51
|
Sun Y, Chen Z, Liu G, Chen X, Shi Z, Feng H, Yu L, Li G, Ding K, Huang H, Zhang Z, Xu S. Discovery of a potent and selective covalent threonine tyrosine kinase (TTK) inhibitor. Bioorg Chem 2024; 143:107053. [PMID: 38159497 DOI: 10.1016/j.bioorg.2023.107053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/07/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Threonine tyrosine kinase (TTK) is a critical component of the spindle assembly checkpoint and plays a pivotal role in mitosis. TTK has been identified as a potential therapeutic target for human cancers. Here, we describe our design, synthesis and evaluation of a class of covalent TTK inhibitors, exemplified by 16 (SYL1073). Compound 16 potently inhibits TTK kinase with an IC50 of 0.016 μM and displays improved selectivity in a panel of kinases. Mass spectrometry analysis reveals that 16 covalently binds to the C604 cysteine residue in the hinge region of the TTK kinase domain. Furthermore, 16 achieves strong potency in inhibiting the growth of various human cancer cell lines, outperforming its relative reversible inhibitor, and eliciting robust downstream effects. Taken together, compound 16 provides a valuable lead compound for further optimization toward the development of drug for treatment of human cancers.
Collapse
Affiliation(s)
- Yaoliang Sun
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhiwen Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Guobin Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoai Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zihan Shi
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Science, 19 Yuquan Road, Beijing 100049, China
| | - Huixu Feng
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lei Yu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Guodong Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - He Huang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Zhang Zhang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Shilin Xu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; University of Chinese Academy of Science, 19 Yuquan Road, Beijing 100049, China.
| |
Collapse
|
52
|
Cao J, Hori T, Ariyoshi M, Fukagawa T. Artificial tethering of constitutive centromere-associated network proteins induces CENP-A deposition without Knl2 in DT40 cells. J Cell Sci 2024; 137:jcs261639. [PMID: 38319136 DOI: 10.1242/jcs.261639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/22/2023] [Indexed: 02/07/2024] Open
Abstract
The kinetochore is an essential structure for chromosome segregation. Although the kinetochore is usually formed on a centromere locus, it can be artificially formed at a non-centromere locus by protein tethering. An artificial kinetochore can be formed by tethering of CENP-C or CENP-I, members of the constitutive centromere-associated network (CCAN). However, how CENP-C or CENP-I recruit the centromere-specific histone CENP-A to form an artificial kinetochore remains unclear. In this study, we analyzed this issue using the tethering assay combined with an auxin-inducible degron (AID)-based knockout method in chicken DT40 cells. We found that tethering of CENP-C or CENP-I induced CENP-A incorporation at the non-centromeric locus in the absence of Knl2 (or MIS18BP1), a component of the Mis18 complex, and that Knl2 tethering recruited CENP-A in the absence of CENP-C. We also showed that CENP-C coimmunoprecipitated with HJURP, independently of Knl2. Considering these results, we propose that CENP-C recruits CENP-A by HJURP binding to form an artificial kinetochore. Our results suggest that CENP-C or CENP-I exert CENP-A recruitment activity, independently of Knl2, for artificial kinetochore formation in chicken DT40 cells. This gives us a new insight into mechanisms for CENP-A incorporation.
Collapse
Affiliation(s)
- JingHui Cao
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Tetsuya Hori
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mariko Ariyoshi
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Tatsuo Fukagawa
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
53
|
Mihalas BP, Pieper GH, Aboelenain M, Munro L, Srsen V, Currie CE, Kelly DA, Hartshorne GM, Telfer EE, McAinsh AD, Anderson RA, Marston AL. Age-dependent loss of cohesion protection in human oocytes. Curr Biol 2024; 34:117-131.e5. [PMID: 38134935 PMCID: PMC7617652 DOI: 10.1016/j.cub.2023.11.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 11/05/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023]
Abstract
Aneuploid human eggs (oocytes) are a major cause of infertility, miscarriage, and chromosomal disorders. Such aneuploidies increase greatly as women age, with defective linkages between sister chromatids (cohesion) in meiosis as a common cause. We found that loss of a specific pool of the cohesin protector protein, shugoshin 2 (SGO2), may contribute to this phenomenon. Our data indicate that SGO2 preserves sister chromatid cohesion in meiosis by protecting a "cohesin bridge" between sister chromatids. In human oocytes, SGO2 localizes to both sub-centromere cups and the pericentromeric bridge, which spans the sister chromatid junction. SGO2 normally colocalizes with cohesin; however, in meiosis II oocytes from older women, SGO2 is frequently lost from the pericentromeric bridge and sister chromatid cohesion is weakened. MPS1 and BUB1 kinase activities maintain SGO2 at sub-centromeres and the pericentromeric bridge. Removal of SGO2 throughout meiosis I by MPS1 inhibition reduces cohesion protection, increasing the incidence of single chromatids at meiosis II. Therefore, SGO2 deficiency in human oocytes can exacerbate the effects of maternal age by rendering residual cohesin at pericentromeres vulnerable to loss in anaphase I. Our data show that impaired SGO2 localization weakens cohesion integrity and may contribute to the increased incidence of aneuploidy observed in human oocytes with advanced maternal age.
Collapse
Affiliation(s)
- Bettina P Mihalas
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Gerard H Pieper
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Mansour Aboelenain
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK; Theriogenology department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Lucy Munro
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Vlastimil Srsen
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Cerys E Currie
- Centre for Mechanochemical Cell Biology & Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK
| | - David A Kelly
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Geraldine M Hartshorne
- Centre for Mechanochemical Cell Biology & Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK; University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
| | - Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Andrew D McAinsh
- Centre for Mechanochemical Cell Biology & Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Gibbet Hill, Coventry CV4 7AL, UK
| | - Richard A Anderson
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Adele L Marston
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, UK.
| |
Collapse
|
54
|
Fuentes‐Antrás J, Bedard PL, Cescon DW. Seize the engine: Emerging cell cycle targets in breast cancer. Clin Transl Med 2024; 14:e1544. [PMID: 38264947 PMCID: PMC10807317 DOI: 10.1002/ctm2.1544] [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: 10/09/2023] [Revised: 12/10/2023] [Accepted: 12/31/2023] [Indexed: 01/25/2024] Open
Abstract
Breast cancer arises from a series of molecular alterations that disrupt cell cycle checkpoints, leading to aberrant cell proliferation and genomic instability. Targeted pharmacological inhibition of cell cycle regulators has long been considered a promising anti-cancer strategy. Initial attempts to drug critical cell cycle drivers were hampered by poor selectivity, modest efficacy and haematological toxicity. Advances in our understanding of the molecular basis of cell cycle disruption and the mechanisms of resistance to CDK4/6 inhibitors have reignited interest in blocking specific components of the cell cycle machinery, such as CDK2, CDK4, CDK7, PLK4, WEE1, PKMYT1, AURKA and TTK. These targets play critical roles in regulating quiescence, DNA replication and chromosome segregation. Extensive preclinical data support their potential to overcome CDK4/6 inhibitor resistance, induce synthetic lethality or sensitise tumours to immune checkpoint inhibitors. This review provides a biological and drug development perspective on emerging cell cycle targets and novel inhibitors, many of which exhibit favourable safety profiles and promising activity in clinical trials.
Collapse
Affiliation(s)
- Jesús Fuentes‐Antrás
- Division of Medical Oncology and HematologyDepartment of MedicinePrincess Margaret Cancer CentreUniversity Health NetworkUniversity of TorontoTorontoOntarioCanada
- NEXT OncologyHospital Universitario QuironSalud MadridMadridSpain
| | - Philippe L. Bedard
- Division of Medical Oncology and HematologyDepartment of MedicinePrincess Margaret Cancer CentreUniversity Health NetworkUniversity of TorontoTorontoOntarioCanada
| | - David W. Cescon
- Division of Medical Oncology and HematologyDepartment of MedicinePrincess Margaret Cancer CentreUniversity Health NetworkUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
55
|
Zeng Y, Ren X, Jin P, Zhang Y, Zhuo M, Wang J. Development of MPS1 Inhibitors: Recent Advances and Perspectives. J Med Chem 2023; 66:16484-16514. [PMID: 38095579 DOI: 10.1021/acs.jmedchem.3c00963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Monopolar spindle kinase 1 (MPS1) plays a pivotal role as a dual-specificity kinase governing spindle assembly checkpoint activation and sister chromatid separation in mitosis. Its overexpression has been observed in various human malignancies. MPS1 reduces spindle assembly checkpoint sensitivity, allowing tumor cells with a high degree of aneuploidy to complete mitosis and survive. Thus, MPS1 has emerged as a promising candidate for cancer therapy. Despite the identification of numerous MPS1 inhibitors, only five have advanced to clinical trials with none securing FDA approval for cancer treatment. In this perspective, we provide a concise overview of the structural and functional characteristics of MPS1 by highlighting its relevance to cancer. Additionally, we explore the structure-activity relationships, selectivity, and pharmacokinetics of MPS1 inhibitors featuring diverse scaffolds. Moreover, we review the reported work on enhancing MPS1 inhibitor selectivity, offering valuable insights into the discovery of novel, highly potent small-molecule MPS1 inhibitors.
Collapse
Affiliation(s)
- Yangjie Zeng
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Xiaodong Ren
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Pengyao Jin
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Yali Zhang
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Ming Zhuo
- Medical College, Guizhou University, Guiyang, Guizhou 550025, China
| | - Jubo Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
56
|
Campos G, Sciorio R, Fleming S. Healthy Live Births after the Transfer of Mosaic Embryos: Self-Correction or PGT-A Overestimation? Genes (Basel) 2023; 15:18. [PMID: 38275600 PMCID: PMC10815078 DOI: 10.3390/genes15010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/27/2024] Open
Abstract
The implementation of next generation sequencing (NGS) in preimplantation genetic testing for aneuploidy (PGT-A) has led to a higher prevalence of mosaic diagnosis within the trophectoderm (TE) sample. Regardless, mosaicism could potentially increase the rate of live-born children with chromosomic syndromes, though available data from the transfer of embryos with putative PGT-A mosaicism are scarce but reassuring. Even with lower implantation and higher miscarriage rates, mosaic embryos can develop into healthy live births. Therefore, this urges an explanation for the disappearance of aneuploid cells throughout development, to provide guidance in the management of mosaicism in clinical practice. Technical overestimation of mosaicism, together with some sort of "self-correction" mechanisms during the early post-implantation stages, emerged as potential explanations. Unlike the animal model, in which the elimination of genetically abnormal cells from the future fetal lineage has been demonstrated, in human embryos this capability remains unverified even though the germ layer displays an aneuploidy-induced cell death lineage preference with higher rates of apoptosis in the inner cell mass (ICM) than in the TE cells. Moreover, the reported differential dynamics of cell proliferation and apoptosis between euploid, mosaic, and aneuploid embryos, together with pro-apoptosis gene products (cfDNA and mRNA) and extracellular vesicles identified in the blastocoel fluid, may support the hypothesis of apoptosis as a mechanism to purge the preimplantation embryo of aneuploid cells. Alternative hypotheses, like correction of aneuploidy by extrusion of a trisomy chromosome or by monosomic chromosome duplication, are even, though they represent an extremely rare phenomenon. On the other hand, the technical limitations of PGT-A analysis may lead to inaccuracy in embryo diagnoses, identifying as "mosaic" those embryos that are uniformly euploid or aneuploid. NGS assumption of "intermediate copy number profiles" as evidence of a mixture of euploid and aneuploid cells in a single biopsy has been reported to be poorly predictive in cases of mosaicism diagnosis. Additionally, the concordance found between the TE and the ICM in cases of TE biopsies displaying mosaicism is lower than expected, and it correlates differently depending on the type (whole chromosome versus segmental) and the level of mosaicism reported. Thus, in cases of low-/medium-level mosaicism (<50%), aneuploid cells would rarely involve the ICM and other regions. However, in high-level mosaics (≥50%), abnormal cells in the ICM should display higher prevalence, revealing more uniform aneuploidy in most embryos, representing a technical variation in the uniform aneuploidy range, and therefore might impair the live birth rate.
Collapse
Affiliation(s)
- Gerard Campos
- Geisinger Medical Center, Women’s Health Fertility Clinic, Danville, PA 17821, USA;
- GIREXX Fertility Clinics, C. de Cartagena, 258, 08025 Girona, Spain
| | - Romualdo Sciorio
- Fertility Medicine and Gynaecological Endocrinology Unit, Department Woman-Mother-Child, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Steven Fleming
- Discipline of Anatomy & Histology, School of Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia;
| |
Collapse
|
57
|
Cmentowski V, Ciossani G, d'Amico E, Wohlgemuth S, Owa M, Dynlacht B, Musacchio A. RZZ-Spindly and CENP-E form an integrated platform to recruit dynein to the kinetochore corona. EMBO J 2023; 42:e114838. [PMID: 37984321 PMCID: PMC10711656 DOI: 10.15252/embj.2023114838] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/22/2023] Open
Abstract
Chromosome biorientation on the mitotic spindle is prerequisite to errorless genome inheritance. CENP-E (kinesin-7) and dynein-dynactin (DD), microtubule motors with opposite polarity, promote biorientation from the kinetochore corona, a polymeric structure whose assembly requires MPS1 kinase. The corona's building block consists of ROD, Zwilch, ZW10, and the DD adaptor Spindly (RZZS). How CENP-E and DD are scaffolded and mutually coordinated in the corona remains unclear. Here, we show that when corona assembly is prevented through MPS1 inhibition, CENP-E is absolutely required to retain RZZS at kinetochores. An RZZS phosphomimetic mutant bypasses this requirement, demonstrating the existence of a second receptor for polymeric RZZS. With active MPS1, CENP-E is dispensable for corona expansion, but strictly required for physiological kinetochore accumulation of DD. Thus, we identify the corona as an integrated scaffold where CENP-E kinesin controls DD kinetochore loading for coordinated bidirectional transport of chromosome cargo.
Collapse
Affiliation(s)
- Verena Cmentowski
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
- Centre for Medical Biotechnology, Faculty of BiologyUniversity Duisburg‐EssenEssenGermany
| | - Giuseppe Ciossani
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
- Present address:
European Institute of OncologyMilanItaly
| | - Ennio d'Amico
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
- Present address:
Division of Structural StudiesMRC Laboratory of Molecular BiologyCambridgeUK
| | - Sabine Wohlgemuth
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Mikito Owa
- Department of PathologyNew York University Cancer Institute, New York University School of MedicineNew YorkNYUSA
| | - Brian Dynlacht
- Department of PathologyNew York University Cancer Institute, New York University School of MedicineNew YorkNYUSA
| | - Andrea Musacchio
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
- Centre for Medical Biotechnology, Faculty of BiologyUniversity Duisburg‐EssenEssenGermany
| |
Collapse
|
58
|
Parnell EJ, Jenson E, Miller MP. An interaction hub on Ndc80 complex facilitates dynamic recruitment of Mps1 to yeast kinetochores to promote accurate chromosome segregation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.566082. [PMID: 37986816 PMCID: PMC10659343 DOI: 10.1101/2023.11.07.566082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Accurate chromosome segregation relies on kinetochores carrying out multiple functions, including establishing and maintaining microtubule attachments, forming precise bioriented attachments between sister chromatids, and activating the spindle assembly checkpoint. Central to these processes is the highly conserved Ndc80 complex. This kinetochore subcomplex interacts directly with microtubules, but also serves as a critical platform for recruiting kinetochore-associated factors and as a key substrate for error correction kinases. The precise manner in which these kinetochore factors interact, and regulate each other's function, remains unknown - considerably hindering our understanding of how Ndc80 complex-dependent processes function together to orchestrate accurate chromosome segregation. Here, we aimed to uncover the role of Nuf2's CH domain, a component of the Ndc80 complex, in ensuring accurate chromosome segregation. Through extensive mutational analysis, we identified a conserved "interaction hub" comprising two segments in Nuf2's CH domain, forming the binding site for Mps1 within the yeast Ndc80 complex. Intriguingly, the interaction between Mps1 and the Ndc80 complex seems to be subject to regulation by competitive binding with other factors. Mutants disrupting this interaction hub exhibit defects in spindle assembly checkpoint function and severe chromosome segregation errors. Significantly, specifically restoring Mps1-Ndc80 complex association rescues these defects. Our findings shed light on the intricate regulation of Ndc80 complex-dependent functions and highlight the essential role of Mps1 in kinetochore biorientation and accurate chromosome segregation.
Collapse
Affiliation(s)
- Emily J. Parnell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Erin Jenson
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Matthew P. Miller
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
59
|
Mihajlović AI, Byers C, Reinholdt L, FitzHarris G. Spindle assembly checkpoint insensitivity allows meiosis-II despite chromosomal defects in aged eggs. EMBO Rep 2023; 24:e57227. [PMID: 37795949 PMCID: PMC10626445 DOI: 10.15252/embr.202357227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023] Open
Abstract
Chromosome segregation errors in mammalian oocyte meiosis lead to developmentally compromised aneuploid embryos and become more common with advancing maternal age. Known contributors include age-related chromosome cohesion loss and spindle assembly checkpoint (SAC) fallibility in meiosis-I. But how effective the SAC is in meiosis-II and how this might contribute to age-related aneuploidy is unknown. Here, we developed genetic and pharmacological approaches to directly address the function of the SAC in meiosis-II. We show that the SAC is insensitive in meiosis-II oocytes and that as a result misaligned chromosomes are randomly segregated. Whilst SAC ineffectiveness in meiosis-II is not age-related, it becomes most prejudicial in oocytes from older females because chromosomes that prematurely separate by age-related cohesion loss become misaligned in meiosis-II. We show that in the absence of a robust SAC in meiosis-II these age-related misaligned chromatids are missegregated and lead to aneuploidy. Our data demonstrate that the SAC fails to prevent cell division in the presence of misaligned chromosomes in oocyte meiosis-II, which explains how age-related cohesion loss can give rise to aneuploid embryos.
Collapse
Affiliation(s)
| | - Candice Byers
- The Institute for Experiential AI, Roux InstituteNortheastern UniversityPortlandMEUSA
| | | | | |
Collapse
|
60
|
Dong J, Jin L, Bao S, Chen B, Zeng Y, Luo Y, Du X, Sang Q, Wu T, Wang L. Ectopic expression of human TUBB8 leads to increased aneuploidy in mouse oocytes. Cell Discov 2023; 9:105. [PMID: 37875488 PMCID: PMC10598138 DOI: 10.1038/s41421-023-00599-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 08/26/2023] [Indexed: 10/26/2023] Open
Abstract
Aneuploidy seriously compromises female fertility and increases incidence of birth defects. Rates of aneuploidy in human eggs from even young women are significantly higher than those in other mammals. However, intrinsic genetic factors underlying this high incidence of aneuploidy in human eggs remain largely unknown. Here, we found that ectopic expression of human TUBB8 in mouse oocytes increases rates of aneuploidy by causing kinetochore-microtubule (K-MT) attachment defects. Stretched bivalents in mouse oocytes expressing TUBB8 are under less tension, resulting in continuous phosphorylation status of HEC1 by AURKB/C at late metaphase I that impairs the established correct K-MT attachments. This reduced tension in stretched bivalents likely correlates with decreased recruitment of KIF11 on meiotic spindles. We also found that ectopic expression of TUBB8 without its C-terminal tail decreases aneuploidy rates by reducing erroneous K-MT attachments. Importantly, variants in the C-terminal tail of TUBB8 were identified in patients with recurrent miscarriages. Ectopic expression of an identified TUBB8 variant in mouse oocytes also compromises K-MT attachments and increases aneuploidy rates. In conclusion, our study provides novel understanding for physiological mechanisms of aneuploidy in human eggs as well as for pathophysiological mechanisms involved in recurrent miscarriages.
Collapse
Affiliation(s)
- Jie Dong
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Liping Jin
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shihua Bao
- Department of Reproductive Immunology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Fudan University, Shanghai, China
| | - Yang Zeng
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Yuxi Luo
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Xingzhu Du
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China.
| | - Tianyu Wu
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China.
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University and Institutes of Biomedical Sciences, The State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China.
| |
Collapse
|
61
|
Corno A, Cordeiro MH, Allan LA, Lim Q, Harrington E, Smith RJ, Saurin AT. A bifunctional kinase-phosphatase module balances mitotic checkpoint strength and kinetochore-microtubule attachment stability. EMBO J 2023; 42:e112630. [PMID: 37712330 PMCID: PMC10577578 DOI: 10.15252/embj.2022112630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023] Open
Abstract
Two major mechanisms safeguard genome stability during mitosis: the mitotic checkpoint delays mitosis until all chromosomes have attached to microtubules, and the kinetochore-microtubule error-correction pathway keeps this attachment process free from errors. We demonstrate here that the optimal strength and dynamics of these processes are set by a kinase-phosphatase pair (PLK1-PP2A) that engage in negative feedback from adjacent phospho-binding motifs on the BUB complex. Uncoupling this feedback to skew the balance towards PLK1 produces a strong checkpoint, hypostable microtubule attachments and mitotic delays. Conversely, skewing the balance towards PP2A causes a weak checkpoint, hyperstable microtubule attachments and chromosome segregation errors. These phenotypes are associated with altered BUB complex recruitment to KNL1-MELT motifs, implicating PLK1-PP2A in controlling auto-amplification of MELT phosphorylation. In support, KNL1-BUB disassembly becomes contingent on PLK1 inhibition when KNL1 is engineered to contain excess MELT motifs. This elevates BUB-PLK1/PP2A complex levels on metaphase kinetochores, stabilises kinetochore-microtubule attachments, induces chromosome segregation defects and prevents KNL1-BUB disassembly at anaphase. Together, these data demonstrate how a bifunctional PLK1/PP2A module has evolved together with the MELT motifs to optimise BUB complex dynamics and ensure accurate chromosome segregation.
Collapse
Affiliation(s)
- Andrea Corno
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Marilia H Cordeiro
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Lindsey A Allan
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Qian‐Wei Lim
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Elena Harrington
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Richard J Smith
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| | - Adrian T Saurin
- Cellular and Systems Medicine, School of MedicineUniversity of DundeeDundeeUK
| |
Collapse
|
62
|
Zhou KD, Zhang CX, Niu FR, Bai HC, Wu DD, Deng JC, Qian HY, Jiang YL, Ma W. Exploring Plant Meiosis: Insights from the Kinetochore Perspective. Curr Issues Mol Biol 2023; 45:7974-7995. [PMID: 37886947 PMCID: PMC10605258 DOI: 10.3390/cimb45100504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/12/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
The central player for chromosome segregation in both mitosis and meiosis is the macromolecular kinetochore structure, which is assembled by >100 structural and regulatory proteins on centromere DNA. Kinetochores play a crucial role in cell division by connecting chromosomal DNA and microtubule polymers. This connection helps in the proper segregation and alignment of chromosomes. Additionally, kinetochores can act as a signaling hub, regulating the start of anaphase through the spindle assembly checkpoint, and controlling the movement of chromosomes during anaphase. However, the role of various kinetochore proteins in plant meiosis has only been recently elucidated, and these proteins differ in their functionality from those found in animals. In this review, our current knowledge of the functioning of plant kinetochore proteins in meiosis will be summarized. In addition, the functional similarities and differences of core kinetochore proteins in meiosis between plants and other species are discussed, and the potential applications of manipulating certain kinetochore genes in meiosis for breeding purposes are explored.
Collapse
Affiliation(s)
- Kang-Di Zhou
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (K.-D.Z.); (C.-X.Z.)
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (H.-C.B.); (J.-C.D.); (H.-Y.Q.); (Y.-L.J.)
| | - Cai-Xia Zhang
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (K.-D.Z.); (C.-X.Z.)
| | - Fu-Rong Niu
- College of Forestry, Gansu Agricultural University, Lanzhou 730070, China;
| | - Hao-Chen Bai
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (H.-C.B.); (J.-C.D.); (H.-Y.Q.); (Y.-L.J.)
| | - Dan-Dan Wu
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu 611130, China;
| | - Jia-Cheng Deng
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (H.-C.B.); (J.-C.D.); (H.-Y.Q.); (Y.-L.J.)
| | - Hong-Yuan Qian
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (H.-C.B.); (J.-C.D.); (H.-Y.Q.); (Y.-L.J.)
| | - Yun-Lei Jiang
- School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (H.-C.B.); (J.-C.D.); (H.-Y.Q.); (Y.-L.J.)
| | - Wei Ma
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China; (K.-D.Z.); (C.-X.Z.)
| |
Collapse
|
63
|
Johnson BA, Liu AZ, Bi T, Dong Y, Li T, Zhou D, Narkar A, Wu Y, Sun SX, Larman TC, Zhu J, Li R. Differential effects of aneuploidy on growth and differentiation in human intestinal stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.23.559117. [PMID: 37790420 PMCID: PMC10542480 DOI: 10.1101/2023.09.23.559117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Aneuploidy, a near ubiquitous genetic feature of tumors, is a context-dependent driver of cancer evolution; however, the mechanistic basis of this role remains unclear. Here, by inducing heterogeneous aneuploidy in non-transformed human colon organoids (colonoids), we investigate how the effects of aneuploidy on cell growth and differentiation may promote malignant transformation. Single-cell RNA sequencing reveals that the gene expression signature across over 100 unique aneuploid karyotypes is enriched with p53 responsive genes. The primary driver of p53 activation is karyotype complexity. Complex aneuploid cells with multiple unbalanced chromosomes activate p53 and undergo G1 cell-cycle arrest, independent of DNA damage and without evidence of senescence. By contrast, simple aneuploid cells with 1-3 chromosomes gained or lost continue to proliferate, demonstrated by single cell tracking in colonoids. Notably, simple aneuploid cells exhibit impaired differentiation when niche factors are withdrawn. These findings suggest that while complex aneuploid cells are eliminated from the normal epithelium due to p53 activation, simple aneuploid cells can escape this checkpoint and may contribute to niche factor-independent growth of cancer-initiating cells.
Collapse
|
64
|
Uno N, Satofuka H, Miyamoto H, Honma K, Suzuki T, Yamazaki K, Ito R, Moriwaki T, Hamamichi S, Tomizuka K, Oshimura M, Kazuki Y. Treatment of CHO cells with Taxol and reversine improves micronucleation and microcell-mediated chromosome transfer efficiency. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:391-403. [PMID: 37547291 PMCID: PMC10403731 DOI: 10.1016/j.omtn.2023.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 07/11/2023] [Indexed: 08/08/2023]
Abstract
Microcell-mediated chromosome transfer is an attractive technique for transferring chromosomes from donor cells to recipient cells and has enabled the generation of cell lines and humanized animal models that contain megabase-sized gene(s). However, improvements in chromosomal transfer efficiency are still needed to accelerate the production of these cells and animals. The chromosomal transfer protocol consists of micronucleation, microcell formation, and fusion of donor cells with recipient cells. We found that the combination of Taxol (paclitaxel) and reversine rather than the conventional reagent colcemid resulted in highly efficient micronucleation and substantially improved chromosomal transfer efficiency from Chinese hamster ovary donor cells to HT1080 and NIH3T3 recipient cells by up to 18.3- and 4.9-fold, respectively. Furthermore, chromosome transfer efficiency to human induced pluripotent stem cells, which rarely occurred with colcemid, was also clearly improved after Taxol and reversine treatment. These results might be related to Taxol increasing the number of spindle poles, leading to multinucleation and delaying mitosis, and reversine inducing mitotic slippage and decreasing the duration of mitosis. Here, we demonstrated that an alternative optimized protocol improved chromosome transfer efficiency into various cell lines. These data advance chromosomal engineering technology and the use of human artificial chromosomes in genetic and regenerative medical research.
Collapse
Affiliation(s)
- Narumi Uno
- Laboratory of Bioengineering, Faculty of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Hiroyuki Satofuka
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Hitomaru Miyamoto
- Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Kazuhisa Honma
- Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Teruhiko Suzuki
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Kyotaro Yamazaki
- Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
- Chromosome Engineering Research Group, The Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - Ryota Ito
- Laboratory of Bioengineering, Faculty of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Takashi Moriwaki
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
- Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Shusei Hamamichi
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Kazuma Tomizuka
- Laboratory of Bioengineering, Faculty of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Mitsuo Oshimura
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Yasuhiro Kazuki
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
- Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
- Chromosome Engineering Research Group, The Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
65
|
Truong MA, Cané-Gasull P, Lens SMA. Modeling specific aneuploidies: from karyotype manipulations to biological insights. Chromosome Res 2023; 31:25. [PMID: 37640903 PMCID: PMC10462580 DOI: 10.1007/s10577-023-09735-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/11/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023]
Abstract
An abnormal chromosome number, or aneuploidy, underlies developmental disorders and is a common feature of cancer, with different cancer types exhibiting distinct patterns of chromosomal gains and losses. To understand how specific aneuploidies emerge in certain tissues and how they contribute to disease development, various methods have been developed to alter the karyotype of mammalian cells and mice. In this review, we provide an overview of both classic and novel strategies for inducing or selecting specific chromosomal gains and losses in human and murine cell systems. We highlight how these customized aneuploidy models helped expanding our knowledge of the consequences of specific aneuploidies to (cancer) cell physiology.
Collapse
Affiliation(s)
- My Anh Truong
- Oncode Institute and Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584, CG, Utrecht, The Netherlands
| | - Paula Cané-Gasull
- Oncode Institute and Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584, CG, Utrecht, The Netherlands
| | - Susanne M A Lens
- Oncode Institute and Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584, CG, Utrecht, The Netherlands.
| |
Collapse
|
66
|
Normandin K, Coulombe-Huntington J, St-Denis C, Bernard A, Bourouh M, Bertomeu T, Tyers M, Archambault V. Genetic enhancers of partial PLK1 inhibition reveal hypersensitivity to kinetochore perturbations. PLoS Genet 2023; 19:e1010903. [PMID: 37639469 PMCID: PMC10491399 DOI: 10.1371/journal.pgen.1010903] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/08/2023] [Accepted: 08/06/2023] [Indexed: 08/31/2023] Open
Abstract
Polo-like kinase 1 (PLK1) is a serine/threonine kinase required for mitosis and cytokinesis. As cancer cells are often hypersensitive to partial PLK1 inactivation, chemical inhibitors of PLK1 have been developed and tested in clinical trials. However, these small molecule inhibitors alone are not completely effective. PLK1 promotes numerous molecular and cellular events in the cell division cycle and it is unclear which of these events most crucially depend on PLK1 activity. We used a CRISPR-based genome-wide screening strategy to identify genes whose inactivation enhances cell proliferation defects upon partial chemical inhibition of PLK1. Genes identified encode proteins that are functionally linked to PLK1 in multiple ways, most notably factors that promote centromere and kinetochore function. Loss of the kinesin KIF18A or the outer kinetochore protein SKA1 in PLK1-compromised cells resulted in mitotic defects, activation of the spindle assembly checkpoint and nuclear reassembly defects. We also show that PLK1-dependent CENP-A loading at centromeres is extremely sensitive to partial PLK1 inhibition. Our results suggest that partial inhibition of PLK1 compromises the integrity and function of the centromere/kinetochore complex, rendering cells hypersensitive to different kinetochore perturbations. We propose that KIF18A is a promising target for combinatorial therapies with PLK1 inhibitors.
Collapse
Affiliation(s)
- Karine Normandin
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | | | - Corinne St-Denis
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Alexandre Bernard
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Mohammed Bourouh
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Thierry Bertomeu
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Mike Tyers
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Département de médecine, Université de Montréal, Montréal, Canada
| | - Vincent Archambault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Canada
| |
Collapse
|
67
|
Qiao JY, Zhou Q, Xu K, Yue W, Lei WL, Li YY, Gu LJ, Ouyang YC, Hou Y, Schatten H, Meng TG, Wang ZB, Sun QY. Mad2 is dispensable for accurate chromosome segregation but becomes essential when oocytes are subjected to environmental stress. Development 2023; 150:dev201398. [PMID: 37485540 DOI: 10.1242/dev.201398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 06/20/2023] [Indexed: 07/25/2023]
Abstract
Accurate chromosome segregation, monitored by the spindle assembly checkpoint (SAC), is crucial for the production of euploid cells. Previous in vitro studies by us and others showed that Mad2, a core member of the SAC, performs a checkpoint function in oocyte meiosis. Here, through an oocyte-specific knockout approach in mouse, we reconfirmed that Mad2-deficient oocytes exhibit an accelerated metaphase-to-anaphase transition caused by premature degradation of securin and cyclin B1 and subsequent activation of separase in meiosis I. However, it was surprising that the knockout mice were completely fertile and the resulting oocytes were euploid. In the absence of Mad2, other SAC proteins, including BubR1, Bub3 and Mad1, were normally recruited to the kinetochores, which likely explains the balanced chromosome separation. Further studies showed that the chromosome separation in Mad2-null oocytes was particularly sensitive to environmental changes and, when matured in vitro, showed chromosome misalignment, lagging chromosomes, and aneuploidy with premature separation of sister chromatids, which was exacerbated at a lower temperature. We reveal for the first time that Mad2 is dispensable for proper chromosome segregation but acts to mitigate environmental stress in meiotic oocytes.
Collapse
Affiliation(s)
- Jing-Yi Qiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Zhou
- Department of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Ke Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Yue
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wen-Long Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuan-Yuan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lin-Jian Gu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Tie-Gang Meng
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qing-Yuan Sun
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, China
| |
Collapse
|
68
|
Agustinus AS, Al-Rawi D, Dameracharla B, Raviram R, Jones BSCL, Stransky S, Scipioni L, Luebeck J, Di Bona M, Norkunaite D, Myers RM, Duran M, Choi S, Weigelt B, Yomtoubian S, McPherson A, Toufektchan E, Keuper K, Mischel PS, Mittal V, Shah SP, Maciejowski J, Storchova Z, Gratton E, Ly P, Landau D, Bakhoum MF, Koche RP, Sidoli S, Bafna V, David Y, Bakhoum SF. Epigenetic dysregulation from chromosomal transit in micronuclei. Nature 2023; 619:176-183. [PMID: 37286593 PMCID: PMC10322720 DOI: 10.1038/s41586-023-06084-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/14/2023] [Indexed: 06/09/2023]
Abstract
Chromosomal instability (CIN) and epigenetic alterations are characteristics of advanced and metastatic cancers1-4, but whether they are mechanistically linked is unknown. Here we show that missegregation of mitotic chromosomes, their sequestration in micronuclei5,6 and subsequent rupture of the micronuclear envelope7 profoundly disrupt normal histone post-translational modifications (PTMs), a phenomenon conserved across humans and mice, as well as in cancer and non-transformed cells. Some of the changes in histone PTMs occur because of the rupture of the micronuclear envelope, whereas others are inherited from mitotic abnormalities before the micronucleus is formed. Using orthogonal approaches, we demonstrate that micronuclei exhibit extensive differences in chromatin accessibility, with a strong positional bias between promoters and distal or intergenic regions, in line with observed redistributions of histone PTMs. Inducing CIN causes widespread epigenetic dysregulation, and chromosomes that transit in micronuclei experience heritable abnormalities in their accessibility long after they have been reincorporated into the primary nucleus. Thus, as well as altering genomic copy number, CIN promotes epigenetic reprogramming and heterogeneity in cancer.
Collapse
Affiliation(s)
- Albert S Agustinus
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pharmacology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Duaa Al-Rawi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bhargavi Dameracharla
- Department of Computer Science, University of California, San Diego, La Jolla, CA, USA
| | | | - Bailey S C L Jones
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie Stransky
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
| | - Lorenzo Scipioni
- School of Engineering, University of California, Irvine, Irvine, CA, USA
| | - Jens Luebeck
- Department of Computer Science, University of California, San Diego, La Jolla, CA, USA
| | - Melody Di Bona
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Danguole Norkunaite
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert M Myers
- New York Genome Center, New York, NY, USA
- Tri-institutional MD-PhD Program, New York, NY, USA
| | - Mercedes Duran
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Seongmin Choi
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shira Yomtoubian
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Andrew McPherson
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eléonore Toufektchan
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kristina Keuper
- Department of Molecular Genetics, University of Kaiserslautern, Kaiserslautern, Germany
| | - Paul S Mischel
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Vivek Mittal
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, USA
| | - Sohrab P Shah
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John Maciejowski
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zuzana Storchova
- Department of Molecular Genetics, University of Kaiserslautern, Kaiserslautern, Germany
| | - Enrico Gratton
- School of Engineering, University of California, Irvine, Irvine, CA, USA
| | - Peter Ly
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dan Landau
- New York Genome Center, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Mathieu F Bakhoum
- Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
| | - Vineet Bafna
- Department of Computer Science, University of California, San Diego, La Jolla, CA, USA
| | - Yael David
- Pharmacology Graduate Program, Weill Cornell Medicine, New York, NY, USA.
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Tri-institutional PhD Program in Chemical Biology, New York, NY, USA.
| | - Samuel F Bakhoum
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
69
|
Ide AH, DeLuca KF, Wiggan O, Markus SM, DeLuca JG. The role of kinetochore dynein in checkpoint silencing is restricted to disassembly of the corona. Mol Biol Cell 2023; 34:ar76. [PMID: 37126397 PMCID: PMC10295480 DOI: 10.1091/mbc.e23-04-0130] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 05/02/2023] Open
Abstract
During mitosis, kinetochore-microtubule attachments are monitored by a molecular surveillance system known as the spindle assembly checkpoint. The prevailing model posits that dynein evicts checkpoint proteins (e.g., Mad1, Mad2) from stably attached kinetochores by transporting them away from kinetochores, thus contributing to checkpoint silencing. However, the mechanism by which dynein performs this function, and its precise role in checkpoint silencing remain unresolved. Here, we find that dynein's role in checkpoint silencing is restricted to evicting checkpoint effectors from the fibrous corona, and not the outer kinetochore. Dynein evicts these molecules from the corona in a manner that does not require stable, end-on microtubule attachments. Thus, by disassembling the corona through indiscriminate microtubule encounters, dynein primes the checkpoint signaling apparatus so it can respond to stable end-on microtubule attachments and permit cells to progress through mitosis. Accordingly, we find that dynein function in checkpoint silencing becomes largely dispensable in cells in which checkpoint effectors are excluded from the corona.
Collapse
Affiliation(s)
- Amy H. Ide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523
| | - Keith F. DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523
| | - O’Neil Wiggan
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523
| | - Steven M. Markus
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523
| | - Jennifer G. DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523
| |
Collapse
|
70
|
Hayes BH, Zhu PK, Wang M, Pfeifer CR, Xia Y, Phan S, Andrechak JC, Du J, Tobin MP, Anlas A, Dooling LJ, Vashisth M, Irianto J, Lampson MA, Discher DE. Confinement plus myosin-II suppression maximizes heritable loss of chromosomes, as revealed by live-cell ChReporters. J Cell Sci 2023; 136:jcs260753. [PMID: 37288769 PMCID: PMC10309578 DOI: 10.1242/jcs.260753] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 04/27/2023] [Indexed: 05/10/2023] Open
Abstract
The mechanical environment of a cell can have many effects, but whether it impacts the DNA sequence of a cell has remained unexamined. To investigate this, we developed a live-cell method to measure changes in chromosome numbers. We edited constitutive genes with GFP or RFP tags on single alleles and discovered that cells that lose Chromosome reporters (ChReporters) become non-fluorescent. We applied our new tools to confined mitosis and to inhibition of the putative tumor suppressor myosin-II. We quantified compression of mitotic chromatin in vivo and demonstrated that similar compression in vitro resulted in cell death, but also rare and heritable ChReptorter loss. Myosin-II suppression rescued lethal multipolar divisions and maximized ChReporter loss during three-dimensional (3D) compression and two-dimensional (2D) lateral confinement, but not in standard 2D culture. ChReporter loss was associated with chromosome mis-segregation, rather than just the number of divisions, and loss in vitro and in mice was selected against in subsequent 2D cultures. Inhibition of the spindle assembly checkpoint (SAC) caused ChReporter loss in 2D culture, as expected, but not during 3D compression, suggesting a SAC perturbation. Thus, ChReporters enable diverse studies of viable genetic changes, and show that confinement and myosin-II affect DNA sequence and mechano-evolution.
Collapse
Affiliation(s)
- Brandon H. Hayes
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter Kuangzheng Zhu
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mai Wang
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charlotte R. Pfeifer
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yuntao Xia
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven Phan
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason C. Andrechak
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junhong Du
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael P. Tobin
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alisya Anlas
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lawrence J. Dooling
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Manasvita Vashisth
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jerome Irianto
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael A. Lampson
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dennis E. Discher
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
71
|
Bosco N, Goldberg A, Zhao X, Mays JC, Cheng P, Johnson AF, Bianchi JJ, Toscani C, Di Tommaso E, Katsnelson L, Annuar D, Mei S, Faitelson RE, Pesselev IY, Mohamed KS, Mermerian A, Camacho-Hernandez EM, Gionco CA, Manikas J, Tseng YS, Sun Z, Fani S, Keegan S, Lippman SM, Fenyö D, Giunta S, Santaguida S, Davoli T. KaryoCreate: A CRISPR-based technology to study chromosome-specific aneuploidy by targeting human centromeres. Cell 2023; 186:1985-2001.e19. [PMID: 37075754 PMCID: PMC10676289 DOI: 10.1016/j.cell.2023.03.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/17/2022] [Accepted: 03/23/2023] [Indexed: 04/21/2023]
Abstract
Aneuploidy, the presence of chromosome gains or losses, is a hallmark of cancer. Here, we describe KaryoCreate (karyotype CRISPR-engineered aneuploidy technology), a system that enables the generation of chromosome-specific aneuploidies by co-expression of an sgRNA targeting chromosome-specific CENPA-binding ɑ-satellite repeats together with dCas9 fused to mutant KNL1. We design unique and highly specific sgRNAs for 19 of the 24 chromosomes. Expression of these constructs leads to missegregation and induction of gains or losses of the targeted chromosome in cellular progeny, with an average efficiency of 8% for gains and 12% for losses (up to 20%) validated across 10 chromosomes. Using KaryoCreate in colon epithelial cells, we show that chromosome 18q loss, frequent in gastrointestinal cancers, promotes resistance to TGF-β, likely due to synergistic hemizygous deletion of multiple genes. Altogether, we describe an innovative technology to create and study chromosome missegregation and aneuploidy in the context of cancer and beyond.
Collapse
Affiliation(s)
- Nazario Bosco
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Aleah Goldberg
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Xin Zhao
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Joseph C Mays
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Pan Cheng
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Adam F Johnson
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Joy J Bianchi
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Cecilia Toscani
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Elena Di Tommaso
- Department of Biology and Biotechnology Charles Darwin, University of Rome "La Sapienza", 00185 Rome, Italy
| | - Lizabeth Katsnelson
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Dania Annuar
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Sally Mei
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Roni E Faitelson
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Ilan Y Pesselev
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Kareem S Mohamed
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Angela Mermerian
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Elaine M Camacho-Hernandez
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Courtney A Gionco
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Julie Manikas
- Department of Cell Biology, NYU Langone Health, New York, NY, USA
| | - Yi-Shuan Tseng
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Zhengxi Sun
- Department of Pathology and Laura & Isaac Perlmutter Cancer Center, NYU School of Medicine, New York, NY, USA
| | - Somayeh Fani
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Sarah Keegan
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Scott M Lippman
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093, USA
| | - David Fenyö
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA
| | - Simona Giunta
- Department of Biology and Biotechnology Charles Darwin, University of Rome "La Sapienza", 00185 Rome, Italy
| | - Stefano Santaguida
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| | - Teresa Davoli
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY, USA.
| |
Collapse
|
72
|
Cmentowski V, Ciossani G, d’Amico E, Wohlgemuth S, Owa M, Dynlacht B, Musacchio A. A mechanism that integrates microtubule motors of opposite polarity at the kinetochore corona. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.538277. [PMID: 37163019 PMCID: PMC10168246 DOI: 10.1101/2023.04.25.538277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Chromosome biorientation on the mitotic spindle is prerequisite to errorless genome inheritance. CENP-E (kinesin 7) and Dynein-Dynactin (DD), microtubule motors with opposite polarity, promote biorientation from the kinetochore corona, a polymeric structure whose assembly requires MPS1 kinase. The corona's building block consists of ROD, Zwilch, ZW10, and the DD adaptor Spindly (RZZS). How CENP-E and DD are scaffolded and mutually coordinated in the corona remains unclear. Here, we report near-complete depletion of RZZS and DD from kinetochores after depletion of CENP-E and the outer kinetochore protein KNL1. With inhibited MPS1, CENP-E, which we show binds directly to RZZS, is required to retain kinetochore RZZS. An RZZS phosphomimetic mutant bypasses this requirement. With active MPS1, CENP-E is dispensable for corona expansion, but strictly required for physiological kinetochore accumulation of DD. Thus, we identify the corona as an integrated scaffold where CENP-E kinesin controls DD kinetochore loading for coordinated bidirectional transport of chromosome cargo.
Collapse
Affiliation(s)
- Verena Cmentowski
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Giuseppe Ciossani
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Ennio d’Amico
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Sabine Wohlgemuth
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
| | - Mikito Owa
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY, USA
| | - Brian Dynlacht
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY, USA
| | - Andrea Musacchio
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227 Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
73
|
Jiang H, Kong N, Liu Z, West SC, Chan YW. Human Endonuclease ANKLE1 Localizes at the Midbody and Processes Chromatin Bridges to Prevent DNA Damage and cGAS-STING Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204388. [PMID: 36825683 PMCID: PMC10131833 DOI: 10.1002/advs.202204388] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/12/2022] [Indexed: 06/18/2023]
Abstract
Chromatin bridges connecting the two segregating daughter nuclei arise from chromosome fusion or unresolved interchromosomal linkage. Persistent chromatin bridges are trapped in the cleavage plane, triggering cytokinesis delay. The trapped bridges occasionally break during cytokinesis, inducing DNA damage and chromosomal rearrangements. Recently, Caenorhabditis elegans LEM-3 and human TREX1 nucleases have been shown to process chromatin bridges. Here, it is shown that ANKLE1 endonuclease, the human ortholog of LEM-3, accumulates at the bulge-like structure of the midbody via its N-terminal ankyrin repeats. Importantly, ANKLE1-/- knockout cells display an elevated level of G1-specific 53BP1 nuclear bodies, prolonged activation of the DNA damage response, and replication stress. Increased DNA damage observed in ANKLE1-/- cells is rescued by inhibiting actin polymerization or reducing actomyosin contractility. ANKLE1 does not act in conjunction with structure-selective endonucleases, GEN1 and MUS81 in resolving recombination intermediates. Instead, ANKLE1 acts on chromatin bridges by priming TREX1 nucleolytic activity and cleaving bridge DNA to prevent the formation of micronuclei and cytosolic dsDNA that activate the cGAS-STING pathway. It is therefore proposed that ANKLE1 prevents DNA damage and autoimmunity by cleaving chromatin bridges to avoid catastrophic breakage mediated by actomyosin contractile forces.
Collapse
Affiliation(s)
- Huadong Jiang
- School of Biological SciencesThe University of Hong KongPokfulamHong Kong
| | - Nannan Kong
- School of Biological SciencesThe University of Hong KongPokfulamHong Kong
| | - Zeyuan Liu
- School of Biological SciencesThe University of Hong KongPokfulamHong Kong
| | - Stephen C. West
- The Francis Crick InstituteDNA Recombination and Repair Laboratory1 Midland RoadLondonNW1 1ATUK
| | - Ying Wai Chan
- School of Biological SciencesThe University of Hong KongPokfulamHong Kong
| |
Collapse
|
74
|
Zhou Q, Xu K, Zhao BW, Qiao JY, Li YY, Lei WL, Li J, Ouyang YC, Hou Y, Schatten H, Wang ZB, Sun QY. Mitochondrial E3 ubiquitin ligase MARCH5 is required for mouse oocyte meiotic maturation†. Biol Reprod 2023; 108:437-446. [PMID: 36503987 DOI: 10.1093/biolre/ioac215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 07/26/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
As the most abundant organelles in oocytes, mitochondria play an important role in maintaining oocyte quality. Here, we report that March5, encoding a mitochondrial ubiquitin ligase that promotes mitochondrial elongation, plays a critical role in mouse oocyte meiotic maturation via regulating mitochondrial function. The subcellular localization of MARCH5 was similar to the mitochondrial distribution during mouse oocyte meiotic progression. Knockdown of March5 caused decreased ratios of the first polar body extrusion. March5-siRNA injection resulted in oocyte mitochondrial dysfunctions, manifested by increased reactive oxygen species, decreased ATP content as well as decreased mitochondrial membrane potential, leading to reduced ability of spindle formation and an increased ratio of kinetochore-microtubule detachment. Further study showed that the continuous activation of the spindle assembly checkpoint and the failure of Cyclin B1 degradation caused MI arrest and first polar body (PB1) extrusion failure in March5 knockdown oocytes. Taken together, our results demonstrated that March5 plays an essential role in mouse oocyte meiotic maturation, possibly via regulation of mitochondrial function and/or ubiquitination of microtubule dynamics- or cell cycle-regulating proteins.
Collapse
Affiliation(s)
- Qian Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- International Cancer Center, Shenzhen University Medical School, Shenzhen, China
- Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ke Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Bing-Wang Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jing-Yi Qiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yuan-Yuan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wen-Long Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jian Li
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
75
|
Garribba L, De Feudis G, Martis V, Galli M, Dumont M, Eliezer Y, Wardenaar R, Ippolito MR, Iyer DR, Tijhuis AE, Spierings DCJ, Schubert M, Taglietti S, Soriani C, Gemble S, Basto R, Rhind N, Foijer F, Ben-David U, Fachinetti D, Doksani Y, Santaguida S. Short-term molecular consequences of chromosome mis-segregation for genome stability. Nat Commun 2023; 14:1353. [PMID: 36906648 PMCID: PMC10008630 DOI: 10.1038/s41467-023-37095-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 03/01/2023] [Indexed: 03/13/2023] Open
Abstract
Chromosome instability (CIN) is the most common form of genome instability and is a hallmark of cancer. CIN invariably leads to aneuploidy, a state of karyotype imbalance. Here, we show that aneuploidy can also trigger CIN. We found that aneuploid cells experience DNA replication stress in their first S-phase and precipitate in a state of continuous CIN. This generates a repertoire of genetically diverse cells with structural chromosomal abnormalities that can either continue proliferating or stop dividing. Cycling aneuploid cells display lower karyotype complexity compared to the arrested ones and increased expression of DNA repair signatures. Interestingly, the same signatures are upregulated in highly-proliferative cancer cells, which might enable them to proliferate despite the disadvantage conferred by aneuploidy-induced CIN. Altogether, our study reveals the short-term origins of CIN following aneuploidy and indicates the aneuploid state of cancer cells as a point mutation-independent source of genome instability, providing an explanation for aneuploidy occurrence in tumors.
Collapse
Affiliation(s)
- Lorenza Garribba
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Giuseppina De Feudis
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Valentino Martis
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Martina Galli
- IFOM ETS - The AIRC Institute of Molecular Oncology, via Adamello 16, 20139, Milan, Italy
| | - Marie Dumont
- Institut Curie, PSL Research University, CNRS, UMR144, Paris, France
| | - Yonatan Eliezer
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - René Wardenaar
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, the Netherlands
| | - Marica Rosaria Ippolito
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Divya Ramalingam Iyer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA, 01605, USA
| | - Andréa E Tijhuis
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, the Netherlands
| | - Diana C J Spierings
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, the Netherlands
| | - Michael Schubert
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, the Netherlands
| | - Silvia Taglietti
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Chiara Soriani
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy
| | - Simon Gemble
- Institut Curie, PSL Research University, CNRS, UMR144, Paris, France
| | - Renata Basto
- Institut Curie, PSL Research University, CNRS, UMR144, Paris, France
| | - Nick Rhind
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, 364 Plantation Street, Worcester, MA, 01605, USA
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, the Netherlands
| | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Ylli Doksani
- IFOM ETS - The AIRC Institute of Molecular Oncology, via Adamello 16, 20139, Milan, Italy
| | - Stefano Santaguida
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Via Adamello 16, 20139, Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9/1, 20122, Milan, Italy.
| |
Collapse
|
76
|
Adell MAY, Klockner TC, Höfler R, Wallner L, Schmid J, Markovic A, Martyniak A, Campbell CS. Adaptation to spindle assembly checkpoint inhibition through the selection of specific aneuploidies. Genes Dev 2023; 37:171-190. [PMID: 36859339 PMCID: PMC10111865 DOI: 10.1101/gad.350182.122] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/30/2023] [Indexed: 03/03/2023]
Abstract
Both the presence of an abnormal complement of chromosomes (aneuploidy) and an increased frequency of chromosome missegregation (chromosomal instability) are hallmarks of cancer. Analyses of cancer genome data have identified certain aneuploidy patterns in tumors; however, the bases behind their selection are largely unexplored. By establishing time-resolved long-term adaptation protocols, we found that human cells adapt to persistent spindle assembly checkpoint (SAC) inhibition by acquiring specific chromosome arm gains and losses. Independently adapted populations converge on complex karyotypes, which over time are refined to contain ever smaller chromosomal changes. Of note, the frequencies of chromosome arm gains in adapted cells correlate with those detected in cancers, suggesting that our cellular adaptation approach recapitulates selective traits that dictate the selection of aneuploidies frequently observed across many cancer types. We further engineered specific aneuploidies to determine the genetic basis behind the observed karyotype patterns. These experiments demonstrated that the adapted and engineered aneuploid cell lines limit CIN by extending mitotic duration. Heterozygous deletions of key SAC and APC/C genes recapitulated the rescue phenotypes of the monosomic chromosomes. We conclude that aneuploidy-induced gene dosage imbalances of individual mitotic regulators are sufficient for altering mitotic timing to reduce CIN.
Collapse
Affiliation(s)
- Manuel Alonso Y Adell
- Department of Chromosome Biology, Max Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| | - Tamara C Klockner
- Department of Chromosome Biology, Max Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| | - Rudolf Höfler
- Department of Chromosome Biology, Max Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| | - Lea Wallner
- Department of Chromosome Biology, Max Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| | - Julia Schmid
- Department of Chromosome Biology, Max Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| | - Ana Markovic
- Department of Chromosome Biology, Max Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| | - Anastasiia Martyniak
- Department of Chromosome Biology, Max Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| | - Christopher S Campbell
- Department of Chromosome Biology, Max Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), A-1030 Vienna, Austria
| |
Collapse
|
77
|
Lambuta RA, Nanni L, Liu Y, Diaz-Miyar J, Iyer A, Tavernari D, Katanayeva N, Ciriello G, Oricchio E. Whole-genome doubling drives oncogenic loss of chromatin segregation. Nature 2023; 615:925-933. [PMID: 36922594 PMCID: PMC10060163 DOI: 10.1038/s41586-023-05794-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 02/03/2023] [Indexed: 03/17/2023]
Abstract
Whole-genome doubling (WGD) is a recurrent event in human cancers and it promotes chromosomal instability and acquisition of aneuploidies1-8. However, the three-dimensional organization of chromatin in WGD cells and its contribution to oncogenic phenotypes are currently unknown. Here we show that in p53-deficient cells, WGD induces loss of chromatin segregation (LCS). This event is characterized by reduced segregation between short and long chromosomes, A and B subcompartments and adjacent chromatin domains. LCS is driven by the downregulation of CTCF and H3K9me3 in cells that bypassed activation of the tetraploid checkpoint. Longitudinal analyses revealed that LCS primes genomic regions for subcompartment repositioning in WGD cells. This results in chromatin and epigenetic changes associated with oncogene activation in tumours ensuing from WGD cells. Notably, subcompartment repositioning events were largely independent of chromosomal alterations, which indicates that these were complementary mechanisms contributing to tumour development and progression. Overall, LCS initiates chromatin conformation changes that ultimately result in oncogenic epigenetic and transcriptional modifications, which suggests that chromatin evolution is a hallmark of WGD-driven cancer.
Collapse
Affiliation(s)
- Ruxandra A Lambuta
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Écublens, Switzerland
- Swiss Cancer Center Leman, Lausanne, Switzerland
| | - Luca Nanni
- Swiss Cancer Center Leman, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Yuanlong Liu
- Swiss Cancer Center Leman, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Juan Diaz-Miyar
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Écublens, Switzerland
- Swiss Cancer Center Leman, Lausanne, Switzerland
| | - Arvind Iyer
- Swiss Cancer Center Leman, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Daniele Tavernari
- Swiss Cancer Center Leman, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Natalya Katanayeva
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Écublens, Switzerland
- Swiss Cancer Center Leman, Lausanne, Switzerland
| | - Giovanni Ciriello
- Swiss Cancer Center Leman, Lausanne, Switzerland.
- Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland.
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland.
| | - Elisa Oricchio
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Écublens, Switzerland.
- Swiss Cancer Center Leman, Lausanne, Switzerland.
| |
Collapse
|
78
|
Kong N, Liu Z, Chan YW. RIF1 suppresses the formation of single-stranded ultrafine anaphase bridges via protein phosphatase 1. Cell Rep 2023; 42:112032. [PMID: 36719798 DOI: 10.1016/j.celrep.2023.112032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/07/2022] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
Resolution of ultrafine anaphase bridges (UFBs) must be completed before cytokinesis to ensure sister-chromatid disjunction. RIF1 is involved in UFB resolution by a mechanism that is not yet clear. Here, we show that RIF1 functions in mitosis to inhibit the formation of 53BP1 nuclear bodies and micronuclei. Meanwhile, RIF1 localizes on PICH-coated double-stranded UFBs but not on RPA-coated single-stranded UFBs. Depletion of RIF1 leads to an elevated level of RPA-coated UFBs, in a BLM-dependent manner. RIF1 interacts with all three isoforms of protein phosphatase 1 (PP1) at its CI domain in anaphase when CDK1 activity declines. CDK1 negatively regulates RIF1-PP1 interaction via the CIII domain of RIF1. Importantly, depletion of PP1 phenocopies RIF1 depletion, and phosphorylation-resistant mutant of PICH shows reduced interaction with the BTR complex and bypasses the need of RIF1 in preventing the formation of single-stranded UFBs. Overall, our data show that PP1 is the effector of RIF1 in UFB resolution.
Collapse
Affiliation(s)
- Nannan Kong
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Zeyuan Liu
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Ying Wai Chan
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China.
| |
Collapse
|
79
|
Partscht P, Simon A, Chen NP, Erhardt S, Schiebel E. The HIPK2/CDC14B-MeCP2 axis enhances the spindle assembly checkpoint block by promoting cyclin B translation. SCIENCE ADVANCES 2023; 9:eadd6982. [PMID: 36662865 PMCID: PMC9858502 DOI: 10.1126/sciadv.add6982] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 12/16/2022] [Indexed: 05/12/2023]
Abstract
Mitotic perturbations activate the spindle assembly checkpoint (SAC) that keeps cells in prometaphase with high CDK1 activity. Prolonged mitotic arrest is eventually bypassed by gradual cyclin B decline followed by slippage of cells into G1 without chromosome segregation, a process that promotes cell transformation and drug resistance. Hitherto, the cyclin B1 decay is exclusively defined by mechanisms that involve its proteasomal degradation. Here, we report that hyperphosphorylated HIPK2 kinase accumulates in mitotic cells and phosphorylates the Rett syndrome protein MeCP2 at Ser92, a regulation that is counteracted by CDC14B phosphatase. MeCP2S92 phosphorylation leads to the enhanced translation of cyclin B1, which is important for cells with persistent SAC activation to counteract the proteolytic decline of cyclin B1 and therefore to suspend mitotic slippage. Hence, the HIPK2/CDC14B-MeCP2 axis functions as an enhancer of the SAC-induced mitotic block. Collectively, our study revises the prevailing view of how cells confer a sustainable SAC.
Collapse
Affiliation(s)
- Patrick Partscht
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg 69120, Germany
- Heidelberg Biosciences International Graduate School (HBIGS), Universität Heidelberg, Heidelberg, Germany
| | - Alexander Simon
- Heidelberg Biosciences International Graduate School (HBIGS), Universität Heidelberg, Heidelberg, Germany
- Zoological Institute, Karlsruhe Institute of Technology (KIT), Karlsruhe 76131, Germany
| | - Nan-Peng Chen
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg 69120, Germany
| | - Sylvia Erhardt
- Zoological Institute, Karlsruhe Institute of Technology (KIT), Karlsruhe 76131, Germany
| | - Elmar Schiebel
- Zentrum für Molekulare Biologie, Universität Heidelberg, DKFZ-ZMBH Allianz, Heidelberg 69120, Germany
| |
Collapse
|
80
|
Roca M, Besnardeau L, Christians E, McDougall A, Chenevert J, Castagnetti S. Acquisition of the spindle assembly checkpoint and its modulation by cell fate and cell size in a chordate embryo. Development 2023; 150:285941. [PMID: 36515557 DOI: 10.1242/dev.201145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022]
Abstract
The spindle assembly checkpoint (SAC) is a surveillance system that preserves genome integrity by delaying anaphase onset until all chromosomes are correctly attached to spindle microtubules. Recruitment of SAC proteins to unattached kinetochores generates an inhibitory signal that prolongs mitotic duration. Chordate embryos are atypical in that spindle defects do not delay mitotic progression during early development, implying that either the SAC is inactive or the cell-cycle target machinery is unresponsive. Here, we show that in embryos of the chordate Phallusia mammillata, the SAC delays mitotic progression from the 8th cleavage divisions. Unattached kinetochores are not recognized by the SAC machinery until the 7th cell cycle, when the SAC is acquired. After acquisition, SAC strength, which manifests as the degree of mitotic lengthening induced by spindle perturbations, is specific to different cell types and is modulated by cell size, showing similarity to SAC control in early Caenorhabditis elegans embryos. We conclude that SAC acquisition is a process that is likely specific to chordate embryos, while modulation of SAC efficiency in SAC proficient stages depends on cell fate and cell size, which is similar to non-chordate embryos.
Collapse
Affiliation(s)
- Marianne Roca
- Sorbonne Universités, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (LBDV), 06230 Villefranche-sur-Mer, France
| | - Lydia Besnardeau
- Sorbonne Universités, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (LBDV), 06230 Villefranche-sur-Mer, France
| | - Elisabeth Christians
- Sorbonne Universités, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (LBDV), 06230 Villefranche-sur-Mer, France
| | - Alex McDougall
- Sorbonne Universités, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (LBDV), 06230 Villefranche-sur-Mer, France
| | - Janet Chenevert
- Sorbonne Universités, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (LBDV), 06230 Villefranche-sur-Mer, France
| | - Stefania Castagnetti
- Sorbonne Universités, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-Mer (LBDV), 06230 Villefranche-sur-Mer, France
| |
Collapse
|
81
|
Tucker JB, Bonema SC, García-Varela R, Denu RA, Hu Y, McGregor SM, Burkard ME, Weaver BA. Misaligned Chromosomes are a Major Source of Chromosomal Instability in Breast Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:54-65. [PMID: 36968230 PMCID: PMC10035514 DOI: 10.1158/2767-9764.crc-22-0302] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/17/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
Chromosomal instability (CIN), the persistent reshuffling of chromosomes during mitosis, is a hallmark of human cancers that contributes to tumor heterogeneity and has been implicated in driving metastasis and altering responses to therapy. Though multiple mechanisms can produce CIN, lagging chromosomes generated from abnormal merotelic attachments are the major cause of CIN in a variety of cell lines, and are expected to predominate in cancer. Here, we quantify CIN in breast cancer using a tumor microarray, matched primary and metastatic samples, and patient-derived organoids from primary breast cancer. Surprisingly, misaligned chromosomes are more common than lagging chromosomes and represent a major source of CIN in primary and metastatic tumors. This feature of breast cancers is conserved in a majority of breast cancer cell lines. Importantly, though a portion of misaligned chromosomes align before anaphase onset, the fraction that remain represents the largest source of CIN in these cells. Metastatic breast cancers exhibit higher rates of CIN than matched primary cancers, primarily due to increases in misaligned chromosomes. Whether CIN causes immune activation or evasion is controversial. We find that misaligned chromosomes result in immune-activating micronuclei substantially less frequently than lagging and bridge chromosomes and that breast cancers with greater frequencies of lagging chromosomes and chromosome bridges recruit more stromal tumor-infiltrating lymphocytes. These data indicate misaligned chromosomes represent a major mechanism of CIN in breast cancer and provide support for differential immunostimulatory effects of specific types of CIN. Significance We surveyed the single-cell landscape of mitotic defects that generate CIN in primary and metastatic breast cancer and relevant models. Misaligned chromosomes predominate, and are less immunostimulatory than other chromosome segregation errors.
Collapse
Affiliation(s)
- John B. Tucker
- Cancer Biology Graduate Training Program, University of Wisconsin–Madison, Madison, Wisconsin
| | - Sarah C. Bonema
- Molecular and Cellular Pharmacology Graduate Training Program, University of Wisconsin–Madison, Madison, Wisconsin
| | | | - Ryan A. Denu
- Medical Scientist Training Program, University of Wisconsin–Madison, Madison, Wisconsin
| | - Yang Hu
- Medical Scientist Training Program, University of Wisconsin–Madison, Madison, Wisconsin
| | - Stephanie M. McGregor
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin
| | - Mark E. Burkard
- Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin
- Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
| | - Beth A. Weaver
- Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin
- Carbone Cancer Center, University of Wisconsin–Madison, Madison, Wisconsin
- Department of Cell and Regenerative Biology, University of Wisconsin–Madison, Madison, Wisconsin
| |
Collapse
|
82
|
Fischer ES. Kinetochore‐catalyzed MCC
formation: A structural perspective. IUBMB Life 2022; 75:289-310. [PMID: 36518060 DOI: 10.1002/iub.2697] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/08/2022] [Indexed: 12/23/2022]
Abstract
The spindle assembly checkpoint (SAC) is a cellular surveillance mechanism that functions to ensure accurate chromosome segregation during mitosis. Macromolecular complexes known as kinetochores, act as the interface of sister chromatid attachment to spindle microtubules. In response to unattached kinetochores, the SAC activates its effector, the mitotic checkpoint complex (MCC), which delays mitotic exit until all sister chromatid pairs have achieved successful attachment to the bipolar mitotic spindle. Formation of the MCC (composed of Mad2, BubR1, Bub3 and Cdc20) is regulated by an Mps1 kinase-dependent phosphorylation signaling cascade which assembles and repositions components of the MCC onto a catalytic scaffold. This scaffold functions to catalyze the conversion of the HORMA-domain protein Mad2 from an "inactive" open-state (O-Mad2) into an "active" closed-Mad2 (C-Mad2), and simultaneous Cdc20 binding. Here, our current understanding of the molecular mechanisms underlying the kinetic barrier to C-Mad2:Cdc20 formation will be reviewed. Recent progress in elucidating the precise molecular choreography orchestrated by the catalytic scaffold to rapidly assemble the MCC will be examined, and unresolved questions will be highlighted. Ultimately, understanding how the SAC rapidly activates the checkpoint not only provides insights into how cells maintain genomic integrity during mitosis, but also provides a paradigm for how cells can utilize molecular switches, including other HORMA domain-containing proteins, to make rapid changes to a cell's physiological state.
Collapse
Affiliation(s)
- Elyse S. Fischer
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus Cambridge UK
| |
Collapse
|
83
|
Hayward D, Roberts E, Gruneberg U. MPS1 localizes to end-on microtubule-attached kinetochores to promote microtubule release. Curr Biol 2022; 32:5200-5208.e8. [PMID: 36395767 DOI: 10.1016/j.cub.2022.10.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/07/2022] [Accepted: 10/20/2022] [Indexed: 11/17/2022]
Abstract
In eukaryotes, the spindle assembly checkpoint protects genome stability in mitosis by preventing chromosome segregation until incorrect microtubule-kinetochore attachment geometries have been eliminated and chromosome biorientation has been completed. These error correction and checkpoint processes are linked by the conserved Aurora B and MPS1 Ser/Thr kinases.1,2 MPS1-dependent checkpoint signaling is believed to be initiated by kinetochores without end-on microtubule attachments,3,4 including those generated by Aurora B-mediated error correction. The current model posits that MPS1 competes with microtubules for binding sites at the kinetochore.3,4 MPS1 is thought to first recognize kinetochores not blocked by microtubules and then initiate checkpoint signaling. However, MPS1 is also required for chromosome biorientation and correction of microtubule-kinetochore attachment errors.5,6,7,8,9 This latter function, which must require direct interaction with microtubule-attached kinetochores, is not readily explained within the constraints of the current model. Here, we show that MPS1 transiently localizes to end-on attached kinetochores and that this recruitment depends on the relative activities of Aurora B and its counteracting phosphatase PP2A-B56 rather than microtubule-attachment state per se. MPS1 autophosphorylation also regulates MPS1 kinetochore levels but does not determine the response to microtubule attachment. At end-on attached kinetochores, MPS1 actively promotes microtubule release together with Aurora B. Furthermore, in live cells, MPS1 is detected at attached kinetochores before the removal of microtubules. During chromosome alignment, MPS1, therefore, coordinates both the resolution of incorrect microtubule-kinetochore attachments and the initiation of spindle checkpoint signaling.
Collapse
Affiliation(s)
- Daniel Hayward
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, Oxfordshire OX1 3RE, UK
| | - Emile Roberts
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, Oxfordshire OX1 3RE, UK
| | - Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, Oxfordshire OX1 3RE, UK.
| |
Collapse
|
84
|
Lin YF, Hu Q, Guyer A, Fachinetti D, Ly P. Induction of chromosome-specific micronuclei and chromothripsis by centromere inactivation. Methods Cell Biol 2022; 182:1-20. [PMID: 38359973 PMCID: PMC11008423 DOI: 10.1016/bs.mcb.2022.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Chromothripsis describes the catastrophic fragmentation of individual chromosomes followed by its haphazard reassembly into a derivative chromosome harboring complex rearrangements. This process can be initiated by mitotic cell division errors when one or more chromosomes aberrantly mis-segregate into micronuclei and acquire extensive DNA damage. Approaches to induce the formation of micronuclei encapsulating random chromosomes have been used; however, the eventual reincorporation of the micronucleated chromosome into daughter cell nuclei poses a challenge in tracking the chromosome for multiple cell cycles. Here we outline an approach to genetically engineer stable human cell lines capable of efficient chromosome-specific micronuclei induction. This strategy, which targets the CENP-B-deficient Y chromosome centromere for inactivation, allows the stepwise process of chromothripsis to be experimentally recapitulated, including the mechanisms and timing of chromosome fragmentation. Lastly, we describe the integration of a selection marker onto the micronucleated Y chromosome that enables the diverse genomic rearrangement landscape arising from micronuclei formation to be interrogated.
Collapse
Affiliation(s)
- Yu-Fen Lin
- Department of Pathology, Department of Cell Biology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Qing Hu
- Department of Pathology, Department of Cell Biology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Alison Guyer
- Department of Pathology, Department of Cell Biology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | | | - Peter Ly
- Department of Pathology, Department of Cell Biology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
85
|
Ippolito MR, Santaguida S. Generation of aneuploid cells and assessment of their ability to survive in presence of chemotherapeutic agents. Methods Cell Biol 2022; 182:21-33. [PMID: 38359978 DOI: 10.1016/bs.mcb.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Aneuploidy is a condition in which cells have an abnormal number of chromosomes that is not a multiple of the haploid complement. It is known that aneuploidy has detrimental consequences on cell physiology, such as genome instability, metabolic and proteotoxic stress and decreased cellular fitness. Importantly, aneuploidy is a hallmark of tumors and it is associated with resistance to chemotherapeutic agents and poor clinical outcome. To shed light into how aneuploidy contributes to chemoresistance, we induced chromosome mis-segregation in human cancer cell lines, then treated them with several chemotherapeutic agents and evaluated the emergence of chemoresistance. By doing so, we found that elevation of chromosome mis-segregation promotes resistance to chemotherapeutic agents through the expansion of aneuploid karyotypes and subsequent selection of specific aneuploidies essential for cellular viability under those stressful conditions. Here, we describe a method to generate aneuploid cell populations and to evaluate their resistance to anti-cancer agents. This protocol has been already successfully employed and can be further utilized to accelerate the exploration of the role of aneuploidy in chemoresistance.
Collapse
Affiliation(s)
- Marica Rosaria Ippolito
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Stefano Santaguida
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
86
|
Herman JA, Romain RR, Hoellerbauer P, Shirnekhi HK, King DC, DeLuca KF, Osborne Nishimura E, Paddison PJ, DeLuca JG. Hyper-active RAS/MAPK introduces cancer-specific mitotic vulnerabilities. Proc Natl Acad Sci U S A 2022; 119:e2208255119. [PMID: 36191188 PMCID: PMC9565228 DOI: 10.1073/pnas.2208255119] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/26/2022] [Indexed: 01/04/2023] Open
Abstract
Aneuploidy, the incorrect number of whole chromosomes, is a common feature of tumors that contributes to their initiation and evolution. Preventing aneuploidy requires properly functioning kinetochores, which are large protein complexes assembled on centromeric DNA that link mitotic chromosomes to dynamic spindle microtubules and facilitate chromosome segregation. The kinetochore leverages at least two mechanisms to prevent aneuploidy: error correction and the spindle assembly checkpoint (SAC). BubR1, a factor involved in both processes, was identified as a cancer dependency and therapeutic target in multiple tumor types; however, it remains unclear what specific oncogenic pressures drive this enhanced dependency on BubR1 and whether it arises from BubR1's regulation of the SAC or error-correction pathways. Here, we use a genetically controlled transformation model and glioblastoma tumor isolates to show that constitutive signaling by RAS or MAPK is necessary for cancer-specific BubR1 vulnerability. The MAPK pathway enzymatically hyperstimulates a network of kinetochore kinases that compromises chromosome segregation, rendering cells more dependent on two BubR1 activities: counteracting excessive kinetochore-microtubule turnover for error correction and maintaining the SAC. This work expands our understanding of how chromosome segregation adapts to different cellular states and reveals an oncogenic trigger of a cancer-specific defect.
Collapse
Affiliation(s)
- Jacob A. Herman
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523
| | - Romario R. Romain
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523
| | - Pia Hoellerbauer
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109
| | - Hazheen K. Shirnekhi
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523
| | - David C. King
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523
| | - Keith F. DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523
| | - Erin Osborne Nishimura
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523
| | | | - Jennifer G. DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523
| |
Collapse
|
87
|
Sedzro DM, Yuan X, Mullen M, Ejaz U, Yang T, Liu X, Song X, Tang YC, Pan W, Zou P, Gao X, Wang D, Wang Z, Dou Z, Liu X, Yao X. Phosphorylation of CENP-R by Aurora B regulates kinetochore-microtubule attachment for accurate chromosome segregation. J Mol Cell Biol 2022; 14:6693714. [PMID: 36069839 PMCID: PMC9802239 DOI: 10.1093/jmcb/mjac051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/14/2022] [Accepted: 09/01/2022] [Indexed: 01/14/2023] Open
Abstract
Error-free mitosis depends on accurate chromosome attachment to spindle microtubules via a fine structure called the centromere that is epigenetically specified by the enrichment of CENP-A nucleosomes. Centromere maintenance during mitosis requires CENP-A-mediated deposition of constitutive centromere-associated network that establishes the inner kinetochore and connects centromeric chromatin to spindle microtubules during mitosis. Although previously proposed to be an adaptor of retinoic acid receptor, here, we show that CENP-R synergizes with CENP-OPQU to regulate kinetochore-microtubule attachment stability and ensure accurate chromosome segregation in mitosis. We found that a phospho-mimicking mutation of CENP-R weakened its localization to the kinetochore, suggesting that phosphorylation may regulate its localization. Perturbation of CENP-R phosphorylation is shown to prevent proper kinetochore-microtubule attachment at metaphase. Mechanistically, CENP-R phosphorylation disrupts its binding with CENP-U. Thus, we speculate that Aurora B-mediated CENP-R phosphorylation promotes the correction of improper kinetochore-microtubule attachment in mitosis. As CENP-R is absent from yeast, we reasoned that metazoan evolved an elaborate chromosome stability control machinery to ensure faithful chromosome segregation in mitosis.
Collapse
Affiliation(s)
- Divine Mensah Sedzro
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China,Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, National Center for Cross-Disciplinary Sciences & CAS Center for Excellence in Molecular Cell Science, Hefei 230026, China
| | - Xiao Yuan
- Correspondence to: Xiao Yuan, E-mail:
| | - McKay Mullen
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China,Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, National Center for Cross-Disciplinary Sciences & CAS Center for Excellence in Molecular Cell Science, Hefei 230026, China,Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Umer Ejaz
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China,Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, National Center for Cross-Disciplinary Sciences & CAS Center for Excellence in Molecular Cell Science, Hefei 230026, China
| | - Tongtong Yang
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China,Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, National Center for Cross-Disciplinary Sciences & CAS Center for Excellence in Molecular Cell Science, Hefei 230026, China
| | - Xu Liu
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China,Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, National Center for Cross-Disciplinary Sciences & CAS Center for Excellence in Molecular Cell Science, Hefei 230026, China,Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xiaoyu Song
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China,Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, National Center for Cross-Disciplinary Sciences & CAS Center for Excellence in Molecular Cell Science, Hefei 230026, China,Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Yun-Chi Tang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Weijun Pan
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Peng Zou
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Xinjiao Gao
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China,Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, National Center for Cross-Disciplinary Sciences & CAS Center for Excellence in Molecular Cell Science, Hefei 230026, China
| | - Dongmei Wang
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China,Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, National Center for Cross-Disciplinary Sciences & CAS Center for Excellence in Molecular Cell Science, Hefei 230026, China
| | - Zhikai Wang
- MOE Key Laboratory for Cellular Dynamics, University of Science and Technology of China School of Life Sciences, Hefei 230027, China,Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, National Center for Cross-Disciplinary Sciences & CAS Center for Excellence in Molecular Cell Science, Hefei 230026, China,Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Zhen Dou
- Correspondence to: Zhen Dou, E-mail:
| | - Xing Liu
- Correspondence to: Xing Liu, E-mail:
| | | |
Collapse
|
88
|
Dale KL, Armond JW, Hynds RE, Vladimirou E. Modest increase of KIF11 expression exposes fragilities in the mitotic spindle, causing chromosomal instability. J Cell Sci 2022; 135:jcs260031. [PMID: 35929456 PMCID: PMC10500341 DOI: 10.1242/jcs.260031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/01/2022] [Indexed: 11/20/2022] Open
Abstract
Chromosomal instability (CIN), the process of increased chromosomal alterations, compromises genomic integrity and has profound consequences on human health. Yet, our understanding of the molecular and mechanistic basis of CIN initiation remains limited. We developed a high-throughput, single-cell, image-based pipeline employing deep-learning and spot-counting models to detect CIN by automatically counting chromosomes and micronuclei. To identify CIN-initiating conditions, we used CRISPR activation in human diploid cells to upregulate, at physiologically relevant levels, 14 genes that are functionally important in cancer. We found that upregulation of CCND1, FOXA1 and NEK2 resulted in pronounced changes in chromosome counts, and KIF11 upregulation resulted in micronuclei formation. We identified KIF11-dependent fragilities within the mitotic spindle; increased levels of KIF11 caused centrosome fragmentation, higher microtubule stability, lagging chromosomes or mitotic catastrophe. Our findings demonstrate that even modest changes in the average expression of single genes in a karyotypically stable background are sufficient for initiating CIN by exposing fragilities of the mitotic spindle, which can lead to a genomically diverse cell population.
Collapse
Affiliation(s)
- Katie L. Dale
- Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
- Mitotic Dynamics and Chromosomal Instability Laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| | - Jonathan W. Armond
- Mitotic Dynamics and Chromosomal Instability Laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| | - Robert E. Hynds
- Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
- Epithelial Cell Biology in ENT Research Group, UCL Great Ormond Street Institute of Child Health, University College London, London, WC1N 1EH, UK
| | - Elina Vladimirou
- Cancer Research UK Lung Cancer Centre of Excellence, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
- Mitotic Dynamics and Chromosomal Instability Laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| |
Collapse
|
89
|
Bočkaj I, Martini TEI, Smit MJ, Armandari I, Bakker B, Wardenaar R, Meeuwsen-de Boer TGJ, Bakker PL, Spierings DCJ, Hoving EW, Guryev V, Foijer F, Bruggeman SWM. Chromosomal Instability Characterizes Pediatric Medulloblastoma but Is Not Tolerated in the Developing Cerebellum. Int J Mol Sci 2022; 23:ijms23179852. [PMID: 36077248 PMCID: PMC9456393 DOI: 10.3390/ijms23179852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/21/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Medulloblastoma is a pediatric brain malignancy that consists of four transcriptional subgroups. Structural and numerical aneuploidy are common in all subgroups, although they are particularly profound in Group 3 and Group 4 medulloblastoma and in a subtype of SHH medulloblastoma termed SHHα. This suggests that chromosomal instability (CIN), the process leading to aneuploidy, is an important player in medulloblastoma pathophysiology. However, it is not known if there is ongoing CIN in medulloblastoma or if CIN affects the developing cerebellum and promotes tumor formation. To investigate this, we performed karyotyping of single medulloblastoma cells and demonstrated the presence of distinct tumor cell clones harboring unique copy number alterations, which is suggestive of ongoing CIN. We also found enrichment for processes related to DNA replication, repair, and mitosis in both SHH medulloblastoma and in the highly proliferative compartment of the presumed tumor cell lineage-of-origin, the latter also being sensitive to genotoxic stress. However, when challenging these tumor cells-of-origin with genetic lesions inducing CIN using transgenic mouse modeling, we found no evidence for large chromosomal aberrations in the cerebellum or for medulloblastoma formation. We therefore conclude that without a background of specific genetic mutations, CIN is not tolerated in the developing cerebellum in vivo and, thus, by itself is not sufficient to initiate medulloblastoma.
Collapse
Affiliation(s)
- Irena Bočkaj
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Tosca E. I. Martini
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Marlinde J. Smit
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Inna Armandari
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Bjorn Bakker
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - René Wardenaar
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Tiny G. J. Meeuwsen-de Boer
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Petra L. Bakker
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Diana C. J. Spierings
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Eelco W. Hoving
- Princess Máxima Center for Pediatric Oncology, 3584 EA Utrecht, The Netherlands
| | - Victor Guryev
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
- Correspondence: (F.F.); (S.W.M.B.)
| | - Sophia W. M. Bruggeman
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
- Correspondence: (F.F.); (S.W.M.B.)
| |
Collapse
|
90
|
I B, López-Jiménez P, Mena I, Viera A, Page J, González-Martínez J, Maestre C, Malumbres M, Suja JA, Gómez R. Haspin participates in AURKB recruitment to centromeres and contributes to chromosome congression in male mouse meiosis. J Cell Sci 2022; 135:275954. [PMID: 35694956 DOI: 10.1242/jcs.259546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 06/06/2022] [Indexed: 11/20/2022] Open
Abstract
Chromosome segregation requires that centromeres properly attach to spindle microtubules. This essential step regulates the accuracy of cell division and therefore must be precisely regulated. One of the main centromeric regulatory signaling pathways is the Haspin-H3T3ph-chromosomal passenger complex (CPC) cascade, which is responsible for the recruitment of the CPC to the centromeres. In mitosis, Haspin kinase phosphorylates histone H3 at threonine 3 (H3T3ph), an essential epigenetic mark that recruits the CPC, whose catalytic component is Aurora B kinase. However, the centromeric Haspin-H3T3ph-CPC pathway remains largely uncharacterized in mammalian male meiosis. We have analyzed Haspin functions by either its chemical inhibition in cultured spermatocytes using LDN-192960, or the ablation of Haspin gene in Haspin-/-. Our studies suggest that Haspin kinase activity is required for proper chromosome congression during both meiotic divisions and for the recruitment of Aurora B and kinesin MCAK to meiotic centromeres. However, the absence of H3T3ph histone mark does not alter Borealin and SGO2 centromeric localization. These results add new and relevant information regarding the regulation of the Haspin-H3T3ph-CPC pathway and centromere function during meiosis.
Collapse
Affiliation(s)
- Berenguer I
- Cell Biology Unit, Department of Biology, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | - P López-Jiménez
- Cell Biology Unit, Department of Biology, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | - I Mena
- Cell Biology Unit, Department of Biology, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | - A Viera
- Cell Biology Unit, Department of Biology, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | - J Page
- Cell Biology Unit, Department of Biology, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | - J González-Martínez
- Cell Division and Cancer group, Spanish National Cancer Research Centre (CNIO), 29029 Madrid, Spain
| | - C Maestre
- Cell Division and Cancer group, Spanish National Cancer Research Centre (CNIO), 29029 Madrid, Spain
| | - M Malumbres
- Cell Division and Cancer group, Spanish National Cancer Research Centre (CNIO), 29029 Madrid, Spain
| | - J A Suja
- Cell Biology Unit, Department of Biology, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | - R Gómez
- Cell Biology Unit, Department of Biology, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| |
Collapse
|
91
|
Liu S, Yuan X, Gui P, Liu R, Durojaye O, Hill DL, Fu C, Yao X, Dou Z, Liu X. Mad2 promotes Cyclin B2 recruitment to the kinetochore for guiding accurate mitotic checkpoint. EMBO Rep 2022; 23:e54171. [PMID: 35384228 PMCID: PMC9171689 DOI: 10.15252/embr.202154171] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 11/09/2022] Open
Abstract
Accurate mitotic progression relies on the dynamic phosphorylation of multiple substrates by key mitotic kinases. Cyclin-dependent kinase 1 is a master kinase that coordinates mitotic progression and requires its regulatory subunit Cyclin B to ensure full kinase activity and substrate specificity. The function of Cyclin B2, which is a closely related family member of Cyclin B1, remains largely elusive. Here, we show that Mad2 promotes the kinetochore localization of Cyclin B2 and that their interaction at the kinetochores guides accurate chromosome segregation. Our biochemical analyses have characterized the Mad2-Cyclin B2 interaction and delineated a novel Mad2-interacting motif (MIM) on Cyclin B2. The functional importance of the Cyclin B2-Mad2 interaction was demonstrated by real-time imaging in which MIM-deficient mutant Cyclin B2 failed to rescue the chromosomal segregation defects. Taken together, we have delineated a previously undefined function of Cyclin B2 at the kinetochore and have established, in human cells, a mechanism of action by which Mad2 contributes to the spindle checkpoint.
Collapse
Affiliation(s)
- Sikai Liu
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xiao Yuan
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Ping Gui
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Ran Liu
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Olanrewaju Durojaye
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Donald L Hill
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chuanhai Fu
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zhen Dou
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics and The First Affiliated Hospital, School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
92
|
Kim T. Recent Progress on the Localization of PLK1 to the Kinetochore and Its Role in Mitosis. Int J Mol Sci 2022; 23:ijms23095252. [PMID: 35563642 PMCID: PMC9102930 DOI: 10.3390/ijms23095252] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 12/10/2022] Open
Abstract
The accurate distribution of the replicated genome during cell division is essential for cell survival and healthy organismal development. Errors in this process have catastrophic consequences, such as birth defects and aneuploidy, a hallmark of cancer cells. PLK1 is one of the master kinases in mitosis and has multiple functions, including mitotic entry, chromosome segregation, spindle assembly checkpoint, and cytokinesis. To dissect the role of PLK1 in mitosis, it is important to understand how PLK1 localizes in the specific region in cells. PLK1 localizes at the kinetochore and is essential in spindle assembly checkpoint and chromosome segregation. However, how PLK1 localizes at the kinetochore remains elusive. Here, we review the recent literature on the kinetochore recruitment mechanisms of PLK1 and its roles in spindle assembly checkpoint and attachment between kinetochores and spindle microtubules. Together, this review provides an overview of how the local distribution of PLK1 could regulate major pathways in mitosis.
Collapse
Affiliation(s)
- Taekyung Kim
- Department of Biology Education, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Korea
| |
Collapse
|
93
|
Raisch T, Ciossani G, d’Amico E, Cmentowski V, Carmignani S, Maffini S, Merino F, Wohlgemuth S, Vetter IR, Raunser S, Musacchio A. Structure of the RZZ complex and molecular basis of Spindly-driven corona assembly at human kinetochores. EMBO J 2022; 41:e110411. [PMID: 35373361 PMCID: PMC9058546 DOI: 10.15252/embj.2021110411] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 11/09/2022] Open
Abstract
In metazoans, a ≈1 megadalton (MDa) multiprotein complex comprising the dynein-dynactin adaptor Spindly and the ROD-Zwilch-ZW10 (RZZ) complex is the building block of a fibrous biopolymer, the kinetochore fibrous corona. The corona assembles on mitotic kinetochores to promote microtubule capture and spindle assembly checkpoint (SAC) signaling. We report here a high-resolution cryo-EM structure that captures the essential features of the RZZ complex, including a farnesyl-binding site required for Spindly binding. Using a highly predictive in vitro assay, we demonstrate that the SAC kinase MPS1 is necessary and sufficient for corona assembly at supercritical concentrations of the RZZ-Spindly (RZZS) complex, and describe the molecular mechanism of phosphorylation-dependent filament nucleation. We identify several structural requirements for RZZS polymerization in rings and sheets. Finally, we identify determinants of kinetochore localization and corona assembly of Spindly. Our results describe a framework for the long-sought-for molecular basis of corona assembly on metazoan kinetochores.
Collapse
Affiliation(s)
- Tobias Raisch
- Department of Structural BiochemistryMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Giuseppe Ciossani
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
- Present address:
European Institute of OncologyMilanItaly
| | - Ennio d’Amico
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Verena Cmentowski
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Sara Carmignani
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Stefano Maffini
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Felipe Merino
- Department of Structural BiochemistryMax Planck Institute of Molecular PhysiologyDortmundGermany
- Present address:
Department of Protein EvolutionMax Planck Institute for Developmental BiologyTübingenGermany
| | - Sabine Wohlgemuth
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Ingrid R Vetter
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Stefan Raunser
- Department of Structural BiochemistryMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Andrea Musacchio
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
- Centre for Medical BiotechnologyFaculty of BiologyUniversity Duisburg‐EssenEssenGermany
| |
Collapse
|
94
|
SWAP, SWITCH, and STABILIZE: Mechanisms of Kinetochore–Microtubule Error Correction. Cells 2022; 11:cells11091462. [PMID: 35563768 PMCID: PMC9104000 DOI: 10.3390/cells11091462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 11/17/2022] Open
Abstract
For correct chromosome segregation in mitosis, eukaryotic cells must establish chromosome biorientation where sister kinetochores attach to microtubules extending from opposite spindle poles. To establish biorientation, any aberrant kinetochore–microtubule interactions must be resolved in the process called error correction. For resolution of the aberrant interactions in error correction, kinetochore–microtubule interactions must be exchanged until biorientation is formed (the SWAP process). At initiation of biorientation, the state of weak kinetochore–microtubule interactions should be converted to the state of stable interactions (the SWITCH process)—the conundrum of this conversion is called the initiation problem of biorientation. Once biorientation is established, tension is applied on kinetochore–microtubule interactions, which stabilizes the interactions (the STABILIZE process). Aurora B kinase plays central roles in promoting error correction, and Mps1 kinase and Stu2 microtubule polymerase also play important roles. In this article, we review mechanisms of error correction by considering the SWAP, SWITCH, and STABILIZE processes. We mainly focus on mechanisms found in budding yeast, where only one microtubule attaches to a single kinetochore at biorientation, making the error correction mechanisms relatively simpler.
Collapse
|
95
|
Doodhi H, Tanaka TU. Swap and stop - Kinetochores play error correction with microtubules: Mechanisms of kinetochore-microtubule error correction: Mechanisms of kinetochore-microtubule error correction. Bioessays 2022; 44:e2100246. [PMID: 35261042 PMCID: PMC9344824 DOI: 10.1002/bies.202100246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/30/2022]
Abstract
Correct chromosome segregation in mitosis relies on chromosome biorientation, in which sister kinetochores attach to microtubules from opposite spindle poles prior to segregation. To establish biorientation, aberrant kinetochore–microtubule interactions must be resolved through the error correction process. During error correction, kinetochore–microtubule interactions are exchanged (swapped) if aberrant, but the exchange must stop when biorientation is established. In this article, we discuss recent findings in budding yeast, which have revealed fundamental molecular mechanisms promoting this “swap and stop” process for error correction. Where relevant, we also compare the findings in budding yeast with mechanisms in higher eukaryotes. Evidence suggests that Aurora B kinase differentially regulates kinetochore attachments to the microtubule end and its lateral side and switches relative strength of the two kinetochore–microtubule attachment modes, which drives the exchange of kinetochore–microtubule interactions to resolve aberrant interactions. However, Aurora B kinase, recruited to centromeres and inner kinetochores, cannot reach its targets at kinetochore–microtubule interface when tension causes kinetochore stretching, which stops the kinetochore–microtubule exchange once biorientation is established.
Collapse
Affiliation(s)
- Harinath Doodhi
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
| | - Tomoyuki U Tanaka
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK
| |
Collapse
|
96
|
Yue W, Wang Y, Meng T, Zhang H, Zhang X, Ouyang Y, Hou Y, Schatten H, Wang Z, Sun Q. Kinetochore scaffold 1 regulates SAC function during mouse oocyte meiotic maturation. FASEB J 2022; 36:e22210. [DOI: 10.1096/fj.202101586rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Wei Yue
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
- University of Chinese Academy of Sciences Beijing China
| | - Yue Wang
- College of Animal Science and Technology Nanjing Agricultural University Nanjing China
| | - Tie‐Gang Meng
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
| | - Hong‐Yong Zhang
- Department of Reproductive Medicine Peking University Shenzhen Hospital, Shenzhen Peking University‐The Hong Kong University of Science and Technology Medical Center Shenzhen China
| | - Xin‐Ran Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
- University of Chinese Academy of Sciences Beijing China
| | - Ying‐Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
| | - Heide Schatten
- Department of Veterinary Pathobiology University of Missouri Columbia Missouri USA
| | - Zhen‐Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology Institute of Zoology Chinese Academy of Sciences Beijing China
- University of Chinese Academy of Sciences Beijing China
| | - Qing‐Yuan Sun
- Fertility Preservation Lab, Guangdong‐Hong Kong Metabolism & Reproduction Joint Laboratory Reproductive Medicine Center Guangdong Second Provincial General Hospital Guangzhou China
| |
Collapse
|
97
|
Mammel AE, Huang HZ, Gunn AL, Choo E, Hatch EM. Chromosome length and gene density contribute to micronuclear membrane stability. Life Sci Alliance 2022; 5:e202101210. [PMID: 34789512 PMCID: PMC8605325 DOI: 10.26508/lsa.202101210] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 11/24/2022] Open
Abstract
Micronuclei are derived from missegregated chromosomes and frequently lose membrane integrity, leading to DNA damage, innate immune activation, and metastatic signaling. Here, we demonstrate that two characteristics of the trapped chromosome, length and gene density, are key contributors to micronuclei membrane stability and determine the timing of micronucleus rupture. We demonstrate that these results are not due to chromosome-specific differences in spindle position or initial protein recruitment during post-mitotic nuclear envelope assembly. Micronucleus size strongly correlates with lamin B1 levels and nuclear pore density in intact micronuclei, but, unexpectedly, lamin B1 levels do not completely predict nuclear lamina organization or membrane stability. Instead, small gene-dense micronuclei have decreased nuclear lamina gaps compared to large micronuclei, despite very low levels of lamin B1. Our data strongly suggest that nuclear envelope composition defects previously correlated with membrane rupture only partly explain membrane stability in micronuclei. We propose that an unknown factor linked to gene density has a separate function that inhibits the appearance of nuclear lamina gaps and delays membrane rupture until late in the cell cycle.
Collapse
Affiliation(s)
- Anna E Mammel
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Heather Z Huang
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Amanda L Gunn
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Emma Choo
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Emily M Hatch
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
98
|
Barbosa J, Sunkel CE, Conde C. The Role of Mitotic Kinases and the RZZ Complex in Kinetochore-Microtubule Attachments: Doing the Right Link. Front Cell Dev Biol 2022; 10:787294. [PMID: 35155423 PMCID: PMC8832123 DOI: 10.3389/fcell.2022.787294] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/13/2022] [Indexed: 12/31/2022] Open
Abstract
During mitosis, the interaction of kinetochores (KTs) with microtubules (MTs) drives chromosome congression to the spindle equator and supports the segregation of sister chromatids. Faithful genome partition critically relies on the ability of chromosomes to establish and maintain proper amphitelic end-on attachments, a configuration in which sister KTs are connected to robust MT fibers emanating from opposite spindle poles. Because the capture of spindle MTs by KTs is error prone, cells use mechanisms that sense and correct inaccurate KT-MT interactions before committing to segregate sister chromatids in anaphase. If left unresolved, these errors can result in the unequal distribution of chromosomes and lead to aneuploidy, a hallmark of cancer. In this review, we provide an overview of the molecular strategies that monitor the formation and fine-tuning of KT-MT attachments. We describe the complex network of proteins that operates at the KT-MT interface and discuss how AURORA B and PLK1 coordinate several concurrent events so that the stability of KT-MT attachments is precisely modulated throughout mitotic progression. We also outline updated knowledge on how the RZZ complex is regulated to ensure the formation of end-on attachments and the fidelity of mitosis.
Collapse
Affiliation(s)
- João Barbosa
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Claudio E. Sunkel
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Carlos Conde
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
99
|
DeLuca KF, Mick JE, Ide AH, Lima WC, Sherman L, Schaller KL, Anderson SM, Zhao N, Stasevich TJ, Varma D, Nilsson J, DeLuca JG. Generation and diversification of recombinant monoclonal antibodies. eLife 2021; 10:72093. [PMID: 34970967 PMCID: PMC8763395 DOI: 10.7554/elife.72093] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Antibodies are indispensable tools used for a large number of applications in both foundational and translational bioscience research; however, there are drawbacks to using traditional antibodies generated in animals. These include a lack of standardization leading to problems with reproducibility, high costs of antibodies purchased from commercial sources, and ethical concerns regarding the large number of animals used to generate antibodies. To address these issues, we have developed practical methodologies and tools for generating low-cost, high-yield preparations of recombinant monoclonal antibodies and antibody fragments directed to protein epitopes from primary sequences. We describe these methods here, as well as approaches to diversify monoclonal antibodies, including customization of antibody species specificity, generation of genetically encoded small antibody fragments, and conversion of single chain antibody fragments (e.g. scFv) into full-length, bivalent antibodies. This study focuses on antibodies directed to epitopes important for mitosis and kinetochore function; however, the methods and reagents described here are applicable to antibodies and antibody fragments for use in any field.
Collapse
Affiliation(s)
- Keith F DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Jeanne E Mick
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Amy Hodges Ide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Wanessa C Lima
- Geneva Antibody Facility, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Lori Sherman
- CU Cancer Center Cell Technologies Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Kristin L Schaller
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Steven M Anderson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Ning Zhao
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Timothy J Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Dileep Varma
- Department of Cell and Developmental Biology, Northwestern University, Chicago, United States
| | - Jakob Nilsson
- The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Germany
| | - Jennifer G DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| |
Collapse
|
100
|
Sarangapani KK, Koch LB, Nelson CR, Asbury CL, Biggins S. Kinetochore-bound Mps1 regulates kinetochore-microtubule attachments via Ndc80 phosphorylation. J Cell Biol 2021; 220:e202106130. [PMID: 34647959 PMCID: PMC8641409 DOI: 10.1083/jcb.202106130] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/03/2021] [Accepted: 09/09/2021] [Indexed: 12/22/2022] Open
Abstract
Dividing cells detect and correct erroneous kinetochore-microtubule attachments during mitosis, thereby avoiding chromosome missegregation. The Aurora B kinase phosphorylates microtubule-binding elements specifically at incorrectly attached kinetochores, promoting their release and providing another chance for proper attachments to form. However, growing evidence suggests that the Mps1 kinase is also required for error correction. Here we directly examine how Mps1 activity affects kinetochore-microtubule attachments using a reconstitution-based approach that allows us to separate its effects from Aurora B activity. When endogenous Mps1 that copurifies with kinetochores is activated in vitro, it weakens their attachments to microtubules via phosphorylation of Ndc80, a major microtubule-binding protein. This phosphorylation contributes to error correction because phospho-deficient Ndc80 mutants exhibit genetic interactions and segregation defects when combined with mutants in other error correction pathways. In addition, Mps1 phosphorylation of Ndc80 is stimulated on kinetochores lacking tension. These data suggest that Mps1 provides an additional mechanism for correcting erroneous kinetochore-microtubule attachments, complementing the well-known activity of Aurora B.
Collapse
Affiliation(s)
| | - Lori B. Koch
- Howard Hughes Medical Institute, Chevy Chase, MD
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA
| | - Christian R. Nelson
- Howard Hughes Medical Institute, Chevy Chase, MD
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Charles L. Asbury
- Department of Physiology & Biophysics, University of Washington, Seattle, WA
| | - Sue Biggins
- Howard Hughes Medical Institute, Chevy Chase, MD
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|