1
|
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
|
2
|
El Yakoubi W, Pan B, Akera T. Hybrid female sterility due to cohesin protection errors in oocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.16.638358. [PMID: 40027736 PMCID: PMC11870456 DOI: 10.1101/2025.02.16.638358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Hybrid incompatibility can lead to lethality and sterility of F1 hybrids, contributing to speciation. Here we found that female hybrids between Mus musculus domesticus and Mus spicilegus mice are sterile due to the failure of homologous chromosome separation in oocyte meiosis I, producing aneuploid eggs. This non-separation phenotype was driven by the mis- localization of the cohesin protector, SGO2, along the chromosome arms instead of its typical centromeric enrichment, resulting in cohesin over-protection. The upstream kinase, BUB1, showed a significantly higher activity in hybrid oocytes, explaining SGO2 mis-targeting along the chromosome arm. Higher BUB1 activity was not observed in mitosis, consistent with viable hybrid mice. Cohesion defects were also evident in hybrid mice from another genus, Peromyscus , wherein cohesin protection is weakened. Defective cohesion in oocytes is a leading cause of reduced fertility especially with advanced maternal age. Our work provides evidence that a major cause of human infertility may play a positive role in promoting mammalian speciation.
Collapse
|
3
|
Wang M, Robertson D, Zou J, Spanos C, Rappsilber J, Marston AL. Molecular mechanism targeting condensin for chromosome condensation. EMBO J 2025; 44:705-735. [PMID: 39690240 PMCID: PMC11791182 DOI: 10.1038/s44318-024-00336-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024] Open
Abstract
Genomes are organised into DNA loops by the Structural Maintenance of Chromosomes (SMC) proteins. SMCs establish functional chromosomal sub-domains for DNA repair, gene expression and chromosome segregation, but how SMC activity is specifically targeted is unclear. Here, we define the molecular mechanism targeting the condensin SMC complex to specific chromosomal regions in budding yeast. A conserved pocket on the condensin HAWK subunit Ycg1 binds to chromosomal receptors carrying a related motif, CR1. In early mitosis, CR1 motifs in receptors Sgo1 and Lrs4 recruit condensin to pericentromeres and rDNA, to facilitate sister kinetochore biorientation and rDNA condensation, respectively. We additionally find that chromosome arm condensation begins as sister kinetochores come under tension, in a manner dependent on the Ycg1 pocket. We propose that multiple CR1-containing proteins recruit condensin to chromosomes and identify several additional candidates based on their sequence. Overall, we uncover the molecular mechanism that targets condensin to functionalise chromosomal domains to achieve accurate chromosome segregation during mitosis.
Collapse
Affiliation(s)
- Menglu Wang
- Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, United Kingdom
| | - Daniel Robertson
- Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, United Kingdom
| | - Juan Zou
- Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, United Kingdom
| | - Christos Spanos
- Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, United Kingdom
| | - Juri Rappsilber
- Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, United Kingdom
- Institute of Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Adele L Marston
- Centre for Cell Biology, Institute of Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, United Kingdom.
| |
Collapse
|
4
|
Polisetty SD, Bhat K, Das K, Clark I, Hardwick KG, Sanyal K. The dependence of shugoshin on Bub1-kinase activity is dispensable for the maintenance of spindle assembly checkpoint response in Cryptococcus neoformans. PLoS Genet 2025; 21:e1011552. [PMID: 39804939 PMCID: PMC11774493 DOI: 10.1371/journal.pgen.1011552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/28/2025] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
During chromosome segregation, the spindle assembly checkpoint (SAC) detects errors in kinetochore-microtubule attachments. Timely activation and maintenance of the SAC until defects are corrected is essential for genome stability. Here, we show that shugoshin (Sgo1), a conserved tension-sensing protein, ensures the maintenance of SAC signals in response to unattached kinetochores during mitosis in a basidiomycete budding yeast Cryptococcus neoformans. Sgo1 maintains optimum levels of Aurora B kinase Ipl1 and protein phosphatase 1 (PP1) at kinetochores. The absence of Sgo1 results in the loss of Aurora BIpl1 with a concomitant increase in PP1 levels at kinetochores. This leads to a premature reduction in the kinetochore-bound Bub1 levels and early termination of the SAC signals. Intriguingly, the kinase function of Bub1 is dispensable for shugoshin's subcellular localization. Sgo1 is predominantly localized to spindle pole bodies (SPBs) and along the mitotic spindle with a minor pool at kinetochores. In the absence of proper kinetochore-microtubule attachments, Sgo1 reinforces the Aurora B kinaseIpl1-PP1 phosphatase balance, which is critical for prolonged maintenance of the SAC response.
Collapse
Affiliation(s)
- Satya Dev Polisetty
- Molecular Mycology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Krishna Bhat
- Molecular Mycology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Kuladeep Das
- Molecular Mycology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Ivan Clark
- Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Kevin G. Hardwick
- Institute of Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
| | - Kaustuv Sanyal
- Molecular Mycology Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
- Department of Biological Sciences, Bose Institute, Kolkata, India
| |
Collapse
|
5
|
Quaas AM, Penzias AS, Adashi EY. Embryonic aneuploidy - the true "last barrier in assisted reproductive technology"? F&S SCIENCE 2024; 5:303-305. [PMID: 39127422 DOI: 10.1016/j.xfss.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Human embryonic aneuploidy may represent one of the final frontiers in assisted reproductive technology, primarily secondary to oocyte aneuploidy. Mammalian oocytes possess unique characteristics predisposing them to much higher rates of aneuploidy than sperm or most somatic cells. Some of these characteristics are age-independent, whereas others result from reproductive aging and environmental toxicity. A detailed understanding of these properties may lead to novel diagnostic and therapeutic tools designed to detect and prevent oocyte and embryonic aneuploidy to overcome this ultimate barrier to success in assisted reproductive technology.
Collapse
Affiliation(s)
| | - Alan S Penzias
- Boston IVF, Waltham, Massachusetts; Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, Massachusetts
| | - Eli Y Adashi
- Department of Medical Science, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| |
Collapse
|
6
|
Joshi JN, Changela N, Mahal L, Jang J, Defosse T, Wang LI, Das A, Shapiro JG, McKim K. Meiosis-specific functions of kinetochore protein SPC105R required for chromosome segregation in Drosophila oocytes. Mol Biol Cell 2024; 35:ar105. [PMID: 38865189 PMCID: PMC11321039 DOI: 10.1091/mbc.e24-02-0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024] Open
Abstract
The reductional division of meiosis I requires the separation of chromosome pairs towards opposite poles. We have previously implicated the outer kinetochore protein SPC105R/KNL1 in driving meiosis I chromosome segregation through lateral attachments to microtubules and coorientation of sister centromeres. To identify the domains of SPC105R that are critical for meiotic chromosome segregation, an RNAi-resistant gene expression system was developed. We found that the SPC105R C-terminal domain (aa 1284-1960) is necessary and sufficient for recruiting NDC80 to the kinetochore and building the outer kinetochore. Furthermore, the C-terminal domain recruits BUBR1, which in turn recruits the cohesion protection proteins MEI-S332 and PP2A. Of the remaining 1283 amino acids, we found the first 473 are most important for meiosis. The first 123 amino acids of the N-terminal half of SPC105R contain the conserved SLRK and RISF motifs that are targets of PP1 and Aurora B kinase and are most important for regulating the stability of microtubule attachments and maintaining metaphase I arrest. The region between amino acids 124 and 473 are required for lateral microtubule attachments and biorientation of homologues, which are critical for accurate chromosome segregation in meiosis I.
Collapse
Affiliation(s)
- Jay N. Joshi
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854
| | - Neha Changela
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854
| | - Lia Mahal
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854
| | - Janet Jang
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854
| | - Tyler Defosse
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854
| | - Lin-Ing Wang
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854
| | - Arunika Das
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854
| | - Joanatta G. Shapiro
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854
| | - Kim McKim
- Waksman Institute and Department of Genetics, Rutgers, the State University of New Jersey, Piscataway, NJ 08854
| |
Collapse
|
7
|
Pan B, Bruno M, Macfarlan TS, Akera T. Meiosis-specific decoupling of the pericentromere from the kinetochore. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.21.604490. [PMID: 39091844 PMCID: PMC11291024 DOI: 10.1101/2024.07.21.604490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The primary constriction site of the M-phase chromosome is an established marker for the kinetochore position, often used to determine the karyotype of each species. Underlying this observation is the concept that the kinetochore is spatially linked with the pericentromere where sister-chromatids are most tightly cohered. Here, we found an unconventional pericentromere specification with sister chromatids mainly cohered at a chromosome end, spatially separated from the kinetochore in Peromyscus mouse oocytes. This distal locus enriched cohesin protectors, such as the Chromosomal Passenger Complex (CPC) and PP2A, at a higher level compared to its centromere/kinetochore region, acting as the primary site for sister-chromatid cohesion. Chromosomes with the distal cohesion site exhibited enhanced cohesin protection at anaphase I compared to those without it, implying that these distal cohesion sites may have evolved to ensure sister-chromatid cohesion during meiosis. In contrast, mitotic cells enriched CPC only near the kinetochore and the distal locus was not cohered between sister chromatids, suggesting a meiosis-specific mechanism to protect cohesin at this distal locus. We found that this distal locus corresponds to an additional centromeric satellite block, located far apart from the centromeric satellite block that builds the kinetochore. Several Peromyscus species carry chromosomes with two such centromeric satellite blocks. Analyses on three Peromyscus species revealed that the internal satellite consistently assembles the kinetochore in both mitosis and meiosis, whereas the distal satellite selectively enriches cohesin protectors in meiosis to promote sister-chromatid cohesion at that site. Thus, our study demonstrates that pericentromere specification is remarkably flexible and can control chromosome segregation in a cell-type and context dependent manner.
Collapse
Affiliation(s)
- Bo Pan
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health; Bethesda, Maryland 20894, USA
| | - Melania Bruno
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health; Bethesda, Maryland 20894, USA
| | - Todd S Macfarlan
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health; Bethesda, Maryland 20894, USA
| | - Takashi Akera
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health; Bethesda, Maryland 20894, USA
| |
Collapse
|
8
|
Ding Z, Peng L, Zeng J, Yuan K, Tang Y, Yi Q. Functions of HP1 in preventing chromosomal instability. Cell Biochem Funct 2024; 42:e4017. [PMID: 38603595 DOI: 10.1002/cbf.4017] [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: 12/14/2023] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Chromosomal instability (CIN), caused by errors in the segregation of chromosomes during mitosis, is a hallmark of many types of cancer. The fidelity of chromosome segregation is governed by a sophisticated cellular signaling network, one crucial orchestrator of which is Heterochromatin protein 1 (HP1). HP1 dynamically localizes to distinct sites at various stages of mitosis, where it regulates key mitotic events ranging from chromosome-microtubule attachment to sister chromatid cohesion to cytokinesis. Our evolving comprehension of HP1's multifaceted role has positioned it as a central protein in the orchestration of mitotic processes.
Collapse
Affiliation(s)
- Zexian Ding
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Lei Peng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Jinghua Zeng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Kejia Yuan
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Yan Tang
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Qi Yi
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
| |
Collapse
|
9
|
Joshi JN, Changela N, Mahal L, Defosse T, Jang J, Wang LI, Das A, Shapiro JG, McKim K. Meiosis-specific functions of kinetochore protein SPC105R required for chromosome segregation in Drosophila oocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.585003. [PMID: 38559067 PMCID: PMC10980020 DOI: 10.1101/2024.03.14.585003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The reductional division of meiosis I requires the separation of chromosome pairs towards opposite poles. We have previously implicated the outer kinetochore protein SPC105R/KNL1 in driving meiosis I chromosome segregation through lateral attachments to microtubules and co-orientation of sister centromeres. To identify the domains of SPC105R that are critical for meiotic chromosome segregation, an RNAi-resistant gene expression system was developed. We found that SPC105R's C-terminal domain (aa 1284-1960) is necessary and sufficient for recruiting NDC80 to the kinetochore and building the outer kinetochore. Furthermore, the C-terminal domain recruits BUBR1, which in turn recruits the cohesion protection proteins MEI-S332 and PP2A. Of the remaining 1283 amino acids, we found the first 473 are most important for meiosis. The first 123 amino acids of the N-terminal half of SPC105R contain the conserved SLRK and RISF motifs that are targets of PP1 and Aurora B kinase and are most important for regulating the stability of microtubule attachments and maintaining metaphase I arrest. The region between amino acids 124 and 473 are required for two activities that are critical for accurate chromosome segregation in meiosis I, lateral microtubule attachments and bi-orientation of homologs.
Collapse
|
10
|
Reed R, Park K, Waddell B, Timbers TA, Li C, Baxi K, Giacomin RM, Leroux MR, Carvalho CE. The Caenorhabditis elegans Shugoshin regulates TAC-1 in cilia. Sci Rep 2023; 13:9410. [PMID: 37296204 PMCID: PMC10256747 DOI: 10.1038/s41598-023-36430-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
The conserved Shugoshin (SGO) protein family is essential for mediating proper chromosome segregation from yeast to humans but has also been implicated in diverse roles outside of the nucleus. SGO's roles include inhibiting incorrect spindle attachment in the kinetochore, regulating the spindle assembly checkpoint (SAC), and ensuring centriole cohesion in the centrosome, all functions that involve different microtubule scaffolding structures in the cell. In Caenorhabditis elegans, a species with holocentric chromosomes, SGO-1 is not required for cohesin protection or spindle attachment but appears important for licensing meiotic recombination. Here we provide the first functional evidence that in C. elegans, Shugoshin functions in another extranuclear, microtubule-based structure, the primary cilium. We identify the centrosomal and microtubule-regulating transforming acidic coiled-coil protein, TACC/TAC-1, which also localizes to the basal body, as an SGO-1 binding protein. Genetic analyses indicate that TAC-1 activity must be maintained below a threshold at the ciliary base for correct cilia function, and that SGO-1 likely participates in constraining TAC-1 to the basal body by influencing the function of the transition zone 'ciliary gate'. This research expands our understanding of cellular functions of Shugoshin proteins and contributes to the growing examples of overlap between kinetochore, centrosome and cilia proteomes.
Collapse
Affiliation(s)
- R Reed
- Department of Biology, University of Saskatchewan, Saskatoon, Canada
| | - K Park
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
- Terry Fox Laboratory, BC Cancer, Vancouver, BC, V5Z 1L3, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - B Waddell
- Department of Biology, University of Saskatchewan, Saskatoon, Canada
| | - T A Timbers
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - C Li
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - K Baxi
- Department of Biology, University of Saskatchewan, Saskatoon, Canada
| | - R M Giacomin
- Department of Biology, University of Saskatchewan, Saskatoon, Canada
| | - M R Leroux
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| | - C E Carvalho
- Department of Biology, University of Saskatchewan, Saskatoon, Canada.
| |
Collapse
|
11
|
Konecna M, Abbasi Sani S, Anger M. Separase and Roads to Disengage Sister Chromatids during Anaphase. Int J Mol Sci 2023; 24:ijms24054604. [PMID: 36902034 PMCID: PMC10003635 DOI: 10.3390/ijms24054604] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Receiving complete and undamaged genetic information is vital for the survival of daughter cells after chromosome segregation. The most critical steps in this process are accurate DNA replication during S phase and a faithful chromosome segregation during anaphase. Any errors in DNA replication or chromosome segregation have dire consequences, since cells arising after division might have either changed or incomplete genetic information. Accurate chromosome segregation during anaphase requires a protein complex called cohesin, which holds together sister chromatids. This complex unifies sister chromatids from their synthesis during S phase, until separation in anaphase. Upon entry into mitosis, the spindle apparatus is assembled, which eventually engages kinetochores of all chromosomes. Additionally, when kinetochores of sister chromatids assume amphitelic attachment to the spindle microtubules, cells are finally ready for the separation of sister chromatids. This is achieved by the enzymatic cleavage of cohesin subunits Scc1 or Rec8 by an enzyme called Separase. After cohesin cleavage, sister chromatids remain attached to the spindle apparatus and their poleward movement on the spindle is initiated. The removal of cohesion between sister chromatids is an irreversible step and therefore it must be synchronized with assembly of the spindle apparatus, since precocious separation of sister chromatids might lead into aneuploidy and tumorigenesis. In this review, we focus on recent discoveries concerning the regulation of Separase activity during the cell cycle.
Collapse
Affiliation(s)
- Marketa Konecna
- Department of Genetics and Reproduction, Veterinary Research Institute, 621 00 Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Science, 277 21 Libechov, Czech Republic
- Faculty of Science, Masaryk University, 602 00 Brno, Czech Republic
| | - Soodabeh Abbasi Sani
- Department of Genetics and Reproduction, Veterinary Research Institute, 621 00 Brno, Czech Republic
- Faculty of Science, Masaryk University, 602 00 Brno, Czech Republic
| | - Martin Anger
- Department of Genetics and Reproduction, Veterinary Research Institute, 621 00 Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Science, 277 21 Libechov, Czech Republic
- Correspondence:
| |
Collapse
|
12
|
Charalambous C, Webster A, Schuh M. Aneuploidy in mammalian oocytes and the impact of maternal ageing. Nat Rev Mol Cell Biol 2023; 24:27-44. [PMID: 36068367 DOI: 10.1038/s41580-022-00517-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2022] [Indexed: 11/09/2022]
Abstract
During fertilization, the egg and the sperm are supposed to contribute precisely one copy of each chromosome to the embryo. However, human eggs frequently contain an incorrect number of chromosomes - a condition termed aneuploidy, which is much more prevalent in eggs than in either sperm or in most somatic cells. In turn, aneuploidy in eggs is a leading cause of infertility, miscarriage and congenital syndromes. Aneuploidy arises as a consequence of aberrant meiosis during egg development from its progenitor cell, the oocyte. In human oocytes, chromosomes often segregate incorrectly. Chromosome segregation errors increase in women from their mid-thirties, leading to even higher levels of aneuploidy in eggs from women of advanced maternal age, ultimately causing age-related infertility. Here, we cover the two main areas that contribute to aneuploidy: (1) factors that influence the fidelity of chromosome segregation in eggs of women from all ages and (2) factors that change in response to reproductive ageing. Recent discoveries reveal new error-causing pathways and present a framework for therapeutic strategies to extend the span of female fertility.
Collapse
Affiliation(s)
- Chloe Charalambous
- Department of Meiosis, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Alexandre Webster
- Department of Meiosis, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Melina Schuh
- Department of Meiosis, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
13
|
Zhu Z, Xu W, Liu L. Ovarian aging: mechanisms and intervention strategies. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:590-610. [PMID: 37724254 PMCID: PMC10471094 DOI: 10.1515/mr-2022-0031] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/25/2022] [Indexed: 09/20/2023]
Abstract
Ovarian reserve is essential for fertility and influences healthy aging in women. Advanced maternal age correlates with the progressive loss of both the quantity and quality of oocytes. The molecular mechanisms and various contributing factors underlying ovarian aging have been uncovered. In this review, we highlight some of critical factors that impact oocyte quantity and quality during aging. Germ cell and follicle reserve at birth determines reproductive lifespan and timing the menopause in female mammals. Accelerated diminishing ovarian reserve leads to premature ovarian aging or insufficiency. Poor oocyte quality with increasing age could result from chromosomal cohesion deterioration and misaligned chromosomes, telomere shortening, DNA damage and associated genetic mutations, oxidative stress, mitochondrial dysfunction and epigenetic alteration. We also discuss the intervention strategies to delay ovarian aging. Both the efficacy of senotherapies by antioxidants against reproductive aging and mitochondrial therapy are discussed. Functional oocytes and ovarioids could be rejuvenated from pluripotent stem cells or somatic cells. We propose directions for future interventions. As couples increasingly begin delaying parenthood in life worldwide, understanding the molecular mechanisms during female reproductive aging and potential intervention strategies could benefit women in making earlier choices about their reproductive health.
Collapse
Affiliation(s)
- Zhengmao Zhu
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, China
| | - Wanxue Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Lin Liu
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Tianjin Union Medical Center, Institute of Translational Medicine, Nankai University, Tianjin, China
| |
Collapse
|
14
|
Urban JA, Ranjan R, Chen X. Asymmetric Histone Inheritance: Establishment, Recognition, and Execution. Annu Rev Genet 2022; 56:113-143. [PMID: 35905975 PMCID: PMC10054593 DOI: 10.1146/annurev-genet-072920-125226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The discovery of biased histone inheritance in asymmetrically dividing Drosophila melanogaster male germline stem cells demonstrates one means to produce two distinct daughter cells with identical genetic material. This inspired further studies in different systems, which revealed that this phenomenon may be a widespread mechanism to introduce cellular diversity. While the extent of asymmetric histone inheritance could vary among systems, this phenomenon is proposed to occur in three steps: first, establishment of histone asymmetry between sister chromatids during DNA replication; second, recognition of sister chromatids carrying asymmetric histone information during mitosis; and third, execution of this asymmetry in the resulting daughter cells. By compiling the current knowledge from diverse eukaryotic systems, this review comprehensively details and compares known chromatin factors, mitotic machinery components, and cell cycle regulators that may contribute to each of these three steps. Also discussed are potential mechanisms that introduce and regulate variable histone inheritance modes and how these different modes may contribute to cell fate decisions in multicellular organisms.
Collapse
Affiliation(s)
- Jennifer A Urban
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland, USA;
| | - Rajesh Ranjan
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland, USA; .,Howard Hughes Medical Institute, The Johns Hopkins University, Baltimore, Maryland, USA; ,
| | - Xin Chen
- Department of Biology, The Johns Hopkins University, Baltimore, Maryland, USA; .,Howard Hughes Medical Institute, The Johns Hopkins University, Baltimore, Maryland, USA; ,
| |
Collapse
|
15
|
Hu Q, Liu Q, Zhao Y, Zhang L, Li L. SGOL2 is a novel prognostic marker and fosters disease progression via a MAD2-mediated pathway in hepatocellular carcinoma. Biomark Res 2022; 10:82. [PMCID: PMC9664666 DOI: 10.1186/s40364-022-00422-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Background Shugoshin-like protein 2 (SGOL2) is a centromeric protein that ensures the correct and orderly process of mitosis by protecting and maintaining centripetal adhesions during meiosis and mitosis. Here, we examined the potential role of SGOL2 in cancers, especially in hepatocellular carcinoma (HCC). Methods One hundred ninety-nine normal adjacent tissues and 202 HCC samples were collected in this study. Human HCC cells (SK-HEP-1 and HEP-3B) were employed in the present study. Immunohistochemistry, immunofluorescence, western blot, Co-Immunoprecipitation technique, and bioinformatic analysis were utilized to assess the role of SGOL2 in HCC development process. Results Overexpression of SGOL2 predicted an unfavorable prognosis in HCC by The Cancer Genome Atlas database (TCGA), which were further validated in our two independent cohorts. Next, 47 differentially expressed genes positively related to both SGOL2 and MAD2 were identified to be associated with the cell cycle. Subsequently, we demonstrated that SGOL2 downregulation suppressed the malignant activities of HCC in vitro and in vivo. Further investigation showed that SGOL2 promoted tumor proliferation by regulating MAD2-induced cell-cycle dysregulation, which could be reversed by the MAD2 inhibitor M2I-1. Consistently, MAD2 upregulation reversed the knockdown effects of SGOL2-shRNA in HCC. Moreover, we demonstrated that SGOL2 regulated MAD2 expression level by forming a SGOL2-MAD2 complex, which led to cell cycle dysreuglation of HCC cells. Conclusion SGOL2 acts as an oncogene in HCC cells by regulating MAD2 and then dysregulating the cell cycle, providing a potential therapeutic target in HCC. Supplementary Information The online version contains supplementary material available at 10.1186/s40364-022-00422-z.
Collapse
Affiliation(s)
- Qingqing Hu
- grid.13402.340000 0004 1759 700XState Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Qiuhong Liu
- grid.13402.340000 0004 1759 700XState Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Yalei Zhao
- grid.13402.340000 0004 1759 700XState Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Lingjian Zhang
- grid.13402.340000 0004 1759 700XState Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Lanjuan Li
- grid.13402.340000 0004 1759 700XState Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003 China
| |
Collapse
|
16
|
Kumon T, Lampson MA. Evolution of eukaryotic centromeres by drive and suppression of selfish genetic elements. Semin Cell Dev Biol 2022; 128:51-60. [PMID: 35346579 PMCID: PMC9232976 DOI: 10.1016/j.semcdb.2022.03.026] [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: 01/11/2022] [Revised: 03/20/2022] [Accepted: 03/20/2022] [Indexed: 10/18/2022]
Abstract
Despite the universal requirement for faithful chromosome segregation, eukaryotic centromeres are rapidly evolving. It is hypothesized that rapid centromere evolution represents an evolutionary arms race between selfish genetic elements that drive, or propagate at the expense of organismal fitness, and mechanisms that suppress fitness costs. Selfish centromere DNA achieves preferential inheritance in female meiosis by recruiting more effector proteins that alter spindle microtubule interaction dynamics. Parallel pathways for effector recruitment are adaptively evolved to suppress functional differences between centromeres. Opportunities to drive are not limited to female meiosis, and selfish transposons, plasmids and B chromosomes also benefit by maximizing their inheritance. Rapid evolution of selfish genetic elements can diversify suppressor mechanisms in different species that may cause hybrid incompatibility.
Collapse
Affiliation(s)
- Tomohiro Kumon
- Howard Hughes Medical Institute, Whitehead Institute for Biomedical Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Michael A Lampson
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
17
|
Dudka D, Lampson MA. Centromere drive: model systems and experimental progress. Chromosome Res 2022; 30:187-203. [PMID: 35731424 DOI: 10.1007/s10577-022-09696-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 11/28/2022]
Abstract
Centromeres connect chromosomes and spindle microtubules to ensure faithful chromosome segregation. Paradoxically, despite this conserved function, centromeric DNA evolves rapidly and centromeric proteins show signatures of positive selection. The centromere drive hypothesis proposes that centromeric DNA can act like a selfish genetic element and drive non-Mendelian segregation during asymmetric female meiosis. Resulting fitness costs lead to genetic conflict with the rest of the genome and impose a selective pressure for centromeric proteins to adapt by suppressing the costs. Here, we describe experimental model systems for centromere drive in yellow monkeyflowers and mice, summarize key findings demonstrating centromere drive, and explain molecular mechanisms. We further discuss efforts to test if centromeric proteins are involved in suppressing drive-associated fitness costs, highlight a model for centromere drive and suppression in mice, and put forth outstanding questions for future research.
Collapse
Affiliation(s)
- Damian Dudka
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael A Lampson
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
18
|
Osadska M, Selicky T, Kretova M, Jurcik J, Sivakova B, Cipakova I, Cipak L. The Interplay of Cohesin and RNA Processing Factors: The Impact of Their Alterations on Genome Stability. Int J Mol Sci 2022; 23:3939. [PMID: 35409298 PMCID: PMC8999970 DOI: 10.3390/ijms23073939] [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: 03/03/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 12/01/2022] Open
Abstract
Cohesin, a multi-subunit protein complex, plays important roles in sister chromatid cohesion, DNA replication, chromatin organization, gene expression, transcription regulation, and the recombination or repair of DNA damage. Recently, several studies suggested that the functions of cohesin rely not only on cohesin-related protein-protein interactions, their post-translational modifications or specific DNA modifications, but that some RNA processing factors also play an important role in the regulation of cohesin functions. Therefore, the mutations and changes in the expression of cohesin subunits or alterations in the interactions between cohesin and RNA processing factors have been shown to have an impact on cohesion, the fidelity of chromosome segregation and, ultimately, on genome stability. In this review, we provide an overview of the cohesin complex and its role in chromosome segregation, highlight the causes and consequences of mutations and changes in the expression of cohesin subunits, and discuss the RNA processing factors that participate in the regulation of the processes involved in chromosome segregation. Overall, an understanding of the molecular determinants of the interplay between cohesin and RNA processing factors might help us to better understand the molecular mechanisms ensuring the integrity of the genome.
Collapse
Affiliation(s)
- Michaela Osadska
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia; (M.O.); (T.S.); (M.K.); (J.J.)
| | - Tomas Selicky
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia; (M.O.); (T.S.); (M.K.); (J.J.)
| | - Miroslava Kretova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia; (M.O.); (T.S.); (M.K.); (J.J.)
| | - Jan Jurcik
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia; (M.O.); (T.S.); (M.K.); (J.J.)
| | - Barbara Sivakova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska Cesta 9, 845 38 Bratislava, Slovakia;
| | - Ingrid Cipakova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia; (M.O.); (T.S.); (M.K.); (J.J.)
| | - Lubos Cipak
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia; (M.O.); (T.S.); (M.K.); (J.J.)
| |
Collapse
|
19
|
Loss, Gain, and Retention: Mechanisms Driving Late Prophase I Chromosome Remodeling for Accurate Meiotic Chromosome Segregation. Genes (Basel) 2022; 13:genes13030546. [PMID: 35328099 PMCID: PMC8949218 DOI: 10.3390/genes13030546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
To generate gametes, sexually reproducing organisms need to achieve a reduction in ploidy, via meiosis. Several mechanisms are set in place to ensure proper reductional chromosome segregation at the first meiotic division (MI), including chromosome remodeling during late prophase I. Chromosome remodeling after crossover formation involves changes in chromosome condensation and restructuring, resulting in a compact bivalent, with sister kinetochores oriented to opposite poles, whose structure is crucial for localized loss of cohesion and accurate chromosome segregation. Here, we review the general processes involved in late prophase I chromosome remodeling, their regulation, and the strategies devised by different organisms to produce bivalents with configurations that promote accurate segregation.
Collapse
|
20
|
Roles and regulation of Haspin kinase and its impact on carcinogenesis. Cell Signal 2022; 93:110303. [DOI: 10.1016/j.cellsig.2022.110303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 01/15/2023]
|
21
|
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
|
22
|
Kumon T, Ma J, Akins RB, Stefanik D, Nordgren CE, Kim J, Levine MT, Lampson MA. Parallel pathways for recruiting effector proteins determine centromere drive and suppression. Cell 2021; 184:4904-4918.e11. [PMID: 34433012 PMCID: PMC8448984 DOI: 10.1016/j.cell.2021.07.037] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 06/07/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
Selfish centromere DNA sequences bias their transmission to the egg in female meiosis. Evolutionary theory suggests that centromere proteins evolve to suppress costs of this "centromere drive." In hybrid mouse models with genetically different maternal and paternal centromeres, selfish centromere DNA exploits a kinetochore pathway to recruit microtubule-destabilizing proteins that act as drive effectors. We show that such functional differences are suppressed by a parallel pathway for effector recruitment by heterochromatin, which is similar between centromeres in this system. Disrupting the kinetochore pathway with a divergent allele of CENP-C reduces functional differences between centromeres, whereas disrupting heterochromatin by CENP-B deletion amplifies the differences. Molecular evolution analyses using Murinae genomes identify adaptive evolution in proteins in both pathways. We propose that centromere proteins have recurrently evolved to minimize the kinetochore pathway, which is exploited by selfish DNA, relative to the heterochromatin pathway that equalizes centromeres, while maintaining essential functions.
Collapse
Affiliation(s)
- Tomohiro Kumon
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jun Ma
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - R Brian Akins
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Derek Stefanik
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - C Erik Nordgren
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junhyong Kim
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mia T Levine
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael A Lampson
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
23
|
Abstract
Female meiotic drive is the phenomenon where a selfish genetic element alters chromosome segregation during female meiosis to segregate to the egg and transmit to the next generation more frequently than Mendelian expectation. While several examples of female meiotic drive have been known for many decades, a molecular understanding of the underlying mechanisms has been elusive. Recent advances in this area in several model species prompts a comparative re-examination of these drive systems. In this review, we compare female meiotic drive of several animal and plant species, highlighting pertinent similarities.
Collapse
Affiliation(s)
- Frances E. Clark
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, 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
|
24
|
Sane A, Sridhar S, Sanyal K, Ghosh SK. Shugoshin ensures maintenance of the spindle assembly checkpoint response and efficient spindle disassembly. Mol Microbiol 2021; 116:1079-1098. [PMID: 34407255 DOI: 10.1111/mmi.14796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/07/2021] [Accepted: 08/15/2021] [Indexed: 11/27/2022]
Abstract
Shugoshin proteins are evolutionarily conserved across eukaryotes, with some species-specific cellular functions, ensuring the fidelity of chromosome segregation. They act as adaptors at various subcellular locales to mediate several protein-protein interactions in a spatio-temporal manner. Here, we characterize shugoshin (Sgo1) in the human fungal pathogen Candida albicans. We observe that Sgo1 retains its centromeric localization and performs its conserved functions of regulating the sister chromatid biorientation, centromeric condensin localization, and maintenance of chromosomal passenger complex (CPC). We identify novel roles of Sgo1 as a spindle assembly checkpoint (SAC) component with functions in maintaining a prolonged SAC response by retaining Mad2 and Bub1 at the kinetochores in response to improper kinetochore-microtubule attachments. Strikingly, we discover the in vivo localization of Sgo1 along the length of the mitotic spindle. Our results indicate that Sgo1 performs a hitherto unknown function of facilitating timely disassembly of the mitotic spindle in C. albicans. To summarize, this study unravels a unique functional adaptation of shugoshin in maintaining genomic stability.
Collapse
Affiliation(s)
- Aakanksha Sane
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai, India
| | - Shreyas Sridhar
- Molecular Biology & Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India.,Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Kaustuv Sanyal
- Molecular Biology & Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India.,Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Santanu K Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai, India
| |
Collapse
|
25
|
Maier NK, Ma J, Lampson MA, Cheeseman IM. Separase cleaves the kinetochore protein Meikin at the meiosis I/II transition. Dev Cell 2021; 56:2192-2206.e8. [PMID: 34331869 PMCID: PMC8355204 DOI: 10.1016/j.devcel.2021.06.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 05/03/2021] [Accepted: 06/25/2021] [Indexed: 12/19/2022]
Abstract
To generate haploid gametes, germ cells undergo two consecutive meiotic divisions requiring key changes to the cell division machinery. Here, we demonstrate that the protease separase rewires key cell division processes at the meiosis I/II transition by cleaving the meiosis-specific protein Meikin. Separase proteolysis does not inactivate Meikin but instead alters its function to create a distinct activity state. Full-length Meikin and the C-terminal Meikin separase cleavage product both localize to kinetochores, bind to Plk1 kinase, and promote Rec8 cleavage, but our results reveal distinct roles for these proteins in controlling meiosis. Mutations that prevent Meikin cleavage or that conditionally inactivate Meikin at anaphase I result in defective meiosis II chromosome alignment in mouse oocytes. Finally, as oocytes exit meiosis, C-Meikin is eliminated by APC/C-mediated degradation prior to the first mitotic division. Thus, multiple regulatory events irreversibly modulate Meikin activity during successive meiotic divisions to rewire the cell division machinery at two distinct transitions.
Collapse
Affiliation(s)
- Nolan K Maier
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jun Ma
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael A Lampson
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
26
|
Ueki Y, Hadders MA, Weisser MB, Nasa I, Sotelo‐Parrilla P, Cressey LE, Gupta T, Hertz EPT, Kruse T, Montoya G, Jeyaprakash AA, Kettenbach A, Lens SMA, Nilsson J. A highly conserved pocket on PP2A-B56 is required for hSgo1 binding and cohesion protection during mitosis. EMBO Rep 2021; 22:e52295. [PMID: 33973335 PMCID: PMC8256288 DOI: 10.15252/embr.202052295] [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: 12/17/2020] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 01/11/2023] Open
Abstract
The shugoshin proteins are universal protectors of centromeric cohesin during mitosis and meiosis. The binding of human hSgo1 to the PP2A-B56 phosphatase through a coiled-coil (CC) region mediates cohesion protection during mitosis. Here we undertook a structure function analysis of the PP2A-B56-hSgo1 complex, revealing unanticipated aspects of complex formation and function. We establish that a highly conserved pocket on the B56 regulatory subunit is required for hSgo1 binding and cohesion protection during mitosis in human somatic cells. Consistent with this, we show that hSgo1 blocks the binding of PP2A-B56 substrates containing a canonical B56 binding motif. We find that PP2A-B56 bound to hSgo1 dephosphorylates Cdk1 sites on hSgo1 itself to modulate cohesin interactions. Collectively our work provides important insight into cohesion protection during mitosis.
Collapse
Affiliation(s)
- Yumi Ueki
- The Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical ScienceUniversity of CopenhagenCopenhagenDenmark
| | - Michael A Hadders
- Oncode Institute and Center for Molecular MedicineUniversity Medical Center UtrechUtrecht UniversityUtrechtThe Netherlands
| | - Melanie B Weisser
- The Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical ScienceUniversity of CopenhagenCopenhagenDenmark
| | - Isha Nasa
- Biochemistry and Cell BiologyGeisel School of Medicine at Dartmouth CollegeHanoverNHUSA
| | | | - Lauren E Cressey
- Biochemistry and Cell BiologyGeisel School of Medicine at Dartmouth CollegeHanoverNHUSA
| | - Tanmay Gupta
- Wellcome Center for Cell BiologyUniversity of EdinburghEdinburghUK
| | - Emil P T Hertz
- The Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical ScienceUniversity of CopenhagenCopenhagenDenmark
| | - Thomas Kruse
- The Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical ScienceUniversity of CopenhagenCopenhagenDenmark
| | - Guillermo Montoya
- The Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical ScienceUniversity of CopenhagenCopenhagenDenmark
| | | | - Arminja Kettenbach
- Biochemistry and Cell BiologyGeisel School of Medicine at Dartmouth CollegeHanoverNHUSA
| | - Susanne M A Lens
- Oncode Institute and Center for Molecular MedicineUniversity Medical Center UtrechUtrecht UniversityUtrechtThe Netherlands
| | - Jakob Nilsson
- The Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical ScienceUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
27
|
Wartosch L, Schindler K, Schuh M, Gruhn JR, Hoffmann ER, McCoy RC, Xing J. Origins and mechanisms leading to aneuploidy in human eggs. Prenat Diagn 2021; 41:620-630. [PMID: 33860956 PMCID: PMC8237340 DOI: 10.1002/pd.5927] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/02/2021] [Accepted: 02/21/2021] [Indexed: 11/18/2022]
Abstract
The gain or loss of a chromosome-or aneuploidy-acts as one of the major triggers for infertility and pregnancy loss in humans. These chromosomal abnormalities affect more than 40% of eggs in women at both ends of the age spectrum, that is, young girls as well as women of advancing maternal age. Recent studies in human oocytes and embryos using genomics, cytogenetics, and in silico modeling all provide new insight into the rates and potential genetic and cellular factors associated with aneuploidy at varying stages of development. Here, we review recent studies that are shedding light on potential molecular mechanisms of chromosome missegregation in oocytes and embryos across the entire female reproductive life span.
Collapse
Affiliation(s)
- Lena Wartosch
- Department of MeiosisMax Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Karen Schindler
- Department of GeneticsRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
- Human Genetics Institute of New JerseyRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
| | - Melina Schuh
- Department of MeiosisMax Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Jennifer R. Gruhn
- DNRF Center for Chromosome StabilityDepartment of Cellular and Molecular MedicineFaculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Eva R. Hoffmann
- DNRF Center for Chromosome StabilityDepartment of Cellular and Molecular MedicineFaculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Rajiv C. McCoy
- Department of BiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jinchuan Xing
- Department of GeneticsRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
- Human Genetics Institute of New JerseyRutgers, The State University of New JerseyPiscatawayNew JerseyUSA
| |
Collapse
|
28
|
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
|
29
|
Specificity determinants of phosphoprotein phosphatases controlling kinetochore functions. Essays Biochem 2021; 64:325-336. [PMID: 32501472 DOI: 10.1042/ebc20190065] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022]
Abstract
Kinetochores are instrumental for accurate chromosome segregation by binding to microtubules in order to move chromosomes and by delaying anaphase onset through the spindle assembly checkpoint (SAC). Dynamic phosphorylation of kinetochore components is key to control these activities and is tightly regulated by temporal and spatial recruitment of kinases and phosphoprotein phosphatases (PPPs). Here we focus on PP1, PP2A-B56 and PP2A-B55, three PPPs that are important regulators of mitosis. Despite the fact that these PPPs share a very similar active site, they target unique ser/thr phosphorylation sites to control kinetochore function. Specificity is in part achieved by PPPs binding to short linear motifs (SLiMs) that guide their substrate specificity. SLiMs bind to conserved pockets on PPPs and are degenerate in nature, giving rise to a range of binding affinities. These SLiMs control the assembly of numerous substrate specifying complexes and their position and binding strength allow PPPs to target specific phosphorylation sites. In addition, the activity of PPPs is regulated by mitotic kinases and inhibitors, either directly at the activity level or through affecting PPP-SLiM interactions. Here, we discuss recent progress in understanding the regulation of PPP specificity and activity and how this controls kinetochore biology.
Collapse
|
30
|
Cairo G, MacKenzie AM, Lacefield S. Differential requirement for Bub1 and Bub3 in regulation of meiotic versus mitotic chromosome segregation. J Cell Biol 2020; 219:133770. [PMID: 32328625 PMCID: PMC7147105 DOI: 10.1083/jcb.201909136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 01/02/2020] [Accepted: 01/15/2020] [Indexed: 01/21/2023] Open
Abstract
Accurate chromosome segregation depends on the proper attachment of kinetochores to spindle microtubules before anaphase onset. The Ipl1/Aurora B kinase corrects improper attachments by phosphorylating kinetochore components and so releasing aberrant kinetochore–microtubule interactions. The localization of Ipl1 to kinetochores in budding yeast depends upon multiple pathways, including the Bub1–Bub3 pathway. We show here that in meiosis, Bub3 is crucial for correction of attachment errors. Depletion of Bub3 results in reduced levels of kinetochore-localized Ipl1 and concomitant massive chromosome missegregation caused by incorrect chromosome–spindle attachments. Depletion of Bub3 also results in shorter metaphase I and metaphase II due to premature localization of protein phosphatase 1 (PP1) to kinetochores, which antagonizes Ipl1-mediated phosphorylation. We propose a new role for the Bub1–Bub3 pathway in maintaining the balance between kinetochore localization of Ipl1 and PP1, a balance that is essential for accurate meiotic chromosome segregation and timely anaphase onset.
Collapse
Affiliation(s)
- Gisela Cairo
- Department of Biology, Indiana University, Bloomington, IN
| | | | - Soni Lacefield
- Department of Biology, Indiana University, Bloomington, IN
| |
Collapse
|
31
|
Yahya G, Wu Y, Peplowska K, Röhrl J, Soh YM, Bürmann F, Gruber S, Storchova Z. Phospho-regulation of the Shugoshin - Condensin interaction at the centromere in budding yeast. PLoS Genet 2020; 16:e1008569. [PMID: 32810145 PMCID: PMC7454948 DOI: 10.1371/journal.pgen.1008569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 08/28/2020] [Accepted: 05/22/2020] [Indexed: 01/07/2023] Open
Abstract
Correct bioriented attachment of sister chromatids to the mitotic spindle is essential for chromosome segregation. In budding yeast, the conserved protein shugoshin (Sgo1) contributes to biorientation by recruiting the protein phosphatase PP2A-Rts1 and the condensin complex to centromeres. Using peptide prints, we identified a Serine-Rich Motif (SRM) of Sgo1 that mediates the interaction with condensin and is essential for centromeric condensin recruitment and the establishment of biorientation. We show that the interaction is regulated via phosphorylation within the SRM and we determined the phospho-sites using mass spectrometry. Analysis of the phosphomimic and phosphoresistant mutants revealed that SRM phosphorylation disrupts the shugoshin–condensin interaction. We present evidence that Mps1, a central kinase in the spindle assembly checkpoint, directly phosphorylates Sgo1 within the SRM to regulate the interaction with condensin and thereby condensin localization to centromeres. Our findings identify novel mechanisms that control shugoshin activity at the centromere in budding yeast. Proper chromosome segregation in eukaryotes is ensured through correct attachment of the spindle microtubules to the centromeric chromosomal regions. The attachment is mediated via the multimolecular proteinaceous complex called the kinetochore. This enables the establishment of bioirentation, when each sister chromatid is attached to microtubules emanating from opposite spindle poles. Shugoshin (Sgo1) is a conserved centromeric protein that facilitates biorientation through its interactions with the protein phosphatase PP2A-Rts1, chromosome passenger complex and centromeric condensin. Here, we identified a serine-rich motif that is required for the interaction of shugoshin with the condensin complex. We show that loss of this region impairs condensin enrichment at the centromere, chromosome biorientation, segregation as well as the function of the chromosome passenger complex in the error correction. Moreover, the interaction is phosphoregulated, as phosphorylation of the serine-rich motif on Sgo1 disrupts its interaction with condensin. Finally, we show that the conserved spindle assembly checkpoint kinase Mps1 is responsible for this phosphorylation. Our findings uncover novel regulatory mechanisms that facilitate proper chromosome segregation.
Collapse
Affiliation(s)
- Galal Yahya
- Department of Microbiology and Immunology, School of Pharmacy, Zagazig University, Egypt
- Department of Molecular Genetics, TU Kaiserlautern, Kaiserslautern, Germany
| | - Yehui Wu
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Karolina Peplowska
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Genomics and Bioinformatics Shared Resource, University of Hawaii Cancer Center, Honolulu, United States of America
| | - Jennifer Röhrl
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Young-Min Soh
- Department of Fundamental Microbiology, UNIL-Sorge District, Lausanne, Switzerland
| | - Frank Bürmann
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, United Kingdom
| | - Stephan Gruber
- Department of Fundamental Microbiology, UNIL-Sorge District, Lausanne, Switzerland
| | - Zuzana Storchova
- Department of Molecular Genetics, TU Kaiserlautern, Kaiserslautern, Germany
- * E-mail:
| |
Collapse
|
32
|
Jiménez J, Queralt E, Posas F, de Nadal E. The regulation of Net1/Cdc14 by the Hog1 MAPK upon osmostress unravels a new mechanism regulating mitosis. Cell Cycle 2020; 19:2105-2118. [PMID: 32794416 PMCID: PMC7513861 DOI: 10.1080/15384101.2020.1804222] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
During evolution, cells have developed a plethora of mechanisms to optimize survival in a changing and unpredictable environment. In this regard, they have evolved networks that include environmental sensors, signaling transduction molecules and response mechanisms. Hog1 (yeast) and p38 (mammals) stress-activated protein kinases (SAPKs) are activated upon stress and they drive a full collection of cell adaptive responses aimed to maximize survival. SAPKs are extensively used to learn about the mechanisms through which cells adapt to changing environments. In addition to regulating gene expression and metabolism, SAPKs control cell cycle progression. In this review, we will discuss the latest findings related to the SAPK-driven regulation of mitosis upon osmostress in yeast.
Collapse
Affiliation(s)
- Javier Jiménez
- Departament De Ciències Experimentals I De La Salut, Universitat Pompeu Fabra (UPF) , Barcelona, Spain.,Department of Ciències Bàsiques, Facultat De Medicina I Ciències De La Salut, Universitat Internacional De Catalunya , Barcelona, Spain
| | - Ethel Queralt
- Cell Cycle Group, Institut d'Investigacions Biomèdica De Bellvitge (IDIBELL), L'Hospitalet De Llobregat , Barcelona, Spain
| | - Francesc Posas
- Departament De Ciències Experimentals I De La Salut, Universitat Pompeu Fabra (UPF) , Barcelona, Spain.,Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute of Science and Technology , 08028 Barcelona, Spain
| | - Eulàlia de Nadal
- Departament De Ciències Experimentals I De La Salut, Universitat Pompeu Fabra (UPF) , Barcelona, Spain.,Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute of Science and Technology , 08028 Barcelona, Spain
| |
Collapse
|
33
|
CDK Regulation of Meiosis: Lessons from S. cerevisiae and S. pombe. Genes (Basel) 2020; 11:genes11070723. [PMID: 32610611 PMCID: PMC7397238 DOI: 10.3390/genes11070723] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 12/13/2022] Open
Abstract
Meiotic progression requires precise orchestration, such that one round of DNA replication is followed by two meiotic divisions. The order and timing of meiotic events is controlled through the modulation of the phosphorylation state of proteins. Key components of this phospho-regulatory system include cyclin-dependent kinase (CDK) and its cyclin regulatory subunits. Over the past two decades, studies in budding and fission yeast have greatly informed our understanding of the role of CDK in meiotic regulation. In this review, we provide an overview of how CDK controls meiotic events in both budding and fission yeast. We discuss mechanisms of CDK regulation through post-translational modifications and changes in the levels of cyclins. Finally, we highlight the similarities and differences in CDK regulation between the two yeast species. Since CDK and many meiotic regulators are highly conserved, the findings in budding and fission yeasts have revealed conserved mechanisms of meiotic regulation among eukaryotes.
Collapse
|
34
|
Matellán L, Monje-Casas F. Regulation of Mitotic Exit by Cell Cycle Checkpoints: Lessons From Saccharomyces cerevisiae. Genes (Basel) 2020; 11:E195. [PMID: 32059558 PMCID: PMC7074328 DOI: 10.3390/genes11020195] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
In order to preserve genome integrity and their ploidy, cells must ensure that the duplicated genome has been faithfully replicated and evenly distributed before they complete their division by mitosis. To this end, cells have developed highly elaborated checkpoints that halt mitotic progression when problems in DNA integrity or chromosome segregation arise, providing them with time to fix these issues before advancing further into the cell cycle. Remarkably, exit from mitosis constitutes a key cell cycle transition that is targeted by the main mitotic checkpoints, despite these surveillance mechanisms being activated by specific intracellular signals and acting at different stages of cell division. Focusing primarily on research carried out using Saccharomyces cerevisiae as a model organism, the aim of this review is to provide a general overview of the molecular mechanisms by which the major cell cycle checkpoints control mitotic exit and to highlight the importance of the proper regulation of this process for the maintenance of genome stability during the distribution of the duplicated chromosomes between the dividing cells.
Collapse
Affiliation(s)
| | - Fernando Monje-Casas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Spanish National Research Council (CSIC)—University of Seville—University Pablo de Olavide, Avda, Américo Vespucio, 24, 41092 Sevilla, Spain;
| |
Collapse
|
35
|
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
|
36
|
Kim S, Kim NH, Park JE, Hwang JW, Myung N, Hwang KT, Kim YA, Jang CY, Kim YK. PRMT6-mediated H3R2me2a guides Aurora B to chromosome arms for proper chromosome segregation. Nat Commun 2020; 11:612. [PMID: 32001712 PMCID: PMC6992762 DOI: 10.1038/s41467-020-14511-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 01/10/2020] [Indexed: 11/09/2022] Open
Abstract
The kinase Aurora B forms the chromosomal passenger complex (CPC) together with Borealin, INCENP, and Survivin to mediate chromosome condensation, the correction of erroneous spindle-kinetochore attachments, and cytokinesis. Phosphorylation of histone H3 Thr3 by Haspin kinase and of histone H2A Thr120 by Bub1 concentrates the CPC at the centromere. However, how the CPC is recruited to chromosome arms upon mitotic entry is unknown. Here, we show that asymmetric dimethylation at Arg2 on histone H3 (H3R2me2a) by protein arginine methyltransferase 6 (PRMT6) recruits the CPC to chromosome arms and facilitates histone H3S10 phosphorylation by Aurora B for chromosome condensation. Furthermore, in vitro assays show that Aurora B preferentially binds to the H3 peptide containing H3R2me2a and phosphorylates H3S10. Our findings indicate that the long-awaited key histone mark for CPC recruitment onto mitotic chromosomes is H3R2me2a, which is indispensable for maintaining appropriate CPC levels in dynamic translocation throughout mitosis. The proteins of the chromosomal passenger complex help chromosomes condense before cell division, but how this complex arrives at chromosomes was not known. Here the authors show that PRMT6 methylates histone H3 to recruit the chromosomal passenger complex.
Collapse
Affiliation(s)
- Seul Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Nam Hyun Kim
- Department of Pharmacology, College of Medicine, Catholic Kwandong University, Gangneung, 25601, Republic of Korea
| | - Ji Eun Park
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Jee Won Hwang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Nayeon Myung
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Ki-Tae Hwang
- Department of Surgery, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Young A Kim
- Department of Pathology, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Chang-Young Jang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Yong Kee Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
37
|
Takii R, Fujimoto M, Matsumoto M, Srivastava P, Katiyar A, Nakayama KI, Nakai A. The pericentromeric protein shugoshin 2 cooperates with HSF1 in heat shock response and RNA Pol II recruitment. EMBO J 2019; 38:e102566. [PMID: 31657478 DOI: 10.15252/embj.2019102566] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/12/2019] [Accepted: 09/12/2019] [Indexed: 12/17/2022] Open
Abstract
The recruitment of RNA polymerase II (Pol II) to core promoters is highly regulated during rapid induction of genes. In response to heat shock, heat shock transcription factor 1 (HSF1) is activated and occupies heat shock gene promoters. Promoter-bound HSF1 recruits general transcription factors and Mediator, which interact with Pol II, but stress-specific mechanisms of Pol II recruitment are unclear. Here, we show in comparative analyses of HSF1 paralogs and their mutants that HSF1 interacts with the pericentromeric adaptor protein shugoshin 2 (SGO2) during heat shock in mouse cells, in a manner dependent on inducible phosphorylation of HSF1 at serine 326, and recruits SGO2 to the HSP70 promoter. SGO2-mediated binding and recruitment of Pol II with a hypophosphorylated C-terminal domain promote expression of HSP70, implicating SGO2 as one of the coactivators that facilitate Pol II recruitment by HSF1. Furthermore, the HSF1-SGO2 complex supports cell survival and maintenance of proteostasis in heat shock conditions. These results exemplify a proteotoxic stress-specific mechanism of Pol II recruitment, which is triggered by phosphorylation of HSF1 during the heat shock response.
Collapse
Affiliation(s)
- Ryosuke Takii
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Mitsuaki Fujimoto
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Masaki Matsumoto
- Division of Proteomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Pratibha Srivastava
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Arpit Katiyar
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| | - Keiich I Nakayama
- Division of Proteomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.,Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Akira Nakai
- Department of Biochemistry and Molecular Biology, Yamaguchi University School of Medicine, Ube, Japan
| |
Collapse
|
38
|
Mirkovic M, Oliveira RA. Centromeric Cohesin: Molecular Glue and Much More. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2019; 56:485-513. [PMID: 28840250 DOI: 10.1007/978-3-319-58592-5_20] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sister chromatid cohesion, mediated by the cohesin complex, is a prerequisite for faithful chromosome segregation during mitosis. Premature release of sister chromatid cohesion leads to random segregation of the genetic material and consequent aneuploidy. Multiple regulatory mechanisms ensure proper timing for cohesion establishment, concomitant with DNA replication, and cohesion release during the subsequent mitosis. Here we summarize the most important phases of the cohesin cycle and the coordination of cohesion release with the progression through mitosis. We further discuss recent evidence that has revealed additional functions for centromeric localization of cohesin in the fidelity of mitosis in metazoans. Beyond its well-established role as "molecular glue", centromeric cohesin complexes are now emerging as a scaffold for multiple fundamental processes during mitosis, including the formation of correct chromosome and kinetochore architecture, force balance with the mitotic spindle, and the association with key molecules that regulate mitotic fidelity, particularly at the chromosomal inner centromere. Centromeric chromatin may be thus seen as a dynamic place where cohesin ensures mitotic fidelity by multiple means.
Collapse
Affiliation(s)
- Mihailo Mirkovic
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 2780-156, Oeiras, Portugal
| | - Raquel A Oliveira
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 2780-156, Oeiras, Portugal.
| |
Collapse
|
39
|
Vallardi G, Cordeiro MH, Saurin AT. A Kinase-Phosphatase Network that Regulates Kinetochore-Microtubule Attachments and the SAC. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2019; 56:457-484. [PMID: 28840249 DOI: 10.1007/978-3-319-58592-5_19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The KMN network (for KNL1, MIS12 and NDC80 complexes) is a hub for signalling at the outer kinetochore. It integrates the activities of two kinases (MPS1 and Aurora B) and two phosphatases (PP1 and PP2A-B56) to regulate kinetochore-microtubule attachments and the spindle assembly checkpoint (SAC). We will first discuss each of these enzymes separately, to describe how they are regulated at kinetochores and why this is important for their primary function in controlling either microtubule attachments or the SAC. We will then discuss why inhibiting any one of them individually produces secondary effects on all the others. This cross-talk may help to explain why all enzymes have been linked to both processes, even though the direct evidence suggests they each control only one. This chapter therefore describes how a network of kinases and phosphatases work together to regulate two key mitotic processes.
Collapse
Affiliation(s)
- Giulia Vallardi
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Marilia Henriques Cordeiro
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Adrian Thomas Saurin
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
40
|
Shugoshin protects centromere pairing and promotes segregation of nonexchange partner chromosomes in meiosis. Proc Natl Acad Sci U S A 2019; 116:9417-9422. [PMID: 31019073 PMCID: PMC6511000 DOI: 10.1073/pnas.1902526116] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Faithful chromosome segregation during meiosis I depends upon the formation of connections between homologous chromosomes. Crossovers between homologs connect the partners, allowing them to attach to the meiotic spindle as a unit, such that they migrate away from one another at anaphase I. Homologous partners also become connected by pairing of their centromeres in meiotic prophase. This centromere pairing can promote proper segregation at anaphase I of partners that have failed to become joined by a crossover. Centromere pairing is mediated by synaptonemal complex (SC) proteins that persist at the centromere when the SC disassembles. Here, using mouse spermatocyte and yeast model systems, we tested the role of shugoshin in promoting meiotic centromere pairing by protecting centromeric synaptonemal components from disassembly. The results show that shugoshin protects the centromeric SC in meiotic prophase and, in anaphase, promotes the proper segregation of partner chromosomes that are not linked by a crossover.
Collapse
|
41
|
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: 32] [Impact Index Per Article: 5.3] [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
|
42
|
Liang C, Zhang Z, Chen Q, Yan H, Zhang M, Xiang X, Yi Q, Pan X, Cheng H, Wang F. A positive feedback mechanism ensures proper assembly of the functional inner centromere during mitosis in human cells. J Biol Chem 2019; 294:1437-1450. [PMID: 30498087 PMCID: PMC6364785 DOI: 10.1074/jbc.ra118.006046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/27/2018] [Indexed: 01/25/2023] Open
Abstract
The inner centromere region of a mitotic chromosome critically regulates sister chromatid cohesion and kinetochore-microtubule attachments. However, the molecular mechanism underlying inner centromere assembly remains elusive. Here, using CRISPR/Cas9-based gene editing in HeLa cells, we disrupted the interaction of Shugoshin 1 (Sgo1) with histone H2A phosphorylated on Thr-120 (H2ApT120) to selectively release Sgo1 from mitotic centromeres. Interestingly, cells expressing the H2ApT120-binding defective mutant of Sgo1 have an elevated rate of chromosome missegregation accompanied by weakened centromeric cohesion and decreased centromere accumulation of the chromosomal passenger complex (CPC), an integral part of the inner centromere and a key player in the correction of erroneous kinetochore-microtubule attachments. When artificially tethered to centromeres, a Sgo1 mutant defective in binding protein phosphatase 2A (PP2A) is not able to support proper centromeric cohesion and CPC accumulation, indicating that the Sgo1-PP2A interaction is essential for the integrity of mitotic centromeres. We further provide evidence indicating that Sgo1 protects centromeric cohesin to create a binding site for the histone H3-associated protein kinase Haspin, which not only inhibits the cohesin release factor Wapl and thereby strengthens centromeric cohesion but also phosphorylates histone H3 at Thr-3 to position CPC at inner centromeres. Taken together, our findings reveal a positive feedback-based mechanism that ensures proper assembly of the functional inner centromere during mitosis. They further suggest a causal link between centromeric cohesion defects and chromosomal instability in cancer cells.
Collapse
Affiliation(s)
- Cai Liang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Zhenlei Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Qinfu Chen
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Haiyan Yan
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Miao Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Xingfeng Xiang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Qi Yi
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Xuan Pan
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Hankun Cheng
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Fangwei Wang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
43
|
Magnaghi-Jaulin L, Eot-Houllier G, Gallaud E, Giet R. Aurora A Protein Kinase: To the Centrosome and Beyond. Biomolecules 2019; 9:biom9010028. [PMID: 30650622 PMCID: PMC6359016 DOI: 10.3390/biom9010028] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/09/2019] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
Accurate chromosome segregation requires the perfect spatiotemporal rearrangement of the cellular cytoskeleton. Isolated more than two decades ago from Drosophila, Aurora A is a widespread protein kinase that plays key roles during cell division. Numerous studies have described the localisation of Aurora A at centrosomes, the mitotic spindle, and, more recently, at mitotic centromeres. In this review, we will summarise the cytoskeletal rearrangements regulated by Aurora A during cell division. We will also discuss the recent discoveries showing that Aurora A also controls not only the dynamics of the cortical proteins but also regulates the centromeric proteins, revealing new roles for this kinase during cell division.
Collapse
Affiliation(s)
- Laura Magnaghi-Jaulin
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| | - Grégory Eot-Houllier
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| | - Emmanuel Gallaud
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| | - Régis Giet
- University of Rennes, CNRS UMR 6290, IGDR-Institute of Genetics and Development of Rennes, F-35000 Rennes, France.
| |
Collapse
|
44
|
Meiosis-specific prophase-like pathway controls cleavage-independent release of cohesin by Wapl phosphorylation. PLoS Genet 2019; 15:e1007851. [PMID: 30605471 PMCID: PMC6317811 DOI: 10.1371/journal.pgen.1007851] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/25/2018] [Indexed: 01/09/2023] Open
Abstract
Sister chromatid cohesion on chromosome arms is essential for the segregation of homologous chromosomes during meiosis I while it is dispensable for sister chromatid separation during mitosis. It was assumed that, unlike the situation in mitosis, chromosome arms retain cohesion prior to onset of anaphase-I. Paradoxically, reduced immunostaining signals of meiosis-specific cohesin, including the kleisin Rec8, were observed on chromosomes during late prophase-I of budding yeast. This decrease is seen in the absence of Rec8 cleavage and depends on condensin-mediated recruitment of Polo-like kinase (PLK/Cdc5). In this study, we confirmed that this release indeed accompanies the dissociation of acetylated Smc3 as well as Rec8 from meiotic chromosomes during late prophase-I. This release requires, in addition to PLK, the cohesin regulator, Wapl (Rad61/Wpl1 in yeast), and Dbf4-dependent Cdc7 kinase (DDK). Meiosis-specific phosphorylation of Rad61/Wpl1 and Rec8 by PLK and DDK collaboratively promote this release. This process is similar to the vertebrate “prophase” pathway for cohesin release during G2 phase and pro-metaphase. In yeast, meiotic cohesin release coincides with PLK-dependent compaction of chromosomes in late meiotic prophase-I. We suggest that yeast uses this highly regulated cleavage-independent pathway to remove cohesin during late prophase-I to facilitate morphogenesis of condensed metaphase-I chromosomes. In meiosis the life and health of future generations is decided upon. Any failure in chromosome segregation has a detrimental impact. Therefore, it is currently believed that the physical connections between homologous chromosomes are maintained by meiotic cohesin with exceptional stability. Indeed, it was shown that cohesive cohesin does not show an appreciable turnover during long periods in oocyte development. In this context, it was long assumed but not properly investigated, that the prophase pathway for cohesin release would be specific to mitosis and would be safely suppressed during meiosis so as not to endanger essential connections between chromosomes. However, a previous study on budding yeast meiosis suggests the presence of cleavage-independent pathway of cohesin release during late prophase-I. In the work presented here we confirmed that the prophase pathway is not suppressed during meiosis, at least in budding yeast and showed that this cleavage-independent release is regulated by meiosis-specific phosphorylation of two cohesin subunits, Rec8 and Rad61(Wapl) by two cell-cycle regulators, PLK and DDK. Our results suggest that late meiotic prophase-I actively controls cohesin dynamics on meiotic chromosomes for chromosome segregation.
Collapse
|
45
|
Bohr T, Nelson CR, Giacopazzi S, Lamelza P, Bhalla N. Shugoshin Is Essential for Meiotic Prophase Checkpoints in C. elegans. Curr Biol 2018; 28:3199-3211.e3. [PMID: 30293721 PMCID: PMC6200582 DOI: 10.1016/j.cub.2018.08.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 07/16/2018] [Accepted: 08/08/2018] [Indexed: 10/28/2022]
Abstract
The conserved factor Shugoshin is dispensable in C. elegans for the two-step loss of sister chromatid cohesion that directs the proper segregation of meiotic chromosomes. We show that the C. elegans ortholog of Shugoshin, SGO-1, is required for checkpoint activity in meiotic prophase. This role in checkpoint function is similar to that of conserved proteins that structure meiotic chromosome axes. Indeed, null sgo-1 mutants exhibit additional phenotypes similar to that of a partial loss-of-function allele of the axis component, HTP-3: premature synaptonemal complex disassembly, the activation of alternate DNA repair pathways, and an inability to recruit a conserved effector of the DNA damage pathway, HUS-1. SGO-1 localizes to pre-meiotic nuclei when HTP-3 is present but not yet loaded onto chromosome axes and genetically interacts with a central component of the cohesin complex, SMC-3, suggesting that it contributes to meiotic chromosome metabolism early in meiosis by regulating cohesin. We propose that SGO-1 acts during pre-meiotic replication to ensure fully functional meiotic chromosome architecture, rendering these chromosomes competent for checkpoint activity and normal progression of meiotic recombination. Given that most research on Shugoshin has focused on its regulation of sister chromatid cohesion during chromosome segregation, this novel role may be conserved but previously uncharacterized in other organisms. Further, our findings expand the repertoire of Shugoshin's functions beyond coordinating regulatory activities at the centromere.
Collapse
Affiliation(s)
- Tisha Bohr
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Christian R Nelson
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Stefani Giacopazzi
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Piero Lamelza
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Needhi Bhalla
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
46
|
Kruitwagen T, Chymkowitch P, Denoth-Lippuner A, Enserink J, Barral Y. Centromeres License the Mitotic Condensation of Yeast Chromosome Arms. Cell 2018; 175:780-795.e15. [PMID: 30318142 PMCID: PMC6197839 DOI: 10.1016/j.cell.2018.09.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 06/14/2018] [Accepted: 09/07/2018] [Indexed: 12/18/2022]
Abstract
During mitosis, chromatin condensation shapes chromosomes as separate, rigid, and compact sister chromatids to facilitate their segregation. Here, we show that, unlike wild-type yeast chromosomes, non-chromosomal DNA circles and chromosomes lacking a centromere fail to condense during mitosis. The centromere promotes chromosome condensation strictly in cis through recruiting the kinases Aurora B and Bub1, which trigger the autonomous condensation of the entire chromosome. Shugoshin and the deacetylase Hst2 facilitated spreading the condensation signal to the chromosome arms. Targeting Aurora B to DNA circles or centromere-ablated chromosomes or releasing Shugoshin from PP2A-dependent inhibition bypassed the centromere requirement for condensation and enhanced the mitotic stability of DNA circles. Our data indicate that yeast cells license the chromosome-autonomous condensation of their chromatin in a centromere-dependent manner, excluding from this process non-centromeric DNA and thereby inhibiting their propagation.
Collapse
Affiliation(s)
- Tom Kruitwagen
- Institute of Biochemistry, Biology Department, ETH Zurich, 8093 Zurich, Switzerland
| | - Pierre Chymkowitch
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
| | | | - Jorrit Enserink
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway; Faculty of Medicine, Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Faculty of Mathematics and Natural Sciences, Department of Biosciences, University of Oslo, Norway
| | - Yves Barral
- Institute of Biochemistry, Biology Department, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
47
|
Shugoshin 1 is dislocated by KSHV-encoded LANA inducing aneuploidy. PLoS Pathog 2018; 14:e1007253. [PMID: 30212568 PMCID: PMC6136811 DOI: 10.1371/journal.ppat.1007253] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/31/2018] [Indexed: 11/30/2022] Open
Abstract
Shugoshin-1 (Sgo1) protects the integrity of the centromeres, and H2A phosphorylation is critical for this process. The mitotic checkpoint kinase Bub1, phosphorylates H2A and ensures fidelity of chromosome segregation and chromosome number. Oncogenic KSHV induces genetic alterations through chromosomal instability (CIN), and its essential antigen LANA regulates Bub1. We show that LANA inhibits Bub1 phosphorylation of H2A and Cdc20, important for chromosome segregation and mitotic signaling. Inhibition of H2A phosphorylation at residue T120 by LANA resulted in dislocation of Sgo1, and cohesin from the centromeres. Arrest of Cdc20 phosphorylation also rescued degradation of Securin and Cyclin B1 at mitotic exit, and interaction of H2A, and Cdc20 with Bub1 was inhibited by LANA. The N-terminal nuclear localization sequence domain of LANA was essential for LANA and Bub1 interaction, reversed LANA inhibited phosphorylation of H2A and Cdc20, and attenuated LANA-induced aneuploidy and cell proliferation. This molecular mechanism whereby KSHV-induced CIN, demonstrated that the NNLS of LANA is a promising target for development of anti-viral therapies targeting KSHV associated cancers. KSHV is a known oncogenic herpes virus associated with human malignancies and lymphoproliferative disorders, which includes Kaposi’s sarcoma, Primary effusion lymphoma, and Multicentric Castleman’s disease. KSHV disrupts the G1 and G2/M checkpoints through multiple pathways. Whether KSHV can directly interfere with spindle checkpoints is not known. Impairment of the mitotic checkpoint protein Bub1 leads to CIN and oncogenesis through displacement of Shugoshin-1. KSHV associated diseases have genetic alterations which are driven by chromosomal instability (CIN), as seen in numerous viral-associated cancer cells. Here we examined the molecular mechanism behind KSHV-induced CIN. We showed that the latent antigen LANA, encoded by KSHV, inhibits Bub1 phosphorylation of H2A and Cdc20, and this led to the dislocation of Shugoshin-1. Our studies demonstrated the direct induction of aneuploidy by LANA. The NNLS domain of LANA serves as an anchor for LANA to promote its multiple functions. We also showed that the NNLS polypeptide can antagonize LANA’s inhibition on Bub1 kinase function, and so rescue the aneuploidy induced by LANA. Development of this property of NNLS is potentially useful for targeted elimination of KSHV-associated cancers.
Collapse
|
48
|
Tripartite Chromatin Localization of Budding Yeast Shugoshin Involves Higher-Ordered Architecture of Mitotic Chromosomes. G3-GENES GENOMES GENETICS 2018; 8:2901-2911. [PMID: 30002083 PMCID: PMC6118306 DOI: 10.1534/g3.118.200522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The spindle assembly checkpoint (SAC) is key to faithful segregation of chromosomes. One requirement that satisfies SAC is appropriate tension between sister chromatids at the metaphase-anaphase juncture. Proper tension generated by poleward pulling of mitotic spindles signals biorientation of the underlying chromosome. In the budding yeast, the tension status is monitored by the conserved Shugoshin protein, Sgo1p, and the tension sensing motif (TSM) of histone H3. ChIP-seq reveals a unique TSM-dependent, tripartite domain of Sgo1p in each mitotic chromosome. This domain consists of one centromeric and two flanking peaks 3 - 4 kb away, present exclusively in mitosis. Strikingly, this trident motif coincides with cohesin localization, but only at the centromere and the two immediate adjacent loci, despite that cohesin is enriched at numerous regions throughout mitotic chromosomes. Chromosome conformation capture assays reveal apparent looping at the centromeric and pericentric regions. The TSM-Sgo1p-cohesin triad is therefore at the center stage of higher-ordered chromatin architecture for error-free segregation.
Collapse
|
49
|
Maeda K, Yoneda M, Nakagawa T, Ikeda K, Higashi M, Nakagawa K, Miyakoda M, Yui K, Oda H, Inoue S, Ito T. Defects in centromeric/pericentromeric histone H2A T120 phosphorylation by hBUB1 cause chromosome missegregation producing multinucleated cells. Genes Cells 2018; 23:828-838. [PMID: 30112853 DOI: 10.1111/gtc.12630] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/17/2018] [Accepted: 07/17/2018] [Indexed: 11/26/2022]
Abstract
Histone H2A phosphorylation plays a role both in chromatin condensation during mitosis and in transcriptional activation during the G1/S transition. Bub1 and NHK1/VRK1 have been identified as histone H2A kinases. However, little is known about the importance of histone H2A phosphorylation in chromosome segregation. Here, we expressed recombinant hBUB1 and confirmed that it phosphorylates histone H2A T120 in the in vitro-assembled nucleosome. Knockdown (KD) of BUB1 decreases bulk H2A T120 phosphorylation in HeLa cells, whereas hBUB1 is upregulated during mitosis, which corresponds with H2A T120 phosphorylation. ChIP-qPCR of the DXZ1 centromeric and γ-ALR pericentromeric region showed that BUB1 localizes to this region and increases local H2A T120 phosphorylation during M phase. BUB1 KD did not induce apoptosis but increased the M phase cell population, as detected by flow cytometry. BUB1 KD also caused an abnormal metaphase and telophase, resulting in multinucleated cells and impaired cancer cell growth both in vitro and in vivo. Over-expression of the histone H2A T120D or T120E mutations, which mimic phosphorylated threonine, decreased the number of multinucleated cells caused by BUB1 KD. These results strengthen the apparent importance of BUB1-mediated H2A T120 phosphorylation in normal mitosis.
Collapse
Affiliation(s)
- Katsutoshi Maeda
- Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki, Japan.,Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Oda Clinic, Hiroshima, Japan
| | - Mitsuhiro Yoneda
- Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Takeya Nakagawa
- Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Miki Higashi
- Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Kaori Nakagawa
- Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Mana Miyakoda
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Katsuyuki Yui
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | | | - Satoshi Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan.,Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Takashi Ito
- Department of Biochemistry, Nagasaki University School of Medicine, Nagasaki, Japan.,Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
50
|
Morales C, Losada A. Establishing and dissolving cohesion during the vertebrate cell cycle. Curr Opin Cell Biol 2018; 52:51-57. [PMID: 29433064 DOI: 10.1016/j.ceb.2018.01.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/18/2018] [Accepted: 01/29/2018] [Indexed: 01/28/2023]
Abstract
Replicated chromatids are held together from the time they emerge from the replication fork until their separation in anaphase. This process, known as cohesion, promotes faithful DNA repair by homologous recombination in interphase and ensures accurate chromosome segregation in mitosis. Identification of cohesin thirty years ago solved a long-standing question about the nature of the linkage keeping together the sister chromatids. Cohesin is an evolutionarily conserved complex composed of a heterodimer of the Structural Maintenance of Chromosomes (SMC) family of ATPases, Smc1 and Smc3, the kleisin subunit Rad21 and a Huntingtin/EF3/PP2A/Tor1 (HEAT) repeat domain-containing subunit named SA/STAG. In addition to mediating cohesion, cohesin plays a major role in genome organization. Cohesin functions rely on the ability of the complex to entrap DNA topologically and in a dynamic manner. Establishment of cohesion during S phase requires coordination with the DNA replication machinery and restricts the dynamic behaviour of at least a fraction of cohesin. Dissolution of cohesion in subsequent mitosis is regulated by multiple mechanisms that ensure that daughter cells receive the correct number of intact chromosomes. We here review recent progress on our understanding of how these processes are regulated in somatic vertebrate cells.
Collapse
Affiliation(s)
- Carmen Morales
- Chromosome Dynamics Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ana Losada
- Chromosome Dynamics Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| |
Collapse
|