (HR) of nuclear DNA occurs within the context of a highly complex chromatin structure. Despite extensive studies of HR in diverse organisms, mechanisms regulating HR within the chromatin context remain poorly elucidated. Here we investigate the role and interplay of the histone chaperones NUCLEOSOME ASSEMBLY PROTEIN1 (NAP1) and NAP1-RELATED PROTEIN (NRP) and the ATP-dependent chromatin-remodeling factor INOSITOL AUXOTROPHY80 (INO80) in regulating somatic HR in Arabidopsis thaliana. We show that simultaneous knockout of the four AtNAP1 genes and the two NRP genes in the sextuple mutant m123456-1 barely affects normal plant growth and development. Interestingly, compared with the respective AtNAP1 (m123-1 and m1234-1) or NRP (m56-1) loss-of-function mutants, the sextuple mutant m123456-1 displays an enhanced plant hypersensitivity to UV or bleomycin treatments. Using HR reporter constructs, we show that AtNAP1 and NRP act in parallel to synergistically promote somatic HR. Distinctively, the AtINO80 loss-of-function mutation (atino80-5) is epistatic to m56-1 in plant phenotype and telomere length but hypostatic to m56-1 in HR determinacy. Further analyses show that expression of HR machinery genes and phosphorylation of H2A.X (γ-H2A.X) are not impaired in the mutants. Collectively, our study indicates that NRP and AtNAP1 synergistically promote HR upstream of AtINO80-mediated chromatin remodeling after the formation of γ-H2A.X foci during DNA damage repair.
Collapse
Affiliation(s)
- Wangbin Zhou
- Department of Biochemistry, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, International Associated Laboratory of CNRS-Fudan-HUNAU on Plant Epigenome Research, School of Life Sciences, Institute of Plant Biology, Fudan University, Shanghai, 20043, China
| | - Juan Gao
- Department of Biochemistry, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, International Associated Laboratory of CNRS-Fudan-HUNAU on Plant Epigenome Research, School of Life Sciences, Institute of Plant Biology, Fudan University, Shanghai, 20043, China
- Institut de Biologie Moléculaire des Plantes (IBMP), UPR2357 CNRS, Université de Strasbourg, 12 rue du Général Zimmer, Strasbourg Cédex, 67084, France
- School of Life Sciences, Shanghai Key Laboratory of Bio-Energy Crops, Shanghai University, Shanghai, 200444, China
| | - Jing Ma
- Department of Biochemistry, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, International Associated Laboratory of CNRS-Fudan-HUNAU on Plant Epigenome Research, School of Life Sciences, Institute of Plant Biology, Fudan University, Shanghai, 20043, China
| | - Lin Cao
- Department of Biochemistry, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, International Associated Laboratory of CNRS-Fudan-HUNAU on Plant Epigenome Research, School of Life Sciences, Institute of Plant Biology, Fudan University, Shanghai, 20043, China
| | - Chi Zhang
- Department of Biochemistry, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, International Associated Laboratory of CNRS-Fudan-HUNAU on Plant Epigenome Research, School of Life Sciences, Institute of Plant Biology, Fudan University, Shanghai, 20043, China
| | - Yan Zhu
- Department of Biochemistry, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, International Associated Laboratory of CNRS-Fudan-HUNAU on Plant Epigenome Research, School of Life Sciences, Institute of Plant Biology, Fudan University, Shanghai, 20043, China
| | - Aiwu Dong
- Department of Biochemistry, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, International Associated Laboratory of CNRS-Fudan-HUNAU on Plant Epigenome Research, School of Life Sciences, Institute of Plant Biology, Fudan University, Shanghai, 20043, China
| | - Wen-Hui Shen
- Department of Biochemistry, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, International Associated Laboratory of CNRS-Fudan-HUNAU on Plant Epigenome Research, School of Life Sciences, Institute of Plant Biology, Fudan University, Shanghai, 20043, China
- Institut de Biologie Moléculaire des Plantes (IBMP), UPR2357 CNRS, Université de Strasbourg, 12 rue du Général Zimmer, Strasbourg Cédex, 67084, France
| |
Collapse
|
41
|
Webster A, Schuh M. Mechanisms of Aneuploidy in Human Eggs. Trends Cell Biol 2016; 27:55-68. [PMID: 27773484 DOI: 10.1016/j.tcb.2016.09.002] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/03/2016] [Accepted: 09/02/2016] [Indexed: 01/24/2023]
Abstract
Eggs and sperm develop through a specialized cell division called meiosis. During meiosis, the number of chromosomes is reduced by two sequential divisions in preparation for fertilization. In human female meiosis, chromosomes frequently segregate incorrectly, resulting in eggs with an abnormal number of chromosomes. When fertilized, these eggs give rise to aneuploid embryos that usually fail to develop. As women become older, errors in meiosis occur more frequently, resulting in increased risks of infertility, miscarriage, and congenital syndromes, such as Down's syndrome. Here, we review recent studies that identify the mechanisms causing aneuploidy in female meiosis, with a particular emphasis on studies in humans.
Collapse
Affiliation(s)
- Alexandre Webster
- Department of Meiosis, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, D-37077, Göttingen, Germany
| | - Melina Schuh
- Department of Meiosis, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, D-37077, Göttingen, Germany.
| |
Collapse
|
42
|
Yoon SW, Lee MS, Xaver M, Zhang L, Hong SG, Kong YJ, Cho HR, Kleckner N, Kim KP. Meiotic prophase roles of Rec8 in crossover recombination and chromosome structure. Nucleic Acids Res 2016; 44:9296-9314. [PMID: 27484478 PMCID: PMC5100558 DOI: 10.1093/nar/gkw682] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 07/22/2016] [Indexed: 12/02/2022] Open
Abstract
Rec8 is a prominent component of the meiotic prophase chromosome axis that mediates sister chromatid cohesion, homologous recombination and chromosome synapsis. Here, we explore the prophase roles of Rec8. (i) During the meiotic divisions, Rec8 phosphorylation mediates its separase-mediated cleavage. We show here that such cleavage plays no detectable role for chromosomal events of prophase. (ii) We have analyzed in detail three rec8 phospho-mutants, with 6, 24 or 29 alanine substitutions. A distinct ‘separation of function’ phenotype is revealed. In the mutants, axis formation and recombination initiation are normal, as is non-crossover recombination; in contrast, crossover (CO)-related events are defective. Moreover, the severities of these defects increase coordinately with the number of substitution mutations, consistent with the possibility that global phosphorylation of Rec8 is important for these effects. (iii) We have analyzed the roles of three kinases that phosphorylate Rec8 during prophase. Timed inhibition of Dbf4-dependent Cdc7 kinase confers defects concordant with rec8 phospho-mutant phenotypes. Inhibition of Hrr25 or Cdc5/polo-like kinase does not. Our results suggest that Rec8's prophase function, independently of cohesin cleavage, contributes to CO-specific events in conjunction with the maintenance of homolog bias at the leptotene/zygotene transition of meiotic prophase.
Collapse
Affiliation(s)
- Sang-Wook Yoon
- Department of Life Science, Chung-Ang University, Seoul 156-756, Korea
| | - Min-Su Lee
- Department of Life Science, Chung-Ang University, Seoul 156-756, Korea
| | - Martin Xaver
- Max Perutz Laboratories, Chromosome Biology, University of Vienna, Vienna Biocenter, Vienna 1030, Austria
| | - Liangran Zhang
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Soo-Gil Hong
- Department of Life Science, Chung-Ang University, Seoul 156-756, Korea
| | - Yoon-Ju Kong
- Department of Life Science, Chung-Ang University, Seoul 156-756, Korea
| | - Hong-Rae Cho
- Department of Life Science, Chung-Ang University, Seoul 156-756, Korea
| | - Nancy Kleckner
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Keun P Kim
- Department of Life Science, Chung-Ang University, Seoul 156-756, Korea
| |
Collapse
|
43
|
Tang A, Shi P, Song A, Zou D, Zhou Y, Gu P, Huang Z, Wang Q, Lin Z, Gao X. PP2A regulates kinetochore-microtubule attachment during meiosis I in oocyte. Cell Cycle 2016; 15:1450-61. [PMID: 27096707 DOI: 10.1080/15384101.2016.1175256] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Studies using in vitro cultured oocytes have indicated that the protein phosphatase 2A (PP2A), a major serine/threonine protein phosphatase, participates in multiple steps of meiosis. Details of oocyte maturation regulation by PP2A remain unclear and an in vivo model can provide more convincing information. Here, we inactivated PP2A by mutating genes encoding for its catalytic subunits (PP2Acs) in mouse oocytes. We found that eliminating both PP2Acs caused female infertility. Oocytes lacking PP2Acs failed to complete 1(st) meiotic division due to chromosome misalignment and abnormal spindle assembly. In mitosis, PP2A counteracts Aurora kinase B/C (AurkB/C) to facilitate correct kinetochore-microtubule (KT-MT) attachment. In meiosis I in oocyte, we found that PP2Ac deficiency destabilized KT-MT attachments. Chemical inhibition of AurkB/C in PP2Ac-null oocytes partly restored the formation of lateral/merotelic KT-MT attachments but not correct KT-MT attachments. Taken together, our findings demonstrate that PP2Acs are essential for chromosome alignments and regulate the formation of correct KT-MT attachments in meiosis I in oocytes.
Collapse
Affiliation(s)
- An Tang
- a State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Collaborative Innovation Center of Genetics and Development, Nanjing University , Nanjing , China
| | - Peiliang Shi
- a State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Collaborative Innovation Center of Genetics and Development, Nanjing University , Nanjing , China
| | - Anying Song
- a State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Collaborative Innovation Center of Genetics and Development, Nanjing University , Nanjing , China
| | - Dayuan Zou
- a State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Collaborative Innovation Center of Genetics and Development, Nanjing University , Nanjing , China
| | - Yue Zhou
- b State Key Laboratory of Food Science and Technology, Jiangnan University , Wuxi , China
| | - Pengyu Gu
- c Neurobiology Department , University of Massachusetts Medical School , Worcester , MA , USA
| | - Zan Huang
- d College of Animal Science & Technology, Nanjing Agricultural University , Nanjing , China
| | - Qinghua Wang
- a State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Collaborative Innovation Center of Genetics and Development, Nanjing University , Nanjing , China
| | - Zhaoyu Lin
- a State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Collaborative Innovation Center of Genetics and Development, Nanjing University , Nanjing , China
| | - Xiang Gao
- a State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Collaborative Innovation Center of Genetics and Development, Nanjing University , Nanjing , China
| |
Collapse
|
44
|
Zhang Q, Giebler HA, Isaacson MK, Nyborg JK. Eviction of linker histone H1 by NAP-family histone chaperones enhances activated transcription. Epigenetics Chromatin 2015; 8:30. [PMID: 26339295 PMCID: PMC4558729 DOI: 10.1186/s13072-015-0022-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 08/13/2015] [Indexed: 12/31/2022] Open
Abstract
Background In the Metazoan nucleus, core histones assemble the genomic DNA to form nucleosome arrays, which are further compacted into dense chromatin structures by the linker histone H1. The extraordinary density of chromatin creates an obstacle for accessing the genetic information. Regulation of chromatin dynamics is therefore critical to cellular homeostasis, and histone chaperones serve as prominent players in these processes. In the current study, we examined the role of specific histone chaperones in negotiating the inherently repressive chromatin structure during transcriptional activation. Results Using a model promoter, we demonstrate that the human nucleosome assembly protein family members hNap1 and SET/Taf1β stimulate transcription in vitro during pre-initiation complex formation, prior to elongation. This stimulatory effect is dependent upon the presence of activators, p300, and Acetyl-CoA. We show that transcription from our chromatin template is strongly repressed by H1, and that both histone chaperones enhance RNA synthesis by overcoming H1-induced repression. Importantly, both hNap1 and SET/Taf1β directly bind H1, and function to enhance transcription by evicting the linker histone from chromatin reconstituted with H1. In vivo studies demonstrate that SET/Taf1β, but not hNap1, strongly stimulates activated transcription from the chromosomally-integrated model promoter, consistent with the observation that SET/Taf1β is nuclear, whereas hNap1 is primarily cytoplasmic. Together, these observations indicate that SET/Taf1β may serve as a critical regulator of H1 dynamics and gene activation in vivo. Conclusions These studies uncover a novel function for SET that mechanistically couples transcriptional derepression with H1 dynamics. Furthermore, they underscore the significance of chaperone-dependent H1 displacement as an essential early step in the transition of a promoter from a dense chromatin state into one that is permissive to transcription factor binding and robust activation.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523-1870 USA
| | - Holli A Giebler
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523-1870 USA
| | - Marisa K Isaacson
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523-1870 USA ; Pace University, 1 Pace Plaza, New York, NY 10038 USA
| | - Jennifer K Nyborg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523-1870 USA
| |
Collapse
|
45
|
Yu C, Ji SY, Sha QQ, Sun QY, Fan HY. CRL4-DCAF1 ubiquitin E3 ligase directs protein phosphatase 2A degradation to control oocyte meiotic maturation. Nat Commun 2015; 6:8017. [PMID: 26281983 PMCID: PMC4557334 DOI: 10.1038/ncomms9017] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 07/07/2015] [Indexed: 01/10/2023] Open
Abstract
Oocyte meiosis is a specialized cell cycle that gives rise to fertilizable haploid gametes and is precisely controlled in various dimensions. We recently found that E3 ubiquitin ligase CRL4 is required for female fertility by regulating DNA hydroxymethylation to maintain oocyte survival and to promote zygotic genome reprogramming. However, not all phenotypes of CRL4-deleted oocytes could be explained by this mechanism. Here we show that CRL4 controls oocyte meiotic maturation by proteasomal degradation of protein phosphatase 2A scaffold subunit, PP2A-A. Oocyte-specific deletion of DDB1 or DCAF1 (also called VPRBP) results in delayed meiotic resumption and failure to complete meiosis I along with PP2A-A accumulation. DCAF1 directly binds to and results in the poly-ubiquitination of PP2A-A. Moreover, combined deletion of Ppp2r1a rescues the meiotic defects caused by DDB1/DCAF1 deficiency. These results provide in vivo evidence that CRL4-directed PP2A-A degradation is physiologically essential for regulating oocyte meiosis and female fertility. The E3 ubiquitin ligase CRL4 regulates oocyte survival through hydroxymethylation of genomic DNA. Here Yu et al. show that CRL4 is also required for oocytes to complete meiosis I by mediating the poly-ubiquitination and proteasomal degradation of the cell cycle regulator protein phosphatase 2A-A subunit.
Collapse
Affiliation(s)
- Chao Yu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Shu-Yan Ji
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Qian-Qian Sha
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| | - Qing-Yuan Sun
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Heng-Yu Fan
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
46
|
Singh DK, Spillane C, Siddiqi I. PATRONUS1 is expressed in meiotic prophase I to regulate centromeric cohesion in Arabidopsis and shows synthetic lethality with OSD1. BMC PLANT BIOLOGY 2015; 15:201. [PMID: 26272661 PMCID: PMC4536785 DOI: 10.1186/s12870-015-0558-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 06/18/2015] [Indexed: 05/18/2023]
Abstract
BACKGROUND Retention of sister centromere cohesion during meiosis I and its dissolution at meiosis II is necessary for balanced chromosome segregation and reduction of chromosome number. PATRONUS1 (PANS1) has recently been proposed to regulate centromere cohesion in Arabidopsis after meiosis I, during interkinesis. pans1 mutants lose centromere cohesion prematurely during interkinesis and segregate randomly at meiosis II. PANS1 protein interacts with components of the Anaphase Promoting Complex/Cyclosome (APC/C). RESULTS We show here that PANS1 protein is found mainly in prophase I of meiosis, with its level declining late in prophase I during diplotene. PANS1 also shows expression in dividing tissues. We demonstrate that, in addition to the previously reported premature loss of centromere cohesion during interkinesis, pans1 mutants show partially penetrant defects in centromere cohesion during meiosis I. We also determine that pans1 shows synthetic lethality at the level of the sporophyte, with Omission of Second Division 1 (osd1), which encodes a known inhibitor of the APC/C that is required for cell cycle progression during mitosis, as well as meiosis I and II. CONCLUSIONS Our results show that PANS1 is expressed mainly in meiosis I where it has an important function and together with previous studies indicate that PANS1 and OSD1 are part of a network linking centromere cohesion and cell cycle progression through control of APC/C activity.
Collapse
Affiliation(s)
- Dipesh Kumar Singh
- Centre for Cellular and Molecular Biology (CSIR), Uppal Road, Hyderabad, 500007, India.
| | - Charles Spillane
- Genetics and Biotechnology Lab, Plant and AgriBiosciences Research Centre (PABC), Botany and Plant Sciences, School of Natural Sciences, National University of Ireland Galway, University Road, Galway, Ireland.
| | - Imran Siddiqi
- Centre for Cellular and Molecular Biology (CSIR), Uppal Road, Hyderabad, 500007, India.
| |
Collapse
|
47
|
Dai XN, Liu S, Shao L, Gao C, Gao L, Liu JY, Cui YG. Expression of the SET protein in testes of mice at different developmental stages. Asian J Androl 2015; 16:689-93. [PMID: 24923460 PMCID: PMC4215651 DOI: 10.4103/1008-682x.129937] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
SET is a multifunctional protein involved in regulating many biological processes of the cell cycle. It is also a regulator of steroidogenesis in the ovary. However, the expression of SET protein in testis, and its function, still remains ambiguous. In this study, we observed the expression of SET in the testes of mice at different developmental stages, and have discussed its potential function in regulating spermatogenesis and androgen production. Forty-eight male mice at different developmental stages (1 week old as the infancy group; 4 weeks old as the prepubertal group; 12 weeks old as the adult group; over 12 months old as the ageing group) were used. Cellular location of SET protein in the testes was observed by immuno-histochemistry. Expression levels of Set mRNA and SET protein were analyzed by quantitative polymerase chain reaction and Western blotting. SET protein was expressed in spermatogonial cells and spermatocytes; the highest level was mainly in haploid and tetraploid cells of the prepubertal and adult groups, and Leydig cells of the adult and ageing groups. There was a low expression in Sertoli cells. Expression of Set mRNA in the prepubertal group was significantly higher than that in the adult group (P < 0.05), while expression of SET protein was at the highest level in the adult group (P < 0.05). SET protein is mainly expressed in spermatogonial cells and spermatocytes, and poorly expressed in Sertoli cells, suggesting that it is involved in spermatogenesis. Expression of SET protein in Leydig cells suggests a possible role in steroidogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yu-Gui Cui
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
48
|
Jin Z, Chung JW, Mei W, Strack S, He C, Lau GW, Yang J. Regulation of nuclear-cytoplasmic shuttling and function of Family with sequence similarity 13, member A (Fam13a), by B56-containing PP2As and Akt. Mol Biol Cell 2015; 26:1160-73. [PMID: 25609086 PMCID: PMC4357514 DOI: 10.1091/mbc.e14-08-1276] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent genome-wide association studies reveal that the FAM13A gene is associated with human lung function and a variety of lung diseases, including chronic obstructive pulmonary disease, asthma, lung cancer, and pulmonary fibrosis. The biological functions of Fam13a, however, have not been studied. In an effort to identify novel substrates of B56-containing PP2As, we found that B56-containing PP2As and Akt act antagonistically to control reversible phosphorylation of Fam13a on Ser-322. We show that Ser-322 phosphorylation acts as a molecular switch to control the subcellular distribution of Fam13a. Fam13a shuttles between the nucleus and cytoplasm. When Ser-322 is phosphorylated by Akt, the binding between Fam13a and 14-3-3 is enhanced, leading to cytoplasmic sequestration of Fam13a. B56-containing PP2As dephosphorylate phospho-Ser-322 and promote nuclear localization of Fam13a. We generated Fam13a-knockout mice. Fam13a-mutant mice are viable and healthy, indicating that Fam13a is dispensable for embryonic development and physiological functions in adult animals. Intriguingly, Fam13a has the ability to activate the Wnt pathway. Although Wnt signaling remains largely normal in Fam13a-knockout lungs, depletion of Fam13a in human lung cancer cells causes an obvious reduction in Wnt signaling activity. Our work provides important clues to elucidating the mechanism by which Fam13a may contribute to human lung diseases.
Collapse
Affiliation(s)
- Zhigang Jin
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802
| | - Jin Wei Chung
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802
| | - Wenyan Mei
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802
| | - Stefan Strack
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Chunyan He
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202
| | - Gee W Lau
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802
| | - Jing Yang
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802
| |
Collapse
|
49
|
Abstract
The shugoshin/Mei-S332 family are proteins that associate with the chromosomal region surrounding the centromere (the pericentromere) and that play multiple and distinct roles in ensuring the accuracy of chromosome segregation during both mitosis and meiosis. The underlying role of shugoshins appears to be to serve as pericentromeric adaptor proteins that recruit several different effectors to this region of the chromosome to regulate processes critical for chromosome segregation. Crucially, shugoshins undergo changes in their localization in response to the tension that is exerted on sister chromosomes by the forces of the spindle that will pull them apart. This has led to the idea that shugoshins provide a platform for activities required at the pericentromere only when sister chromosomes lack tension. Conversely, disassembly of the shugoshin pericentromeric platform may provide a signal that sister chromosomes are under tension. Here the functions and regulation of these important tension-sensitive pericentromeric proteins are discussed.
Collapse
|
50
|
Nerusheva OO, Galander S, Fernius J, Kelly D, Marston AL. Tension-dependent removal of pericentromeric shugoshin is an indicator of sister chromosome biorientation. Genes Dev 2014; 28:1291-309. [PMID: 24939933 PMCID: PMC4066400 DOI: 10.1101/gad.240291.114] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/20/2014] [Indexed: 12/24/2022]
Abstract
During mitosis and meiosis, sister chromatid cohesion resists the pulling forces of microtubules, enabling the generation of tension at kinetochores upon chromosome biorientation. How tension is read to signal the bioriented state remains unclear. Shugoshins form a pericentromeric platform that integrates multiple functions to ensure proper chromosome biorientation. Here we show that budding yeast shugoshin Sgo1 dissociates from the pericentromere reversibly in response to tension. The antagonistic activities of the kinetochore-associated Bub1 kinase and the Sgo1-bound phosphatase protein phosphatase 2A (PP2A)-Rts1 underlie a tension-dependent circuitry that enables Sgo1 removal upon sister kinetochore biorientation. Sgo1 dissociation from the pericentromere triggers dissociation of condensin and Aurora B from the centromere, thereby stabilizing the bioriented state. Conversely, forcing sister kinetochores to be under tension during meiosis I leads to premature Sgo1 removal and precocious loss of pericentromeric cohesion. Overall, we show that the pivotal role of shugoshin is to build a platform at the pericentromere that attracts activities that respond to the absence of tension between sister kinetochores. Disassembly of this platform in response to intersister kinetochore tension signals the bioriented state. Therefore, tension sensing by shugoshin is a central mechanism by which the bioriented state is read.
Collapse
Affiliation(s)
- Olga O. Nerusheva
- The Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| | - Stefan Galander
- The Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| | - Josefin Fernius
- The Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| | - David Kelly
- The Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| | - Adele L. Marston
- The Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, United Kingdom
| |
Collapse
|