1
|
Sethi S, Ghetti S, Cmentowski V, Guerriere TB, Stege P, Piano V, Musacchio A. Interplay of kinetochores and catalysts drives rapid assembly of the mitotic checkpoint complex. Nat Commun 2025; 16:4823. [PMID: 40410156 PMCID: PMC12102207 DOI: 10.1038/s41467-025-59970-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 05/09/2025] [Indexed: 05/25/2025] Open
Abstract
The spindle assembly checkpoint (SAC) ensures mitotic exit occurs only after sister chromatid biorientation, but how this coordination is mechanistically achieved remains unclear. Kinetochores, the megadalton complexes linking chromosomes to spindle microtubules, contribute to SAC signaling. However, whether they act solely as docking platforms or actively promote the co-orientation of SAC catalysts such as MAD1:MAD2 and BUB1:BUB3 remains unresolved. Here, we reconstitute kinetochores and SAC signaling in vitro to address this question. We engineer recombinant kinetochore particles that recruit core SAC components and trigger checkpoint signaling upon Rapamycin induction, and test their function using a panel of targeted mutants. At approximately physiological concentrations of SAC proteins, kinetochores are essential for efficient mitotic checkpoint complex (MCC) assembly, the key effector of SAC signaling. Our results suggest that kinetochores serve not only as structural hubs but also as catalytic platforms that concentrate and spatially organize SAC components to accelerate MCC formation and ensure timely checkpoint activation.
Collapse
Affiliation(s)
- Suruchi Sethi
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227, Dortmund, Germany
- Eradigm Consulting, 6-7 St Cross St, London, EC1N 8UB, UK
| | - Sabrina Ghetti
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227, Dortmund, Germany
| | - Verena Cmentowski
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227, Dortmund, Germany
| | - Teresa Benedetta Guerriere
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227, Dortmund, Germany
| | - Patricia Stege
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227, Dortmund, Germany
| | - Valentina Piano
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227, Dortmund, Germany
- Institute of Human Genetics, Center of Molecular Medicine Cologne (CMMC), University of Cologne, Robert-Koch Str. 21 50931, Cologne, Germany
| | - 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
|
2
|
Piano V. Multitasking Proteins: Exploring Noncanonical Functions of Proteins during Mitosis. Biochemistry 2025; 64:2123-2137. [PMID: 40315343 DOI: 10.1021/acs.biochem.5c00083] [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: 05/04/2025]
Abstract
This review provides a comprehensive overview of how mitotic cells drive the repurposing of proteins to fulfill mitosis-specific functions. To ensure the successful completion of cell division, the cell strategically reallocates its "workforce" by assigning additional functions to available proteins. Protein repurposing occurs at multiple levels of cellular organization and involves diverse mechanisms. At the protein level, proteins may gain mitosis-specific functions through post-translational modifications. At the structural level, proteins that typically maintain cellular architecture in interphase are co-opted to participate in mitotic spindle formation, chromosome condensation, and kinetochore assembly. Furthermore, the dynamic reorganization of the nuclear envelope and other organelles relies on the temporary reassignment of enzymes, structural proteins, and motor proteins to facilitate these changes. These adaptive mechanisms underscore the remarkable versatility of the cellular proteome in responding to the stringent requirements of mitosis. By leveraging the existing proteome for dual or multiple specialized roles, cells optimize resource usage while maintaining the precision needed to preserve genomic integrity and ensure the survival of the next generation of cells.
Collapse
Affiliation(s)
- Valentina Piano
- Institute of Human Genetics, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital, University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
3
|
Hu C, Hu Q, Yang T, Xu P, Xiong F, Wang X, Wang C, Jiang K, Hill DL, Xue L, Tao C, Fu C, Zhang L, Liu D, Xiang S, Zang J, Wang Z, Yao X, Liu X. Condensation-dependent multivalent interactions of EB1 and CENP-R regulate chromosome oscillations in mitosis. Cell Rep 2025; 44:115560. [PMID: 40349345 DOI: 10.1016/j.celrep.2025.115560] [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: 09/04/2024] [Revised: 02/12/2025] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
Mitotic chromosomes oscillate between the spindle poles upon the establishment of bi-orientation, which is essential for chromosome alignment and subsequent synchronous segregation. However, the molecular mechanisms underlying the oscillatory movement remain unclear. Recent studies revealed that phase separation of the end-binding protein 1 (EB1) is essential for eukaryotic cell division. Here, we show that EB1 interacts with CENP-R and that the phase separation-defective EB1 mutant fails to power the chromosome oscillations. Biochemical analyses reveal a co-condensation of EB1 and CENP-R, a subunit of the constitutive centromere-associated network. Nuclear magnetic resonance assays reveal that the interaction and co-condensation are largely mediated by the structured end-binding homology domain of EB1 and the non-structured N-terminal intrinsic disorder region of CENP-R. Chromosome oscillation is perturbed in cells expressing the EB1-binding-defective CENP-R mutant. Thus, phase-separated EB1 binding to CENP-R forms a physical link between inner kinetochore and dynamic spindle microtubule plus-ends to guide accurate chromosome oscillations.
Collapse
Affiliation(s)
- Chengcheng Hu
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Qing Hu
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Tongtong Yang
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Panpan Xu
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Fangyuan Xiong
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Xinyang Wang
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Chao Wang
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Kai Jiang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Donald L Hill
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Lin Xue
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Changlu Tao
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Chuanhai Fu
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Liang Zhang
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Dan Liu
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Shengqi Xiang
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Jianye Zang
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China
| | - Zhikai Wang
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, Hefei 230027, China; School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
4
|
Schweighofer J, Mulay B, Hoffmann I, Vogt D, Pesenti ME, Musacchio A. Interactions with multiple inner kinetochore proteins determine mitotic localization of FACT. J Cell Biol 2025; 224:e202412042. [PMID: 40094435 PMCID: PMC11912937 DOI: 10.1083/jcb.202412042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
The FAcilitates Chromatin Transcription (FACT) complex is a dimeric histone chaperone that operates on chromatin during transcription and replication. FACT also interacts with a specialized centromeric nucleosome containing the histone H3 variant centromere protein A (CENP-A) and with CENP-TW, two subunits of the constitutive centromere-associated network (CCAN), a 16-protein complex associated with CENP-A. The significance of these interactions remains elusive. Here, we show that FACT has multiple additional binding sites on CCAN. The interaction with CCAN is strongly stimulated by casein kinase II phosphorylation of FACT. Mitotic localization of FACT to kinetochores is strictly dependent on specific CCAN subcomplexes. Conversely, CENP-TW requires FACT for stable localization. Unexpectedly, we also find that DNA readily displaces FACT from CCAN, supporting the speculation that FACT becomes recruited through a pool of CCAN that is not stably integrated into chromatin. Collectively, our results point to a potential role of FACT in chaperoning CCAN during transcription or in the stabilization of CCAN at the centromere during the cell cycle.
Collapse
Affiliation(s)
- Julia Schweighofer
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Bhagyashree Mulay
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
| | - Ingrid Hoffmann
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Doro Vogt
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Marion E. Pesenti
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Andrea Musacchio
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany
- Max Planck School Matter to Life, Heidelberg, Germany
| |
Collapse
|
5
|
Durojaye OA, Yang F, Gao X, Aikhionbare F, Zhang L, Liu X, Yao X. Probing centromere-kinetochore core complex CENP-L/M assembly using cenpemlin. J Mol Cell Biol 2025; 16:mjae035. [PMID: 39243121 PMCID: PMC12046508 DOI: 10.1093/jmcb/mjae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/09/2024] [Indexed: 09/09/2024] Open
Affiliation(s)
- Olanrewaju Ayodeji Durojaye
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China
| | - Fengrui Yang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China
- Molecular Imaging Center, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xinjiao Gao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China
| | - Felix Aikhionbare
- Molecular Imaging Center, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Liangyu Zhang
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China
- Molecular Imaging Center, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xing Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China
- Molecular Imaging Center, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
6
|
Bousios A, Kakutani T, Henderson IR. Centrophilic Retrotransposons of Plant Genomes. ANNUAL REVIEW OF PLANT BIOLOGY 2025; 76:579-604. [PMID: 39952673 DOI: 10.1146/annurev-arplant-083123-082220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2025]
Abstract
The centromeres of eukaryotic chromosomes are required to load CENH3/CENP-A variant nucleosomes and the kinetochore complex, which connects to spindle microtubules during cell division. Despite their conserved function, plant centromeres show rapid sequence evolution within and between species and a range of monocentric, holocentric, and polymetacentric architectures, which vary in kinetochore numbers and spacing. Plant centromeres are commonly composed of tandem satellite repeat arrays, which are invaded by specific families of centrophilic retrotransposons, whereas in some species the entire centromere is composed of such retrotransposons. We review the diversity of plant centrophilic retrotransposons and their mechanisms of integration, together with how epigenetic information and small RNAs control their proliferation. We discuss models for rapid centromere sequence evolution and speculate on the roles that centrophilic retrotransposons may play in centromere dynamics. We focus on plants but draw comparisons with animal and fungal centromeric transposons to highlight conserved and divergent themes across the eukaryotes.
Collapse
Affiliation(s)
| | - Tetsuji Kakutani
- Department of Biological Sciences, University of Tokyo, Tokyo, Japan
| | - Ian R Henderson
- Department of Plant Sciences, University of Cambridge, Cambridge, United Kingdom;
| |
Collapse
|
7
|
Dudziak A, Pleuger R, Schmidt J, Hamm F, Tendulkar S, Jänen K, Vetter IR, Singh S, Fischböck J, Herzog F, Westermann S. The Spc105/Kre28 complex promotes mitotic error correction by outer kinetochore recruitment of Ipl1/Sli15. EMBO J 2025:10.1038/s44318-025-00437-w. [PMID: 40281358 DOI: 10.1038/s44318-025-00437-w] [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: 10/04/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Kinetochores link chromosomes to dynamic microtubules of the mitotic spindle. To ensure equal chromosome segregation, sister chromatids must achieve biorientation. The conserved kinase Aurora B phosphorylates outer kinetochore proteins on attachments lacking tension, allowing the re-establishment of new connections until biorientation is achieved. Aurora B localizes to the centromere as part of the chromosomal passenger complex (CPC), but the underlying recruitment pathways can be eliminated without disrupting biorientation. It therefore remains unclear how the kinase operates during error correction. Here, we identify the conserved Spc105/Kre28 complex as an outer kinetochore receptor of the Aurora kinase Ipl1 and its activator Sli15 in Saccharomyces cerevisiae. We show that mutations in the helix bundle domain of Spc105/Kre28 impair mitotic error correction, resembling the effects of ipl1 or sli15 mutants. The defects can be suppressed by the artificial recruitment of Ipl1. In biochemical experiments, Ipl1/Sli15 directly associates with Spc105/Kre28, and a conserved segment in the Sli15 central domain is crucially involved in the binding mechanism. These results have important implications for the mechanism of tension-dependent error correction during chromosome biorientation.
Collapse
Affiliation(s)
- Alexander Dudziak
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Universitätsstrasse 5, 45117, Essen, Germany
| | - Richard Pleuger
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Universitätsstrasse 5, 45117, Essen, Germany
| | - Jasmin Schmidt
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Universitätsstrasse 5, 45117, Essen, Germany
| | - Frederik Hamm
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Universitätsstrasse 5, 45117, Essen, Germany
| | - Sharvari Tendulkar
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Universitätsstrasse 5, 45117, Essen, Germany
| | - Karolin Jänen
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Universitätsstrasse 5, 45117, Essen, Germany
| | - Ingrid R Vetter
- Department of Mechanistic Cell Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Straße 11, 44227, Dortmund, Germany
| | - Sylvia Singh
- Gene Center Munich and Department of Biochemistry, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| | - Josef Fischböck
- Gene Center Munich and Department of Biochemistry, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| | - Franz Herzog
- Gene Center Munich and Department of Biochemistry, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 25, 81377, Munich, Germany
- Institute Krems Bioanalytics, IMC University of Applied Sciences, Krems, Piaristengasse 1, A-3500, Krems, Austria
| | - Stefan Westermann
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Universitätsstrasse 5, 45117, Essen, Germany.
| |
Collapse
|
8
|
Barrero DJ, Hedouin S, Mao Y, Asbury CL, Stergachis A, O'Toole E, Biggins S. Centromeres in the thermotolerant yeast K. marxianus mediate attachment to a single microtubule. RESEARCH SQUARE 2025:rs.3.rs-6173630. [PMID: 40313741 PMCID: PMC12045370 DOI: 10.21203/rs.3.rs-6173630/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Eukaryotic chromosome segregation requires spindle microtubules to attach to chromosomes through kinetochores. The chromosomal locus that mediates kinetochore assembly is the centromere and is epigenetically specified in most organisms by a centromeric histone H3 variant called CENP-A. An exception to this is budding yeast which have short, sequenced-defined point centromeres. In S. cerevisiae, a single CENP-A nucleosome is formed at the centromere and is sufficient for kinetochore assembly. The thermophilic budding yeast Kluyveromyces marxianus also has a point centromere but its length is nearly double the S. cerevisiae centromere and the number of centromeric nucleosomes and kinetochore attachment sites is unknown. Purification of native kinetochores from K. marxianus yielded a mixed population, with one subpopulation that appeared to consist of doublets, making it unclear whether K. marxianus shares the same attachment architecture as S. cerevisiae. Here, we demonstrate that though the doublet kinetochores have a functional impact on kinetochore strength, kinetochore localization throughout the cell cycle appears conserved between these two yeasts. In addition, whole spindle electron tomography demonstrates that a single microtubule binds to each chromosome. Single-molecule nucleosome mapping analysis suggests the presence of a single centromeric nucleosome. Taken together, we propose that the K. marxianus point centromere assembles a single centromeric nucleosome that mediates attachment to one microtubule.
Collapse
Affiliation(s)
| | - Sabrine Hedouin
- Howard Hughes Medical Institute, Fred Hutchinson Cancer Center
| | | | | | | | | | - Sue Biggins
- Howard Hughes Medical Institute, Fred Hutchinson Cancer Center
| |
Collapse
|
9
|
Biggs RJ, Sun M, Sundararajan K, Hendrix E, Straight AF, Marko JF. Independence of centromeric and pericentromeric chromatin stability on CCAN components. Mol Biol Cell 2025; 36:ar41. [PMID: 39937678 PMCID: PMC12005111 DOI: 10.1091/mbc.e24-02-0066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 01/28/2025] [Accepted: 02/04/2025] [Indexed: 02/14/2025] Open
Abstract
The chromatin of the centromere provides the assembly site for the mitotic kinetochore that couples microtubule attachment and force production to chromosome movement in mitosis. The chromatin of the centromere is specified by nucleosomes containing the histone H3 variant, CENP-A. The constitutive centromeric-associated network (CCAN) and kinetochore are assembled on CENP-A chromatin to enable chromosome separation. CENP-A chromatin is surrounded by pericentromeric heterochromatin, which itself is bound by the sequence specific binding protein, CENP-B. We performed mechanical experiments on mitotic chromosomes while tracking CENP-A and CENP-B to observe the centromere's stiffness and the role of the CCAN. We degraded CENP-C and CENP-N containing auxin-inducible degrons, which we verified compromises the CCAN via observation of CENP-T loss. Chromosome stretching revealed that the CENP-A domain does not visibly stretch, even in the absence of CENP-C and/or CENP-N. Pericentromeric chromatin deforms upon force application, stretching ∼3-fold less than the entire chromosome. CENP-C and/or CENP-N loss has no impact on pericentromere stretching. Chromosome-disconnecting nuclease treatments showed no structural effects on CENP-A. Our experiments show that the core-centromeric chromatin is more resilient and likely mechanically disconnected from the underlying pericentromeric chromatin, while the pericentric chromatin is deformable yet stiffer than the chromosome arms.
Collapse
Affiliation(s)
- Ronald J. Biggs
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Mingxuan Sun
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | | | - Eline Hendrix
- Department of Biochemistry, Stanford Medical School, Stanford, CA 94305
| | - Aaron F. Straight
- Department of Biochemistry, Stanford Medical School, Stanford, CA 94305
| | - John F. Marko
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
- Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208
| |
Collapse
|
10
|
Melters DP, Bui M, Rakshit T, Grigoryev SA, Sturgill D, Dalal Y. High-resolution analysis of human centromeric chromatin. Life Sci Alliance 2025; 8:e202402819. [PMID: 39848706 PMCID: PMC11757159 DOI: 10.26508/lsa.202402819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/25/2025] Open
Abstract
Centromeres are marked by the centromere-specific histone H3 variant CENP-A/CENH3. Throughout the cell cycle, the constitutive centromere-associated network is bound to CENP-A chromatin, but how this protein network modifies CENP-A nucleosome conformations in vivo is unknown. Here, we purify endogenous centromeric chromatin associated with the CENP-C complex across the cell cycle and analyze the structures by single-molecule imaging and biochemical assays. CENP-C complex-bound chromatin was refractory to MNase digestion. The CENP-C complex increased in height throughout the cell cycle culminating in mitosis, and the smaller CENP-C complex corresponds to the dimensions of in vitro reconstituted constitutive centromere-associated network. In addition, we found two distinct CENP-A nucleosomal configurations; the taller variant was associated with the CENP-C complex. Finally, CENP-A mutants partially corrected CENP-C overexpression-induced centromeric transcription and mitotic defects. In all, our data support a working model in which CENP-C is critical in regulating centromere homeostasis by supporting a unique higher order structure of centromeric chromatin and altering the accessibility of the centromeric chromatin fiber for transcriptional machinery.
Collapse
Affiliation(s)
- Daniël P Melters
- National Cancer Institute, Center for Cancer Research, Laboratory of Receptor Biology and Gene Expression, Bethesda, MD, USA
| | - Minh Bui
- National Cancer Institute, Center for Cancer Research, Laboratory of Receptor Biology and Gene Expression, Bethesda, MD, USA
| | - Tatini Rakshit
- National Cancer Institute, Center for Cancer Research, Laboratory of Receptor Biology and Gene Expression, Bethesda, MD, USA
- Department of Chemistry, Shiv Nadar Institution of Eminence, Delhi, India
| | | | - David Sturgill
- National Cancer Institute, Center for Cancer Research, Laboratory of Receptor Biology and Gene Expression, Bethesda, MD, USA
- National Cancer Institute, Center for Cancer Genomics, Bethesda, MD, USA
| | - Yamini Dalal
- National Cancer Institute, Center for Cancer Research, Laboratory of Receptor Biology and Gene Expression, Bethesda, MD, USA
| |
Collapse
|
11
|
Gimenez S, Eychenne M, Legeai F, Gamble S, d'Alençon E. Towards identification of a holocentromere marker in the lepidopteran model Spodoptera frugiperda. Chromosoma 2025; 134:2. [PMID: 40067534 PMCID: PMC11897090 DOI: 10.1007/s00412-025-00828-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 01/27/2025] [Accepted: 02/12/2025] [Indexed: 03/15/2025]
Abstract
Some insects have holocentric chromosomes, with multiple kinetochores rather than a single centromere. They also lack the CENP-A and CENP-C proteins, suggesting a kinetochore assembly process different from that of monocentric chromosomes. The homolog of CENP-T was recently shown to bind silent chromatin and to play a key role in kinetochore assembly in Bombyx mori, but its role in other insects with holocentric chromosomes is unknown. We identified kinetochore genes and analyzed their expression in Spodoptera frugiperda. We silenced the kinetochore genes cenp-L, cenp-S, cenp-X and ndc80 and searched for chromosome segregation defects in Sf9 cells. All kinetochore genes except cenp-S were more strongly expressed in gonadal than in somatic tissues. Immunofluorescence microscopy and RT-qPCR demonstrated the effective silencing of the target genes by transfection with dsRNA. In Sf9 cells depleted of CENP-L and NDC80, immunofluorescence microscopy revealed increases in mitotic index and in the proportion of cells with unaligned chromosomes or multipolar spindles. The depletion of CENP-S and CENP-X had no effect on mitotic index and no division defects were observed. This suggests that CENP-L and NDC80 play key roles in chromosome segregation, whereas the functions of CENP-S and CENP-X remain unknown. We have begun to characterize the kinetochore proteins (CENP-L, CENP-S, CENP-X, NDC80), a prerequisite for holocentromere identification in S. frugiperda. This study also provides the first information about the role, in Lepidoptera, of CENP-L, a protein essential to the structure of the constitutive centromere-associated network in species with monocentric chromosomes.
Collapse
Affiliation(s)
| | | | - Fabrice Legeai
- BIPAA, IGEPP, INRAE, Institut Agro, University of Rennes, Rennes, France
- University of Rennes, INRIA, CNRS, IRISA, Rennes, France
| | - Sally Gamble
- DGIMI, Univ Montpellier, INRAE, Montpellier, France
| | | |
Collapse
|
12
|
Dudka D, Nguyen AL, Boese KG, Marescal O, Akins RB, Black BE, Cheeseman IM, Lampson MA. Adaptive evolution of CENP-T modulates centromere binding. Curr Biol 2025; 35:1012-1022.e5. [PMID: 39947176 PMCID: PMC11903153 DOI: 10.1016/j.cub.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/19/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025]
Abstract
Centromeric DNA and proteins evolve rapidly despite conserved function in mediating kinetochore-microtubule attachments during cell division. This paradox is explained by selfish DNA sequences preferentially binding centromeric proteins to disrupt attachments and bias their segregation into the egg (drive) during female meiosis. Adaptive centromeric protein evolution is predicted to prevent preferential binding to these sequences and suppress drive. Here, we test this prediction by defining the impact of adaptive evolution of the DNA-binding histone fold domain of CENP-T, a major link between centromeric DNA and microtubules. We reversed adaptive changes by creating chimeric variants of mouse CENP-T with the histone fold domain from closely related species, expressed exogenously in mouse oocytes or in a transgenic mouse model. We show that adaptive evolution of mouse CENP-T reduced centromere binding, which supports robust female gametogenesis. However, this innovation is independent of the centromeric DNA sequence, as shown by comparing the binding of divergent CENP-T variants to distinct centromere satellite arrays in mouse oocytes and in somatic cells from other species. Overall, our findings support a model in which selfish sequences drive to fixation, disrupting attachments of all centromeres to the spindle. DNA sequence-specific innovations are not needed to mitigate fitness costs in this model, so centromeric proteins adapt by modulating their binding to all centromeres in the aftermath of drive.
Collapse
Affiliation(s)
- Damian Dudka
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandra L Nguyen
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Katelyn G Boese
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Océane Marescal
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - R Brian Akins
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ben E Black
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Center for Genome Integrity, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Iain M Cheeseman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Michael A Lampson
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Center for Genome Integrity, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
13
|
Kixmoeller K, Tarasovetc EV, Mer E, Chang YW, Black BE. Centromeric chromatin clearings demarcate the site of kinetochore formation. Cell 2025; 188:1280-1296.e19. [PMID: 39855195 PMCID: PMC11890969 DOI: 10.1016/j.cell.2024.12.025] [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: 05/06/2024] [Revised: 11/24/2024] [Accepted: 12/18/2024] [Indexed: 01/27/2025]
Abstract
The centromere is the chromosomal locus that recruits the kinetochore, directing faithful propagation of the genome during cell division. Using cryo-ET on human mitotic chromosomes, we reveal a distinctive architecture at the centromere: clustered 20- to 25-nm nucleosome-associated complexes within chromatin clearings that delineate them from surrounding chromatin. Centromere components CENP-C and CENP-N are each required for the integrity of the complexes, while CENP-C is also required to maintain the chromatin clearing. We find that CENP-C is required in mitosis, not just for kinetochore assembly, likely reflecting its role in organizing the inner kinetochore during chromosome segregation. We further visualize the scaffold of the fibrous corona, a structure amplified at unattached kinetochores, revealing crescent-shaped parallel arrays of fibrils extending >1 μm. Thus, we reveal how the organization of centromeric chromatin creates a clearing at the site of kinetochore formation as well as the nature of kinetochore amplification mediated by corona fibrils.
Collapse
Affiliation(s)
- Kathryn Kixmoeller
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Biochemistry, Biophysics, Chemical Biology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA; Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ekaterina V Tarasovetc
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elie Mer
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Biochemistry, Biophysics, Chemical Biology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA; Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi-Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Biochemistry, Biophysics, Chemical Biology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA; Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Ben E Black
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Biochemistry, Biophysics, Chemical Biology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA; Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Wong LH, Tremethick DJ. Multifunctional histone variants in genome function. Nat Rev Genet 2025; 26:82-104. [PMID: 39138293 DOI: 10.1038/s41576-024-00759-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2024] [Indexed: 08/15/2024]
Abstract
Histones are integral components of eukaryotic chromatin that have a pivotal role in the organization and function of the genome. The dynamic regulation of chromatin involves the incorporation of histone variants, which can dramatically alter its structural and functional properties. Contrary to an earlier view that limited individual histone variants to specific genomic functions, new insights have revealed that histone variants exert multifaceted roles involving all aspects of genome function, from governing patterns of gene expression at precise genomic loci to participating in genome replication, repair and maintenance. This conceptual change has led to a new understanding of the intricate interplay between chromatin and DNA-dependent processes and how this connection translates into normal and abnormal cellular functions.
Collapse
Affiliation(s)
- Lee H Wong
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - David J Tremethick
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capial Territory, Australia.
| |
Collapse
|
15
|
Barrero DJ, Hedouin S, Mao Y, Asbury CL, Stergachis A, O’Toole E, Biggins S. Centromeres in the thermotolerant yeast K. marxianus mediate attachment to a single microtubule. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634737. [PMID: 39975131 PMCID: PMC11838225 DOI: 10.1101/2025.01.24.634737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Eukaryotic chromosome segregation requires spindle microtubules to attach to chromosomes through kinetochores. The chromosomal locus that mediates kinetochore assembly is the centromere and is epigenetically specified in most organisms by a centromeric histone H3 variant called CENP-A. An exception to this is budding yeast which have short, sequenced-defined point centromeres. In S. cerevisiae, a single CENP-A nucleosome is formed at the centromere and is sufficient for kinetochore assembly. The thermophilic budding yeast Kluyveromyces marxianus also has a point centromere but its length is nearly double the S. cerevisiae centromere and the number of centromeric nucleosomes and kinetochore attachment sites is unknown. Purification of native kinetochores from K. marxianus yielded a mixed population, with one subpopulation that appeared to consist of doublets, making it unclear whether K. marxianus shares the same attachment architecture as S. cerevisiae. Here, we demonstrate that though the doublet kinetochores have a functional impact on kinetochore strength, kinetochore localization throughout the cell cycle appears conserved between these two yeasts. In addition, whole spindle electron tomography demonstrates that a single microtubule binds to each chromosome. Single-molecule nucleosome mapping analysis suggests the presence of a single centromeric nucleosome. Taken together, we propose that the K. marxianus point centromere assembles a single centromeric nucleosome that mediates attachment to one microtubule.
Collapse
Affiliation(s)
- Daniel J. Barrero
- Howard Hughes Medical Institute, Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA
- Molecular and Cellular Biology Program, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Sabrine Hedouin
- Howard Hughes Medical Institute, Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA
| | - Yizi Mao
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Charles L. Asbury
- Department of Neurobiology and Biophysics, 1959 NE Pacific Street, University of Washington, Seattle, WA 98195, USA
| | - Andrew Stergachis
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Eileen O’Toole
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, CO, USA 80309 USA
| | - Sue Biggins
- Howard Hughes Medical Institute, Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA
| |
Collapse
|
16
|
Kozgunova E. Recent advances in plant kinetochore research. Front Cell Dev Biol 2025; 12:1510019. [PMID: 39911184 PMCID: PMC11794483 DOI: 10.3389/fcell.2024.1510019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
Faithful chromosome segregation is crucial for cell division in eukaryotes, facilitated by the kinetochore, a multi-subunit protein complex that connects chromosomes to the spindle microtubules. Recent research has significantly advanced our understanding of kinetochore function in plants, including surprising findings about spindle assembly checkpoint, the composition of the inner kinetochore and unique kinetochore arrangement in holocentric Cuscuta species. Additionally, some kinetochore proteins in plants have been implicated in roles beyond chromosome segregation, such as cytokinesis regulation and involvement in developmental processes. This review summarizes recent insights into plant kinetochore biology, compares plant kinetochores with those of animals and fungi, and highlights key open questions and potential future directions in the field.
Collapse
Affiliation(s)
- Elena Kozgunova
- Institute for Advanced Research, Nagoya University, Nagoya, Japan
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
17
|
Zou C, Zhang Y, Liu C, Li Y, Lin C, Chen H, Hou J, Gao G, Liu Z, Yan Q, Su W. Identification of CENPM as a key gene driving adrenocortical carcinoma metastasis via physical interaction with immune checkpoint ligand FGL1. Clin Transl Med 2025; 15:e70182. [PMID: 39778025 PMCID: PMC11707433 DOI: 10.1002/ctm2.70182] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Distant metastasis occurs in the majority of adrenocortical carcinoma (ACC), leading to an extremely poor prognosis. However, the key genes driving ACC metastasis remain unclear. METHODS Weighted gene co-expression network analysis (WGCNA) and functional enrichment analysis were conducted to identify ACC metastasis-related genes. Data from RNA-seq and microarray were analyzed to reveal correlations of the CENPM gene with cancer, metastasis, and survival in ACC. Immunohistochemistry was used to assess CENPM protein expression. The impact of CENPM on metastasis behaviour was verified in ACC (H295R and SW-13) cells and xenograft NPG mice. DIA quantitative proteomics analysis, western blot, immunofluorescence, and co-immunoprecipitation assay were performed to identify the downstream target of CENPM. RESULTS Among the 12 035 analyzed genes, 363 genes were related to ACC metastasis and CENPM was identified as the hub gene. CENPM was upregulated in ACC samples and associated with metastasis and poor prognosis. Knockdown of CENPM inhibited proliferation, invasion, and migration of ACC cells and suppressed liver metastasis in xenograft NPG mice. Collagen-containing extracellular matrix signalling was primarily downregulated when CENPM was knocked down. FGL1, important components of ECM signalling and immune checkpoint ligand of LAG3, were downregulated following CENPM silence, overexpressed in human advanced ACC samples, and colocalized with CENPM. Physical interaction between CENPM and FGL1 was identified. Overexpression of FGL1 rescued migration and invasion of CENPM knockdown ACC cells. CONCLUSIONS CENPM is a key gene in driving ACC metastasis. CENPM promotes ACC metastasis through physical interaction with the immune checkpoint ligand FGL1. CENPM can be used as a new prognostic biomarker and therapeutic target for metastatic ACC. HIGHLIGHTS CENPM is the key gene that drives ACC metastasis, and a robust biomarker for ACC prognosis. Silencing CENPM impedes ACC metastasis in vitro and in vivo by physical interaction with immune checkpoint ligand FGL1. FGL1 is overexpressed in ACC and promotes ACC metastasis.
Collapse
Affiliation(s)
- Cunru Zou
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Yu Zhang
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Chengyue Liu
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Yaxin Li
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Congjie Lin
- Department of PathologyAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Hao Chen
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| | - Jiangping Hou
- Department of OphthalmologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Guojun Gao
- Department of Urology SurgeryAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Zheng Liu
- Department of Urology SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Qiupeng Yan
- Department of Teaching and Research Section of Introduction to Basic MedicineSchool of Basic MedicineShandong Second Medical UniversityWeifangChina
- Neurologic Disorders and Regenerative Repair Lab of Shandong Higher EducationShandong Second Medical UniversityWeifangChina
| | - Wenxia Su
- Department of PhysiologySchool of Basic MedicineShandong Second Medical UniversityWeifangChina
| |
Collapse
|
18
|
Ali-Ahmad A, Mors M, Carrer M, Li X, Bilokapić S, Halić M, Cascella M, Sekulić N. Non-nucleosomal (CENP-A/H4) 2 - DNA complexes as a possible platform for centromere organization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.31.630874. [PMID: 39803555 PMCID: PMC11722257 DOI: 10.1101/2024.12.31.630874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
The centromere is a part of the chromosome that is essential for the even segregation of duplicated chromosomes during cell division. It is epigenetically defined by the presence of the histone H3 variant CENP-A. CENP-A associates specifically with a group of 16 proteins that form the centromere-associated network of proteins (CCAN). In mitosis, the kinetochore forms on the CCAN to connect the duplicated chromosomes to the microtubules protruding from the cell poles. Previous studies have shown that CENP-A replaces H3 in nucleosomes, and recently the structures of CENP-A-containing nucleosomes in complex with CCANs have been revealed, but they show only a limited interaction between CCANs and CENP-A. Here, we report the cryoEM structure of 2x(CENP-A/H4)2-di-tetramers assembled on DNA in the absence of H2A/H2B histone dimer and speculate how (CENP-A/H4)2-tetramers and -di-tetramers might serve as a platform for CCAN organization.
Collapse
Affiliation(s)
- Ahmad Ali-Ahmad
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, Faculty of Medicine, University of Oslo, Oslo 0318, Norway
| | - Mira Mors
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern 0315, Norway
| | - Manuel Carrer
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern 0315, Norway
| | - Xinmeng Li
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern 0315, Norway
| | - Silvija Bilokapić
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Mario Halić
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Michele Cascella
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern 0315, Norway
- Hylleraas Centre for Quantum Molecular Sciences, University of Oslo, P.O. Box 1033, Blindern, 0315 Oslo, Norway
| | - Nikolina Sekulić
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, Faculty of Medicine, University of Oslo, Oslo 0318, Norway
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern 0315, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
19
|
Takenoshita Y, Hara M, Nakagawa R, Ariyoshi M, Fukagawa T. Molecular details and phosphoregulation of the CENP-T-Mis12 complex interaction during mitosis in DT40 cells. iScience 2024; 27:111295. [PMID: 39628583 PMCID: PMC11612794 DOI: 10.1016/j.isci.2024.111295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/10/2024] [Accepted: 10/29/2024] [Indexed: 12/06/2024] Open
Abstract
To establish bipolar attachments of microtubules to sister chromatids, an inner kinetochore subcomplex, the constitutive centromere-associated network (CCAN), is assembled on centromeric chromatin and recruits the microtubule-binding subcomplex called the KMN network. Since CCAN proteins CENP-C and CENP-T independently bind to the Mis12 complex (Mis12C) of KMN, it is difficult to evaluate the significance of each interaction in cells. Here, we demonstrate the molecular details of the CENP-T-Mis12C interaction using chicken DT40 cells lacking the CENP-C-Mis12C interaction. Using AlphaFold predictions combined with cell biological and biochemical analyses, we identified three binding surfaces of the CENP-T-Mis12C interaction, demonstrating that each interface is important for recruiting Mis12C to CENP-T in cells. This interaction, via three interaction surfaces, is cooperatively regulated by dual phosphorylation of Dsn1 (a Mis12C component) and CENP-T, ensuring a robust CENP-T-Mis12C interaction and proper mitotic progression. These findings deepen our understanding of kinetochore assembly in cells.
Collapse
Affiliation(s)
- Yusuke Takenoshita
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masatoshi Hara
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Reiko Nakagawa
- RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, 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
|
20
|
Biggs RJ, Sundararajan K, Sun M, Hendrix E, Straight AF, Marko JF. Independence of Centromeric and Pericentromeric Chromatin Stability on CCAN Components. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.18.628896. [PMID: 39763731 PMCID: PMC11702632 DOI: 10.1101/2024.12.18.628896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
The chromatin of the centromere provides the assembly site for the mitotic kinetochore that couples microtubule attachment and force production to chromosome movement in mitosis. The chromatin of the centromere is specified by nucleosomes containing the histone H3 variant CENP-A. The constitutive centromeric-associated network (CCAN) and kinetochore are assembled on CENP-A chromatin to enable chromosome separation. CENP-A chromatin is surrounded by pericentromeric heterochromatin and bound by the sequence specific binding protein CENP-B. We performed mechanical experiments on mitotic chromosomes while tracking CENP-A and CENP-B to observe the centromere's stiffness and the role of the CCAN. We degraded CENP-C and CENP-N using auxin-inducible degrons, which we verified compromises the CCAN via observation of CENP-T loss. Chromosome stretching revealed that the CENP-A domain does not visibly stretch, even in the absence of CENP-C and/or CENP-N. Pericentromeric chromatin deforms upon force application, stretching approximately 3-fold less than the entire chromosome. CENP-C and/or CENP-N loss has no impact on pericentromere stretching. Chromosome-disconnecting nuclease treatments showed no structural effects on CENP-A. Our experiments show that the core-centromeric chromatin is more resilient and likely mechanically disconnected from the underlying pericentromeric chromatin, while the pericentric chromatin is deformable yet stiffer than the chromosome arms.
Collapse
|
21
|
Chen YC, Kilic E, Wang E, Rossman W, Suzuki A. CENcyclopedia: Dynamic Landscape of Kinetochore Architecture Throughout the Cell Cycle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.627000. [PMID: 39677682 PMCID: PMC11643120 DOI: 10.1101/2024.12.05.627000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The kinetochore, an intricate macromolecular protein complex located on chromosomes, plays a pivotal role in orchestrating chromosome segregation. It functions as a versatile platform for microtubule assembly, diligently monitors microtubule binding fidelity, and acts as a force coupler. Comprising over 100 distinct proteins, many of which exist in multiple copies, the kinetochore's composition dynamically changes throughout the cell cycle, responding to specific timing and conditions. This dynamicity is important for establishing functional kinetochores, yet the regulatory mechanisms of these dynamics have largely remained elusive. In this study, we employed advanced quantitative immunofluorescence techniques to meticulously chart the dynamics of kinetochore protein levels across the cell cycle. These findings offer a comprehensive view of the dynamic landscape of kinetochore architecture, shedding light on the detailed mechanisms of microtubule interaction and the nuanced characteristics of kinetochore proteins. This study significantly advances our understanding of the molecular coordination underlying chromosome segregation.
Collapse
Affiliation(s)
- Yu-Chia Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ece Kilic
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Evelyn Wang
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Will Rossman
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
22
|
Naish M. Bridging the gap: unravelling plant centromeres in the telomere-to-telomere era. THE NEW PHYTOLOGIST 2024; 244:2143-2149. [PMID: 39329317 PMCID: PMC11579429 DOI: 10.1111/nph.20149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/21/2024] [Indexed: 09/28/2024]
Abstract
Centromeres are specific regions of the chromosomes that play a pivotal role in the segregation of chromosomes, by facilitating the loading of the kinetochore, which forms the link between the chromosomes to the spindle fibres during cell division. In plants and animals, these regions often form megabase-scale loci of tandemly repeated DNA sequences, which have presented a challenge to genomic studies even in model species. The functional designation of centromeres is determined epigenetically by the incorporation of a centromere-specific variant of histone H3. Recent developments in long-read sequencing technology have allowed the assembly of these regions for the first time and have prompted a reassessment of fidelity of centromere function and the evolutionary dynamics of these regions.
Collapse
Affiliation(s)
- Matthew Naish
- Department of Plant SciencesUniversity of CambridgeCambridgeCB2 3EAUK
| |
Collapse
|
23
|
Bellutti L, Macaisne N, El Mossadeq L, Ganeswaran T, Canman JC, Dumont J. Regulation of outer kinetochore assembly during meiosis I and II by CENP-A and KNL-2/M18BP1 in C. elegans oocytes. Curr Biol 2024; 34:4853-4868.e6. [PMID: 39353426 PMCID: PMC11537844 DOI: 10.1016/j.cub.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/24/2024] [Accepted: 09/02/2024] [Indexed: 10/04/2024]
Abstract
During cell division, chromosomes build kinetochores that attach to spindle microtubules. Kinetochores usually form at the centromeres, which contain CENP-A nucleosomes. The outer kinetochore, which is the core attachment site for microtubules, is composed of the KMN network (Knl1c, Mis12c, and Ndc80c complexes) and is recruited downstream of CENP-A and its partner CENP-C. In C. elegans oocytes, kinetochores have been suggested to form independently of CENP-A nucleosomes. Yet kinetochore formation requires CENP-C, which acts in parallel to the nucleoporin MEL-28ELYS. Here, we used a combination of RNAi and Degron-based depletion of CENP-A (or downstream CENP-C) to demonstrate that both proteins are in fact responsible for a portion of outer kinetochore assembly during meiosis I and are essential for accurate chromosome segregation. The remaining part requires the coordinated action of KNL-2 (ortholog of human M18BP1) and of the nucleoporin MEL-28ELYS. Accordingly, co-depletion of CENP-A (or CENP-C) and KNL-2M18BP1 (or MEL-28ELYS) prevented outer kinetochore assembly in oocytes during meiosis I. We further found that KNL-2M18BP1 and MEL-28ELYS are interdependent for kinetochore localization. Using engineered mutants, we demonstrated that KNL-2M18BP1 recruits MEL-28ELYS at meiotic kinetochores through a specific N-terminal domain, independently of its canonical CENP-A loading factor activity. Finally, we found that meiosis II outer kinetochore assembly was solely dependent on the canonical CENP-A/CENP-C pathway. Thus, like in most cells, outer kinetochore assembly in C. elegans oocytes depends on centromeric chromatin. However, during meiosis I, an additional KNL-2M18BP1 and MEL-28ELYS pathway acts in a non-redundant manner and in parallel to canonical centromeric chromatin.
Collapse
Affiliation(s)
- Laura Bellutti
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Nicolas Macaisne
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Layla El Mossadeq
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | | | - Julie C Canman
- Columbia University, Irving Medical Center, Department of Pathology and Cell Biology, New York, NY 10032, USA
| | - Julien Dumont
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France.
| |
Collapse
|
24
|
Kang Z, Li R, Liu C, Dong X, Hu Y, Xu L, Liu X, Xiang Y, Gao L, Si W, Wang L, Li Q, Zhang L, Wang H, Yang X, Liu J. m 6A-modified cenRNA stabilizes CENPA to ensure centromere integrity in cancer cells. Cell 2024; 187:6035-6054.e27. [PMID: 39305902 DOI: 10.1016/j.cell.2024.08.040] [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: 12/12/2023] [Revised: 01/10/2024] [Accepted: 08/20/2024] [Indexed: 10/20/2024]
Abstract
m6A modification is best known for its critical role in controlling multiple post-transcriptional processes of the mRNAs. Here, we discovered elevated levels of m6A modification on centromeric RNA (cenRNA) in cancerous cells compared with non-cancerous cells. We then identified CENPA, an H3 variant, as an m6A reader of cenRNA. CENPA is localized at centromeres and is essential in preserving centromere integrity and function during mitosis. The m6A-modified cenRNA stabilizes centromeric localization of CENPA in cancer cells during the S phase of the cell cycle. Mutations of CENPA at the Leu61 and the Arg63 or removal of cenRNA m6A modification lead to loss of centromere-bound CENPA during S phase. This in turn results in compromised centromere integrity and abnormal chromosome separation and hinders cancer cell proliferation and tumor growth. Our findings unveil an m6A reading mechanism by CENPA that epigenetically governs centromere integrity in cancer cells, providing potential targets for cancer therapy.
Collapse
Affiliation(s)
- Zihong Kang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China
| | - Ruimeng Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Science, Tsinghua University, 100084 Beijing, China
| | - Chang Liu
- Department of Genetics, Stanford University, School of Medicine, Stanford, CA, USA
| | - Xiaozhe Dong
- College of Chemistry and Molecular Engineering, Peking University, 100871 Beijing, China
| | - Yuxuan Hu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 211198 Nanjing, China
| | - Lei Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008 Nanjing, China
| | - Xinyu Liu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China
| | - Yunfan Xiang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China
| | - Liming Gao
- School of Science, China Pharmaceutical University, 211198 Nanjing, China
| | - Wenzhe Si
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Laboratory Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Lei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008 Nanjing, China
| | - Qing Li
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China
| | - Liang Zhang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 310022 Hangzhou, China
| | - Huan Wang
- College of Chemistry and Molecular Engineering, Peking University, 100871 Beijing, China
| | - Xuerui Yang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Science, Tsinghua University, 100084 Beijing, China.
| | - Jun Liu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China.
| |
Collapse
|
25
|
Karami Fath M, Nazari A, Parsania N, Behboodi P, Ketabi SS, Razmjouei P, Farzam F, Shafagh SG, Nabi Afjadi M. Centromeres in cancer: Unraveling the link between chromosomal instability and tumorigenesis. Med Oncol 2024; 41:254. [PMID: 39352464 DOI: 10.1007/s12032-024-02524-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/23/2024] [Indexed: 11/14/2024]
Abstract
Centromeres are critical structures involved in chromosome segregation, maintaining genomic stability, and facilitating the accurate transmission of genetic information. They are key in coordinating the assembly and help keep the correct structure, location, and function of the kinetochore, a proteinaceous structure vital for ensuring proper chromosome segregation during cell division. Abnormalities in centromere structure can lead to aneuploidy or chromosomal instability, which have been implicated in various diseases, including cancer. Accordingly, abnormalities in centromeres, such as structural rearrangements and dysregulation of centromere-associated proteins, disrupt gene function, leading to uncontrolled cell growth and tumor progression. For instance, altered expression of CENP-A, CENP-E, and others such as BUB1, BUBR1, MAD1, and INCENP, have been shown to ascribe to centromere over-amplification, chromosome missegregation, aneuploidy, and chromosomal instability; this, in turn, can culminate in tumor progression. These centromere abnormalities also promoted tumor heterogeneity by generating genetically diverse cell populations within tumors. Advanced techniques like fluorescence in situ hybridization (FISH) and chromosomal microarray analysis are crucial for detecting centromere abnormalities, enabling accurate cancer classification and tailored treatment strategies. Researchers are exploring strategies to disrupt centromere-associated proteins for targeted cancer therapies. Thus, this review explores centromere abnormalities in cancer, their molecular mechanisms, diagnostic implications, and therapeutic targeting. It aims to advance our understanding of centromeres' role in cancer and develop advanced diagnostic tools and targeted therapies for improved cancer management and treatment.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Ahmad Nazari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Noushin Parsania
- Department of Brain and Cognitive Sciences, Cell Science Research Center, ROYAN Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Paria Behboodi
- Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | - Pegah Razmjouei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnoosh Farzam
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
26
|
de Lima LG, Guarracino A, Koren S, Potapova T, McKinney S, Rhie A, Solar SJ, Seidel C, Fagen B, Walenz BP, Bouffard GG, Brooks SY, Peterson M, Hall K, Crawford J, Young AC, Pickett BD, Garrison E, Phillippy AM, Gerton JL. The formation and propagation of human Robertsonian chromosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614821. [PMID: 39386535 PMCID: PMC11463614 DOI: 10.1101/2024.09.24.614821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Robertsonian chromosomes are a type of variant chromosome found commonly in nature. Present in one in 800 humans, these chromosomes can underlie infertility, trisomies, and increased cancer incidence. Recognized cytogenetically for more than a century, their origins have remained mysterious. Recent advances in genomics allowed us to assemble three human Robertsonian chromosomes completely. We identify a common breakpoint and epigenetic changes in centromeres that provide insight into the formation and propagation of common Robertsonian translocations. Further investigation of the assembled genomes of chimpanzee and bonobo highlights the structural features of the human genome that uniquely enable the specific crossover event that creates these chromosomes. Resolving the structure and epigenetic features of human Robertsonian chromosomes at a molecular level paves the way to understanding how chromosomal structural variation occurs more generally, and how chromosomes evolve.
Collapse
Affiliation(s)
| | - Andrea Guarracino
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sergey Koren
- Genome Informatics Section, Center for Genomics and Data Science Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tamara Potapova
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Sean McKinney
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Arang Rhie
- Genome Informatics Section, Center for Genomics and Data Science Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Steven J Solar
- Genome Informatics Section, Center for Genomics and Data Science Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Chris Seidel
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Brandon Fagen
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Brian P Walenz
- Genome Informatics Section, Center for Genomics and Data Science Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gerard G Bouffard
- NIH Intramural Sequencing Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shelise Y Brooks
- NIH Intramural Sequencing Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Kate Hall
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Juyun Crawford
- Genome Informatics Section, Center for Genomics and Data Science Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alice C Young
- NIH Intramural Sequencing Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brandon D Pickett
- Genome Informatics Section, Center for Genomics and Data Science Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Erik Garrison
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Adam M Phillippy
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
- Genome Informatics Section, Center for Genomics and Data Science Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- NIH Intramural Sequencing Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
27
|
Barrero DJ, Wijeratne SS, Zhao X, Cunningham GF, Yan R, Nelson CR, Arimura Y, Funabiki H, Asbury CL, Yu Z, Subramanian R, Biggins S. Architecture of native kinetochores revealed by structural studies utilizing a thermophilic yeast. Curr Biol 2024; 34:3881-3893.e5. [PMID: 39127048 PMCID: PMC11387133 DOI: 10.1016/j.cub.2024.07.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/30/2024] [Accepted: 07/08/2024] [Indexed: 08/12/2024]
Abstract
Eukaryotic chromosome segregation requires kinetochores, multi-megadalton protein machines that assemble on the centromeres of chromosomes and mediate attachments to dynamic spindle microtubules. Kinetochores are built from numerous complexes, and there has been progress in structural studies on recombinant subassemblies. However, there is limited structural information on native kinetochore architecture. To address this, we purified functional, native kinetochores from the thermophilic yeast Kluyveromyces marxianus and examined them by electron microscopy (EM), cryoelectron tomography (cryo-ET), and atomic force microscopy (AFM). The kinetochores are extremely large, flexible assemblies that exhibit features consistent with prior models. We assigned kinetochore polarity by visualizing their interactions with microtubules and locating the microtubule binder, Ndc80c. This work shows that isolated kinetochores are more dynamic and complex than what might be anticipated based on the known structures of recombinant subassemblies and provides the foundation to study the global architecture and functions of kinetochores at a structural level.
Collapse
Affiliation(s)
- Daniel J Barrero
- Howard Hughes Medical Institute, Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Sithara S Wijeratne
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Xiaowei Zhao
- Howard Hughes Medical Institute Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Grace F Cunningham
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Rui Yan
- Howard Hughes Medical Institute Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Christian R Nelson
- Howard Hughes Medical Institute, Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA
| | - Yasuhiro Arimura
- The Rockefeller University, 1230 York Ave., New York, NY 10065, USA
| | | | - Charles L Asbury
- Department of Physiology and Biophysics, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Zhiheng Yu
- Howard Hughes Medical Institute Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Radhika Subramanian
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Sue Biggins
- Howard Hughes Medical Institute, Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA.
| |
Collapse
|
28
|
Salinas-Luypaert C, Fachinetti D. Canonical and noncanonical regulators of centromere assembly and maintenance. Curr Opin Cell Biol 2024; 89:102396. [PMID: 38981198 DOI: 10.1016/j.ceb.2024.102396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/10/2024] [Accepted: 06/15/2024] [Indexed: 07/11/2024]
Abstract
Centromeres are specialized chromosomal domains where the kinetochores assemble during cell division to ensure accurate transmission of the genetic information to the two daughter cells. The centromeric function is evolutionary conserved and, in most organisms, centromeres are epigenetically defined by a unique chromatin containing the histone H3 variant CENP-A. The canonical regulators of CENP-A assembly and maintenance are well-known, yet some of the molecular mechanisms regulating this complex process have only recently been unveiled. We review the most recent advances on the topic, including the emergence of new and unexpected factors that favor and regulate CENP-A assembly and/or maintenance.
Collapse
Affiliation(s)
- Catalina Salinas-Luypaert
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR 144 & UMR3664, 26 rue d'Ulm, 75005, Paris, France.
| | - Daniele Fachinetti
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR 144 & UMR3664, 26 rue d'Ulm, 75005, Paris, France.
| |
Collapse
|
29
|
Andrade Ruiz L, Kops GJPL, Sacristan C. Vertebrate centromere architecture: from chromatin threads to functional structures. Chromosoma 2024; 133:169-181. [PMID: 38856923 PMCID: PMC11266386 DOI: 10.1007/s00412-024-00823-z] [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: 01/06/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 06/11/2024]
Abstract
Centromeres are chromatin structures specialized in sister chromatid cohesion, kinetochore assembly, and microtubule attachment during chromosome segregation. The regional centromere of vertebrates consists of long regions of highly repetitive sequences occupied by the Histone H3 variant CENP-A, and which are flanked by pericentromeres. The three-dimensional organization of centromeric chromatin is paramount for its functionality and its ability to withstand spindle forces. Alongside CENP-A, key contributors to the folding of this structure include components of the Constitutive Centromere-Associated Network (CCAN), the protein CENP-B, and condensin and cohesin complexes. Despite its importance, the intricate architecture of the regional centromere of vertebrates remains largely unknown. Recent advancements in long-read sequencing, super-resolution and cryo-electron microscopy, and chromosome conformation capture techniques have significantly improved our understanding of this structure at various levels, from the linear arrangement of centromeric sequences and their epigenetic landscape to their higher-order compaction. In this review, we discuss the latest insights on centromere organization and place them in the context of recent findings describing a bipartite higher-order organization of the centromere.
Collapse
Affiliation(s)
- Lorena Andrade Ruiz
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, Netherlands
- University Medical Center Utrecht, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Geert J P L Kops
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, Netherlands
- University Medical Center Utrecht, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Carlos Sacristan
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, Netherlands.
- University Medical Center Utrecht, Utrecht, Netherlands.
- Oncode Institute, Utrecht, Netherlands.
| |
Collapse
|
30
|
Yan L, Yuan X, Liu M, Chen Q, Zhang M, Xu J, Zeng LH, Zhang L, Huang J, Lu W, He X, Yan H, Wang F. A non-canonical role of the inner kinetochore in regulating sister-chromatid cohesion at centromeres. EMBO J 2024; 43:2424-2452. [PMID: 38714893 PMCID: PMC11182772 DOI: 10.1038/s44318-024-00104-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/27/2024] [Accepted: 04/12/2024] [Indexed: 06/19/2024] Open
Abstract
The 16-subunit Constitutive Centromere-associated Network (CCAN)-based inner kinetochore is well-known for connecting centromeric chromatin to the spindle-binding outer kinetochore. Here, we report a non-canonical role for the inner kinetochore in directly regulating sister-chromatid cohesion at centromeres. We provide biochemical, X-ray crystal structure, and intracellular ectopic localization evidence that the inner kinetochore directly binds cohesin, a ring-shaped multi-subunit complex that holds sister chromatids together from S-phase until anaphase onset. This interaction is mediated by binding of the 5-subunit CENP-OPQUR sub-complex of CCAN to the Scc1-SA2 sub-complex of cohesin. Mutation in the CENP-U subunit of the CENP-OPQUR complex that abolishes its binding to the composite interface between Scc1 and SA2 weakens centromeric cohesion, leading to premature separation of sister chromatids during delayed metaphase. We further show that CENP-U competes with the cohesin release factor Wapl for binding the interface of Scc1-SA2, and that the cohesion-protecting role for CENP-U can be bypassed by depleting Wapl. Taken together, this study reveals an inner kinetochore-bound pool of cohesin, which strengthens centromeric sister-chromatid cohesion to resist metaphase spindle pulling forces.
Collapse
Affiliation(s)
- Lu Yan
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China
| | - Xueying Yuan
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China
| | - Mingjie Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qinfu Chen
- Department of Gynecological Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Miao Zhang
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China
| | - Junfen Xu
- Department of Gynecological Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Long Zhang
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Jun Huang
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Weiguo Lu
- Department of Gynecological Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Xiaojing He
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Haiyan Yan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| | - Fangwei Wang
- Life Sciences Institute, State Key Laboratory of Transvascular Implantation Devices of the Second Affiliated Hospital of Zhejiang University School of Medicine, MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang University, Hangzhou, 310058, China.
- Department of Gynecological Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
- Zhejiang Provincial Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
31
|
Polley S, Raisch T, Ghetti S, Körner M, Terbeck M, Gräter F, Raunser S, Aponte-Santamaría C, Vetter IR, Musacchio A. Structure of the human KMN complex and implications for regulation of its assembly. Nat Struct Mol Biol 2024; 31:861-873. [PMID: 38459128 PMCID: PMC11189300 DOI: 10.1038/s41594-024-01230-9] [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: 08/07/2023] [Accepted: 01/19/2024] [Indexed: 03/10/2024]
Abstract
Biorientation of chromosomes during cell division is necessary for precise dispatching of a mother cell's chromosomes into its two daughters. Kinetochores, large layered structures built on specialized chromosome loci named centromeres, promote biorientation by binding and sensing spindle microtubules. One of the outer layer main components is a ten-subunit assembly comprising Knl1C, Mis12C and Ndc80C (KMN) subcomplexes. The KMN is highly elongated and docks on kinetochores and microtubules through interfaces at its opposite extremes. Here, we combine cryogenic electron microscopy reconstructions and AlphaFold2 predictions to generate a model of the human KMN that reveals all intra-KMN interfaces. We identify and functionally validate two interaction interfaces that link Mis12C to Ndc80C and Knl1C. Through targeted interference experiments, we demonstrate that this mutual organization strongly stabilizes the KMN assembly. Our work thus reports a comprehensive structural and functional analysis of this part of the kinetochore microtubule-binding machinery and elucidates the path of connections from the chromatin-bound components to the force-generating components.
Collapse
Affiliation(s)
- Soumitra Polley
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Tobias Raisch
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Sabrina Ghetti
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Marie Körner
- Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Melina Terbeck
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Frauke Gräter
- Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
- Max Planck School Matter to Life, Heidelberg, Germany
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | | | - Ingrid R Vetter
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Andrea Musacchio
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
- Max Planck School Matter to Life, Heidelberg, Germany.
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
32
|
Parl FF. Analysis of CENP-B Boxes as Anchor of Kinetochores in Centromeres of Human Chromosomes. Bioinform Biol Insights 2024; 18:11779322241248913. [PMID: 38690324 PMCID: PMC11060027 DOI: 10.1177/11779322241248913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/04/2024] [Indexed: 05/02/2024] Open
Abstract
The kinetochore is a multiprotein structure that attaches at one end to DNA in the centromere and at the other end to microtubules in the mitotic spindle. By connecting centromere and spindle, the kinetochore controls the migration of chromosomes during cell division. The exact position where the kinetochore assembles on each centromere was uncertain because large sections of centromeric DNA had not been sequenced due to highly repetitive alpha-satellite arrays. Embedded in the arrays is a 17 bp consensus sequence, the so-called CENP-B box, which binds the CENP-B protein, the only protein that binds directly to centromeric DNA. Recently, the Telomere-to-Telomere Consortium published the complete centromeric DNA sequences of all chromosomes including their epigenetic modifications in the T2T-CHM13 map. I used data from the T2T-CHM13 map to locate the CENP-B boxes in the centromeres as anchor of kinetochores. Most of the CENP-B boxes in centromeric DNA are methylated with the exception of the so-called centromere dip region (CDR), where CENP-B protein dimers bind to adjacent unmethylated CENP-B boxes and interact with CENP-A and CENP-C proteins to assemble the kinetochore. The centromeres of all chromosomes combined have a size of 407 Mb of which the kinetochores account for 5.0 Mb or 1.2%. There is no correlation between centromere and kinetochore size (P = .77). While the number of CENP-B boxes varies 4-fold between chromosomes, their density (number/Kb) varies less than 2-fold with a mean of 2.61 ± 0.33. The narrow range ensures a uniform pull of the spindle on the centromeres. I illustrate the findings in a model of the human kinetochore anchored at unmethylated CENP-B boxes in the CDR and present circos plots of chromosomes to show the location of kinetochores in their respective centromeres.
Collapse
Affiliation(s)
- Fritz F Parl
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
33
|
Kixmoeller K, Chang YW, Black BE. Centromeric chromatin clearings demarcate the site of kinetochore formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591177. [PMID: 38712116 PMCID: PMC11071481 DOI: 10.1101/2024.04.26.591177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The centromere is the chromosomal locus that recruits the kinetochore, directing faithful propagation of the genome during cell division. The kinetochore has been interrogated by electron microscopy since the middle of the last century, but with methodologies that compromised fine structure. Using cryo-ET on human mitotic chromosomes, we reveal a distinctive architecture at the centromere: clustered 20-25 nm nucleosome-associated complexes within chromatin clearings that delineate them from surrounding chromatin. Centromere components CENP-C and CENP-N are each required for the integrity of the complexes, while CENP-C is also required to maintain the chromatin clearing. We further visualize the scaffold of the fibrous corona, a structure amplified at unattached kinetochores, revealing crescent-shaped parallel arrays of fibrils that extend >1 μm. Thus, we reveal how the organization of centromeric chromatin creates a clearing at the site of kinetochore formation as well as the nature of kinetochore amplification mediated by corona fibrils.
Collapse
Affiliation(s)
- Kathryn Kixmoeller
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, PA, USA
- Biochemistry Biophysics Chemical Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, PA, USA
- Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, PA, USA
- Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, PA, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Yi-Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, PA, USA
- Biochemistry Biophysics Chemical Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, PA, USA
- Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Ben E. Black
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, PA, USA
- Biochemistry Biophysics Chemical Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, PA, USA
- Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, PA, USA
- Penn Center for Genome Integrity, Perelman School of Medicine, University of Pennsylvania, PA, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, PA, USA
| |
Collapse
|
34
|
Wu F, Akbar H, Wang C, Yuan X, Dou Z, Mullen M, Niu L, Zhang L, Zang J, Wang Z, Yao X, Song X, Liu X. Sgo1 interacts with CENP-A to guide accurate chromosome segregation in mitosis. J Mol Cell Biol 2024; 15:mjad061. [PMID: 37777834 PMCID: PMC11181942 DOI: 10.1093/jmcb/mjad061] [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: 09/05/2022] [Revised: 02/21/2023] [Accepted: 09/29/2023] [Indexed: 10/02/2023] Open
Abstract
Shugoshin-1 (Sgo1) is necessary for maintaining sister centromere cohesion and ensuring accurate chromosome segregation during mitosis. It has been reported that the localization of Sgo1 at the centromere is dependent on Bub1-mediated phosphorylation of histone H2A at T120. However, it remains uncertain whether other centromeric proteins play a role in regulating the localization and function of Sgo1 during mitosis. Here, we show that CENP-A interacts with Sgo1 and determines the localization of Sgo1 to the centromere during mitosis. Further biochemical characterization revealed that lysine and arginine residues in the C-terminal domain of Sgo1 are critical for binding CENP-A. Interestingly, the replacement of these basic amino acids with acidic amino acids perturbed the localization of Sgo1 and Aurora B to the centromere, resulting in aberrant chromosome segregation and premature chromatid separation. Taken together, these findings reveal a previously unrecognized but direct link between Sgo1 and CENP-A in centromere plasticity control and illustrate how the Sgo1-CENP-A interaction guides accurate cell division.
Collapse
Affiliation(s)
- Fengge Wu
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Hameed Akbar
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Chunyue Wang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Xiao Yuan
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| | - Zhen Dou
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
- Keck Center for Cellular Dynamics, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - McKay Mullen
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Keck Center for Cellular Dynamics, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Liwen Niu
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| | - Liang Zhang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Jianye Zang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Zhikai Wang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| | - Xiaoyu Song
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| |
Collapse
|
35
|
Dou Z, Liu R, Gui P, Fu C, Lippincott-Schwartz J, Yao X, Liu X. Fluorescence complementation-based FRET imaging reveals centromere assembly dynamics. Mol Biol Cell 2024; 35:ar51. [PMID: 38381564 PMCID: PMC11064673 DOI: 10.1091/mbc.e23-09-0379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/12/2024] [Accepted: 02/16/2024] [Indexed: 02/23/2024] Open
Abstract
Visualization of specific molecules and their assembly in real time and space is essential to delineate how cellular dynamics and signaling circuit are orchestrated during cell division cycle. Our recent studies reveal structural insights into human centromere-kinetochore core CCAN complex. Here we introduce a method for optically imaging trimeric and tetrameric protein interactions at nanometer spatial resolution in live cells using fluorescence complementation-based Förster resonance energy transfer (FC-FRET). Complementary fluorescent protein molecules were first used to visualize dimerization followed by FRET measurements. Using FC-FRET, we visualized centromere CENP-SXTW tetramer assembly dynamics in live cells, and dimeric interactions between CENP-TW dimer and kinetochore protein Spc24/25 dimer in dividing cells. We further delineated the interactions of monomeric CENP-T with Spc24/25 dimer in dividing cells. Surprisingly, our analyses revealed critical role of CDK1 kinase activity in the initial recruitment of Spc24/25 by CENP-T. However, interactions between CENP-T and Spc24/25 during chromosome segregation is independent of CDK1. Thus, FC-FRET provides a unique approach to delineate spatiotemporal dynamics of trimerized and tetramerized proteins at nanometer scale and establishes a platform to report the precise regulation of multimeric protein interactions in space and time in live cells.
Collapse
Affiliation(s)
- Zhen Dou
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Cross-disciplinary Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Ran Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Cross-disciplinary Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Ping Gui
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Cross-disciplinary Sciences, University of Science and Technology of China, Hefei 230027, China
- Molecular Imaging Center, Morehouse School of Medicine, Atlanta, GA 30310
- Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Chuanhai Fu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Cross-disciplinary Sciences, University of Science and Technology of China, Hefei 230027, China
| | | | - Xuebiao Yao
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Cross-disciplinary Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Xing Liu
- MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Hefei National Center for Cross-disciplinary Sciences, University of Science and Technology of China, Hefei 230027, China
- Molecular Imaging Center, Morehouse School of Medicine, Atlanta, GA 30310
| |
Collapse
|
36
|
Naish M, Henderson IR. The structure, function, and evolution of plant centromeres. Genome Res 2024; 34:161-178. [PMID: 38485193 PMCID: PMC10984392 DOI: 10.1101/gr.278409.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Centromeres are essential regions of eukaryotic chromosomes responsible for the formation of kinetochore complexes, which connect to spindle microtubules during cell division. Notably, although centromeres maintain a conserved function in chromosome segregation, the underlying DNA sequences are diverse both within and between species and are predominantly repetitive in nature. The repeat content of centromeres includes high-copy tandem repeats (satellites), and/or specific families of transposons. The functional region of the centromere is defined by loading of a specific histone 3 variant (CENH3), which nucleates the kinetochore and shows dynamic regulation. In many plants, the centromeres are composed of satellite repeat arrays that are densely DNA methylated and invaded by centrophilic retrotransposons. In some cases, the retrotransposons become the sites of CENH3 loading. We review the structure of plant centromeres, including monocentric, holocentric, and metapolycentric architectures, which vary in the number and distribution of kinetochore attachment sites along chromosomes. We discuss how variation in CENH3 loading can drive genome elimination during early cell divisions of plant embryogenesis. We review how epigenetic state may influence centromere identity and discuss evolutionary models that seek to explain the paradoxically rapid change of centromere sequences observed across species, including the potential roles of recombination. We outline putative modes of selection that could act within the centromeres, as well as the role of repeats in driving cycles of centromere evolution. Although our primary focus is on plant genomes, we draw comparisons with animal and fungal centromeres to derive a eukaryote-wide perspective of centromere structure and function.
Collapse
Affiliation(s)
- Matthew Naish
- Department of Plant Sciences, University of Cambridge, Cambridge CB2 3EA, United Kingdom
| | - Ian R Henderson
- Department of Plant Sciences, University of Cambridge, Cambridge CB2 3EA, United Kingdom
| |
Collapse
|
37
|
Barrero DJ, Wijeratne SS, Zhao X, Cunningham GF, Rui Y, Nelson CR, Yasuhiro A, Funabiki H, Asbury CL, Yu Z, Subramanian R, Biggins S. Architecture and flexibility of native kinetochores revealed by structural studies utilizing a thermophilic yeast. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582571. [PMID: 38464254 PMCID: PMC10925344 DOI: 10.1101/2024.02.28.582571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Eukaryotic chromosome segregation requires kinetochores, multi-megadalton protein machines that assemble on the centromeres of chromosomes and mediate attachments to dynamic spindle microtubules. Kinetochores are built from numerous complexes, and understanding how they are arranged is key to understanding how kinetochores perform their multiple functions. However, an integrated understanding of kinetochore architecture has not yet been established. To address this, we purified functional, native kinetochores from Kluyveromyces marxianus and examined them by electron microscopy, cryo-electron tomography and atomic force microscopy. The kinetochores are extremely large, flexible assemblies that exhibit features consistent with prior models. We assigned kinetochore polarity by visualizing their interactions with microtubules and locating the microtubule binder Ndc80c. This work shows that isolated kinetochores are more dynamic and complex than what might be anticipated based on the known structures of recombinant subassemblies, and provides the foundation to study the global architecture and functions of kinetochores at a structural level.
Collapse
Affiliation(s)
- Daniel J. Barrero
- Howard Hughes Medical Institute, Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA
- Molecular and Cellular Biology Program, University of Washington, 1705 NE Pacific Street, Seattle, WA 98195, USA
| | - Sithara S. Wijeratne
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaowei Zhao
- Howard Hughes Medical Institute Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Grace F. Cunningham
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Yan Rui
- Howard Hughes Medical Institute Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Christian R. Nelson
- Howard Hughes Medical Institute, Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA
| | - Arimura Yasuhiro
- The Rockefeller University, 1230 York Ave., New York, NY 10065, USA
| | | | - Charles L. Asbury
- Department of Physiology and Biophysics, 1959 NE Pacific Street, University of Washington, Seattle, WA 98195, USA
| | - Zhiheng Yu
- Howard Hughes Medical Institute Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Radhika Subramanian
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Sue Biggins
- Howard Hughes Medical Institute, Division of Basic Sciences, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA
| |
Collapse
|
38
|
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
|
39
|
Sissoko GB, Tarasovetc EV, Marescal O, Grishchuk EL, Cheeseman IM. Higher-order protein assembly controls kinetochore formation. Nat Cell Biol 2024; 26:45-56. [PMID: 38168769 PMCID: PMC10842828 DOI: 10.1038/s41556-023-01313-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 11/13/2023] [Indexed: 01/05/2024]
Abstract
To faithfully segregate chromosomes during vertebrate mitosis, kinetochore-microtubule interactions must be restricted to a single site on each chromosome. Prior work on pair-wise kinetochore protein interactions has been unable to identify the mechanisms that prevent outer kinetochore formation in regions with a low density of CENP-A nucleosomes. To investigate the impact of higher-order assembly on kinetochore formation, we generated oligomers of the inner kinetochore protein CENP-T using two distinct, genetically engineered systems in human cells. Although individual CENP-T molecules interact poorly with outer kinetochore proteins, oligomers that mimic centromeric CENP-T density trigger the robust formation of functional, cytoplasmic kinetochore-like particles. Both in cells and in vitro, each molecule of oligomerized CENP-T recruits substantially higher levels of outer kinetochore components than monomeric CENP-T molecules. Our work suggests that the density dependence of CENP-T restricts outer kinetochore recruitment to centromeres, where densely packed CENP-A recruits a high local concentration of inner kinetochore proteins.
Collapse
Affiliation(s)
- Gunter B Sissoko
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ekaterina V Tarasovetc
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Océane Marescal
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ekaterina L Grishchuk
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
40
|
Favasuli VK, Ronchetti D, Silvestris I, Puccio N, Fabbiano G, Traini V, Todoerti K, Erratico S, Ciarrocchi A, Fragliasso V, Giannandrea D, Tumiatti F, Chiaramonte R, Torrente Y, Finelli P, Morelli E, Munshi NC, Bolli N, Neri A, Taìana E. DIS3 depletion in multiple myeloma causes extensive perturbation in cell cycle progression and centrosome amplification. Haematologica 2024; 109:231-244. [PMID: 37439377 PMCID: PMC10772536 DOI: 10.3324/haematol.2023.283274] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023] Open
Abstract
DIS3 gene mutations occur in approximately 10% of patients with multiple myeloma (MM); furthermore, DIS3 expression can be affected by monosomy 13 and del(13q), found in roughly 40% of MM cases. Despite the high incidence of DIS3 mutations and deletions, the biological significance of DIS3 and its contribution to MM pathogenesis remain poorly understood. In this study we investigated the functional role of DIS3 in MM, by exploiting a loss-of-function approach in human MM cell lines. We found that DIS3 knockdown inhibits proliferation in MM cell lines and largely affects cell cycle progression of MM plasma cells, ultimately inducing a significant increase in the percentage of cells in the G0/G1 phase and a decrease in the S and G2/M phases. DIS3 plays an important role not only in the control of the MM plasma cell cycle, but also in the centrosome duplication cycle, which are strictly co-regulated in physiological conditions in the G1 phase. Indeed, DIS3 silencing leads to the formation of supernumerary centrosomes accompanied by the assembly of multipolar spindles during mitosis. In MM, centrosome amplification is present in about a third of patients and may represent a mechanism leading to genomic instability. These findings strongly prompt further studies investigating the relevance of DIS3 in the centrosome duplication process. Indeed, a combination of DIS3 defects and deficient spindle-assembly checkpoint can allow cells to progress through the cell cycle without proper chromosome segregation, generating aneuploid cells which ultimately lead to the development of MM.
Collapse
Affiliation(s)
- Vanessa K Favasuli
- Department of Oncology and Hemato-oncology, University of Milan, Italy; Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
| | | | | | - Noemi Puccio
- Laboratory of Translational Research, Azienda USL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy; Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, 41121
| | - Giuseppina Fabbiano
- Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan
| | - Valentina Traini
- Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan
| | - Katia Todoerti
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - Silvia Erratico
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, University of Milan, Centro Dino Ferrari, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; Novystem Spa, Milan
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL-IRCCS Reggio Emilia, 42123 Reggio Emilia
| | - Valentina Fragliasso
- Laboratory of Translational Research, Azienda USL-IRCCS Reggio Emilia, 42123 Reggio Emilia
| | | | - Francesca Tumiatti
- Medical Genetics Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan
| | | | - Yvan Torrente
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, University of Milan, Centro Dino Ferrari, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan
| | - Palma Finelli
- Medical Genetics Laboratory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate, 20090 Milan
| | - Eugenio Morelli
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
| | - Nikhil C Munshi
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
| | - Niccolò Bolli
- Department of Oncology and Hemato-oncology, University of Milan, Italy; Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan
| | - Antonino Neri
- Scientific Directorate, Azienda USL-IRCCS Reggio Emilia, 42123 Reggio Emilia.
| | - Elisa Taìana
- Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan
| |
Collapse
|
41
|
Xu R, Pan Z, Nakagawa T. Gross Chromosomal Rearrangement at Centromeres. Biomolecules 2023; 14:28. [PMID: 38254628 PMCID: PMC10813616 DOI: 10.3390/biom14010028] [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: 11/28/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Centromeres play essential roles in the faithful segregation of chromosomes. CENP-A, the centromere-specific histone H3 variant, and heterochromatin characterized by di- or tri-methylation of histone H3 9th lysine (H3K9) are the hallmarks of centromere chromatin. Contrary to the epigenetic marks, DNA sequences underlying the centromere region of chromosomes are not well conserved through evolution. However, centromeres consist of repetitive sequences in many eukaryotes, including animals, plants, and a subset of fungi, including fission yeast. Advances in long-read sequencing techniques have uncovered the complete sequence of human centromeres containing more than thousands of alpha satellite repeats and other types of repetitive sequences. Not only tandem but also inverted repeats are present at a centromere. DNA recombination between centromere repeats can result in gross chromosomal rearrangement (GCR), such as translocation and isochromosome formation. CENP-A chromatin and heterochromatin suppress the centromeric GCR. The key player of homologous recombination, Rad51, safeguards centromere integrity through conservative noncrossover recombination between centromere repeats. In contrast to Rad51-dependent recombination, Rad52-mediated single-strand annealing (SSA) and microhomology-mediated end-joining (MMEJ) lead to centromeric GCR. This review summarizes recent findings on the role of centromere and recombination proteins in maintaining centromere integrity and discusses how GCR occurs at centromeres.
Collapse
Affiliation(s)
- Ran Xu
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Ziyi Pan
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Takuro Nakagawa
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| |
Collapse
|
42
|
Nagpal H, Ali-Ahmad A, Hirano Y, Cai W, Halic M, Fukagawa T, Sekulić N, Fierz B. CENP-A and CENP-B collaborate to create an open centromeric chromatin state. Nat Commun 2023; 14:8227. [PMID: 38086807 PMCID: PMC10716449 DOI: 10.1038/s41467-023-43739-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Centromeres are epigenetically defined via the presence of the histone H3 variant CENP-A. Contacting CENP-A nucleosomes, the constitutive centromere associated network (CCAN) and the kinetochore assemble, connecting the centromere to spindle microtubules during cell division. The DNA-binding centromeric protein CENP-B is involved in maintaining centromere stability and, together with CENP-A, shapes the centromeric chromatin state. The nanoscale organization of centromeric chromatin is not well understood. Here, we use single-molecule fluorescence and cryoelectron microscopy (cryoEM) to show that CENP-A incorporation establishes a dynamic and open chromatin state. The increased dynamics of CENP-A chromatin create an opening for CENP-B DNA access. In turn, bound CENP-B further opens the chromatin fiber structure and induces nucleosomal DNA unwrapping. Finally, removal of CENP-A increases CENP-B mobility in cells. Together, our studies show that the two centromere-specific proteins collaborate to reshape chromatin structure, enabling the binding of centromeric factors and establishing a centromeric chromatin state.
Collapse
Affiliation(s)
- Harsh Nagpal
- École Polytechnique Fédérale de Lausanne (EPFL), SB ISIC LCBM, Station 6, CH-1015, Lausanne, Switzerland
| | - Ahmad Ali-Ahmad
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, Faculty of Medicine, University of Oslo, Oslo, 0318, Norway
| | - Yasuhiro Hirano
- Graduate School of Frontier Biosciences, Osaka University, Suita, 565-0871, Japan
| | - Wei Cai
- École Polytechnique Fédérale de Lausanne (EPFL), SB ISIC LCBM, Station 6, CH-1015, Lausanne, Switzerland
| | - Mario Halic
- Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105-3678, USA
| | - Tatsuo Fukagawa
- Graduate School of Frontier Biosciences, Osaka University, Suita, 565-0871, Japan
| | - Nikolina Sekulić
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, Faculty of Medicine, University of Oslo, Oslo, 0318, Norway.
- Department of Chemistry, University of Oslo, P.O. Box 1033, Blindern, 0315, Norway.
| | - Beat Fierz
- École Polytechnique Fédérale de Lausanne (EPFL), SB ISIC LCBM, Station 6, CH-1015, Lausanne, Switzerland.
| |
Collapse
|
43
|
Kyriacou E, Heun P. Centromere structure and function: lessons from Drosophila. Genetics 2023; 225:iyad170. [PMID: 37931172 PMCID: PMC10697814 DOI: 10.1093/genetics/iyad170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/01/2023] [Indexed: 11/08/2023] Open
Abstract
The fruit fly Drosophila melanogaster serves as a powerful model organism for advancing our understanding of biological processes, not just by studying its similarities with other organisms including ourselves but also by investigating its differences to unravel the underlying strategies that evolved to achieve a common goal. This is particularly true for centromeres, specialized genomic regions present on all eukaryotic chromosomes that function as the platform for the assembly of kinetochores. These multiprotein structures play an essential role during cell division by connecting chromosomes to spindle microtubules in mitosis and meiosis to mediate accurate chromosome segregation. Here, we will take a historical perspective on the study of fly centromeres, aiming to highlight not only the important similarities but also the differences identified that contributed to advancing centromere biology. We will discuss the current knowledge on the sequence and chromatin organization of fly centromeres together with advances for identification of centromeric proteins. Then, we will describe both the factors and processes involved in centromere organization and how they work together to provide an epigenetic identity to the centromeric locus. Lastly, we will take an evolutionary point of view of centromeres and briefly discuss current views on centromere drive.
Collapse
Affiliation(s)
- Eftychia Kyriacou
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Patrick Heun
- Wellcome Centre of Cell Biology, School of Biological Sciences, University of Edinburgh, EH9 3BF Edinburgh, UK
- Department of Biology, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| |
Collapse
|
44
|
Ariyoshi M, Fukagawa T. An updated view of the kinetochore architecture. Trends Genet 2023; 39:941-953. [PMID: 37775394 DOI: 10.1016/j.tig.2023.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 10/01/2023]
Abstract
The kinetochore is a supramolecular complex that facilitates faithful chromosome segregation by bridging the centromere and spindle microtubules. Recent functional and structural studies on the inner kinetochore subcomplex, constitutive centromere-associated network (CCAN) have updated our understanding of kinetochore architecture. While the CCAN core establishes a stable interface with centromeric chromatin, CCAN organization is dynamically altered and coupled with cell cycle progression. Furthermore, the CCAN components, centromere protein (CENP)-C and CENP-T, mediate higher-order assembly of multiple kinetochore units on the regional centromeres of vertebrates. This review highlights new insights into kinetochore rigidity, plasticity, and clustering, which are key to understanding temporal and spatial regulatory mechanisms of chromosome segregation.
Collapse
Affiliation(s)
- 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
|
45
|
Liu R, Dou Z, Tian T, Gao X, Chen L, Yuan X, Wang C, Hao J, Gui P, Mullen M, Aikhionbare F, Niu L, Bi G, Zou P, Zhang X, Fu C, Yao X, Zang J, Liu X. Dynamic phosphorylation of CENP-N by CDK1 guides accurate chromosome segregation in mitosis. J Mol Cell Biol 2023; 15:mjad041. [PMID: 37365681 PMCID: PMC10799313 DOI: 10.1093/jmcb/mjad041] [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: 07/26/2022] [Revised: 01/09/2023] [Accepted: 06/24/2023] [Indexed: 06/28/2023] Open
Abstract
In mitosis, accurate chromosome segregation depends on the kinetochore, a supermolecular machinery that couples dynamic spindle microtubules to centromeric chromatin. However, the structure-activity relationship of the constitutive centromere-associated network (CCAN) during mitosis remains uncharacterized. Building on our recent cryo-electron microscopic analyses of human CCAN structure, we investigated how dynamic phosphorylation of human CENP-N regulates accurate chromosome segregation. Our mass spectrometric analyses revealed mitotic phosphorylation of CENP-N by CDK1, which modulates the CENP-L-CENP-N interaction for accurate chromosome segregation and CCAN organization. Perturbation of CENP-N phosphorylation is shown to prevent proper chromosome alignment and activate the spindle assembly checkpoint. These analyses provide mechanistic insight into a previously undefined link between the centromere-kinetochore network and accurate chromosome segregation.
Collapse
Affiliation(s)
- Ran Liu
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- CAS Center for Excellence in Molecular and Cell Sciences, Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Zhen Dou
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- CAS Center for Excellence in Molecular and Cell Sciences, Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Tian Tian
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Xinjiao Gao
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- CAS Center for Excellence in Molecular and Cell Sciences, Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Lili Chen
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Xiao Yuan
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- CAS Center for Excellence in Molecular and Cell Sciences, Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Chunyue Wang
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Jiahe Hao
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Ping Gui
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- CAS Center for Excellence in Molecular and Cell Sciences, Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
- Keck Center for Cellular Dynamics, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - McKay Mullen
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Keck Center for Cellular Dynamics, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Felix Aikhionbare
- Keck Center for Cellular Dynamics, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Liwen Niu
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- CAS Center for Excellence in Molecular and Cell Sciences, Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Guoqiang Bi
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- CAS Center for Excellence in Molecular and Cell Sciences, Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Peng Zou
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Xuan Zhang
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Chuanhai Fu
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- CAS Center for Excellence in Molecular and Cell Sciences, Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- CAS Center for Excellence in Molecular and Cell Sciences, Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| | - Jianye Zang
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- CAS Center for Excellence in Molecular and Cell Sciences, Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei 230027, China
| |
Collapse
|
46
|
Wu LZ, Zou Y, Wang BR, Ni HF, Kong YG, Hua QQ, Chen SM. Enhancing nasopharyngeal carcinoma cell radiosensitivity by suppressing AKT/mTOR via CENP-N knockdown. J Transl Med 2023; 21:792. [PMID: 37940975 PMCID: PMC10631041 DOI: 10.1186/s12967-023-04654-x] [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: 07/24/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023] Open
Abstract
OBJECTIVE Investigating the impact of centromere protein N (CENP-N) on radiosensitivity of nasopharyngeal carcinoma (NPC) cells. METHODS Using immunohistochemistry and immunofluorescence to detect CENP-N expression in tissues from 35 patients with radiosensitive or radioresistant NPC. Assessing the effect of combined CENP-N knockdown and radiotherapy on various cellular processes by CCK-8, colony formation, flow cytometry, and Western blotting. Establishing a NPC xenograft model. When the tumor volume reached 100 mm3, a irradiation dose of 6 Gy was given, and the effects of the combined treatment were evaluated in vivo using immunofluorescence and Western blotting techniques. RESULTS The level of CENP-N was significantly reduced in radiosensitive tissues of NPC (p < 0.05). Knockdown of CENP-N enhanced NPC radiosensitivity, resulting in sensitizing enhancement ratios (SER) of 1.44 (5-8 F) and 1.16 (CNE-2Z). The combined treatment showed significantly higher levels of proliferation suppression, apoptosis, and G2/M phase arrest (p < 0.01) compared to either CENP-N knockdown alone or radiotherapy alone. The combined treatment group showed the highest increase in Bax and γH2AX protein levels, whereas the protein Cyclin D1 exhibited the greatest decrease (p < 0.01). However, the above changes were reversed after treatment with AKT activator SC79. In vivo, the mean volume and weight of tumors in the radiotherapy group were 182 ± 54 mm3 and 0.16 ± 0.03 g. The mean tumor volume and weight in the combined treatment group were 84 ± 42 mm3 and 0.04 ± 0.01 g. CONCLUSION Knockdown of CENP-N can enhance NPC radiosensitivity by inhibiting AKT/mTOR.
Collapse
Affiliation(s)
- Li-Zhi Wu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, 430060, Hubei, People's Republic of China
| | - You Zou
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, 430060, Hubei, People's Republic of China
| | - Bin-Ru Wang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, 430060, Hubei, People's Republic of China
| | - Hai-Feng Ni
- Department of Otolaryngology Head and Neck surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, People's Republic of China
| | - Yong-Gang Kong
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, 430060, Hubei, People's Republic of China
| | - Qing-Quan Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, 430060, Hubei, People's Republic of China.
- Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, 430060, Hubei, People's Republic of China.
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, 430060, Hubei, People's Republic of China.
- Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, 430060, Hubei, People's Republic of China.
| |
Collapse
|
47
|
Kale S, Boopathi R, Belotti E, Lone IN, Graies M, Schröder M, Petrova M, Papin C, Bednar J, Ugrinova I, Hamiche A, Dimitrov S. The CENP-A nucleosome: where and when it happens during the inner kinetochore's assembly. Trends Biochem Sci 2023; 48:849-859. [PMID: 37596196 DOI: 10.1016/j.tibs.2023.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 08/20/2023]
Abstract
CENP-A is an essential histone variant that replaces the canonical H3 at the centromeres and marks these regions epigenetically. The CENP-A nucleosome is the specific building block of centromeric chromatin, and it is recognized by CENP-C and CENP-N, two components of the constitutive centromere-associated network (CCAN), the first protein layer of the kinetochore. Recent proposals of the yeast and human (h)CCAN structures position the assembly on exposed DNA, suggesting an elusive spatiotemporal recognition. We summarize the data on the structural organization of the CENP-A nucleosome and the binding of CENP-C and CENP-N. The latter posits an apparent contradiction in engaging the CENP-A nucleosome versus the CCAN. We propose a reconciliatory model for the assembly of CCAN on centromeric chromatin.
Collapse
Affiliation(s)
- Seyit Kale
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Balcova, Izmir 35330, Turkey.
| | - Ramachandran Boopathi
- Université Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), 38000 Grenoble, France; Laboratoire de Biologie et de Modelisation de la Cellule (LBMC), CNRS/ENSL/UCBL, Ecole Normale Supérieure de Lyon, 69007 Lyon, France
| | - Edwige Belotti
- Laboratoire Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR5261, 8 avenue Rockefeller, 69008 Lyon, France
| | - Imtiaz Nisar Lone
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Balcova, Izmir 35330, Turkey
| | - Mohamed Graies
- Institute for Advanced Biosciences, INSERM U1209, CNRS, UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Maria Schröder
- Roumen Tsanev Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Maria Petrova
- Roumen Tsanev Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Christophe Papin
- Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC), UdS, CNRS, INSERM, Illkirch-Graffenstaden, France
| | - Jan Bednar
- Institute for Advanced Biosciences, INSERM U1209, CNRS, UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Iva Ugrinova
- Roumen Tsanev Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Ali Hamiche
- Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC), UdS, CNRS, INSERM, Illkirch-Graffenstaden, France.
| | - Stefan Dimitrov
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Balcova, Izmir 35330, Turkey; Institute for Advanced Biosciences, INSERM U1209, CNRS, UMR 5309, Université Grenoble Alpes, 38000 Grenoble, France; Roumen Tsanev Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| |
Collapse
|
48
|
Dudka D, Akins RB, Lampson MA. FREEDA: An automated computational pipeline guides experimental testing of protein innovation. J Cell Biol 2023; 222:e202212084. [PMID: 37358475 PMCID: PMC10292211 DOI: 10.1083/jcb.202212084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 04/22/2023] [Accepted: 06/07/2023] [Indexed: 06/27/2023] Open
Abstract
Cell biologists typically focus on conserved regions of a protein, overlooking innovations that can shape its function over evolutionary time. Computational analyses can reveal potential innovations by detecting statistical signatures of positive selection that lead to rapid accumulation of beneficial mutations. However, these approaches are not easily accessible to non-specialists, limiting their use in cell biology. Here, we present an automated computational pipeline FREEDA that provides a simple graphical user interface requiring only a gene name; integrates widely used molecular evolution tools to detect positive selection in rodents, primates, carnivores, birds, and flies; and maps results onto protein structures predicted by AlphaFold. Applying FREEDA to >100 centromere proteins, we find statistical evidence of positive selection within loops and turns of ancient domains, suggesting innovation of essential functions. As a proof-of-principle experiment, we show innovation in centromere binding of mouse CENP-O. Overall, we provide an accessible computational tool to guide cell biology research and apply it to experimentally demonstrate functional innovation.
Collapse
Affiliation(s)
- Damian Dudka
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - R. Brian Akins
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael A. Lampson
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
49
|
Yatskevich S, Barford D, Muir KW. Conserved and divergent mechanisms of inner kinetochore assembly onto centromeric chromatin. Curr Opin Struct Biol 2023; 81:102638. [PMID: 37343495 DOI: 10.1016/j.sbi.2023.102638] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/03/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023]
Abstract
Kinetochores are large protein complexes built on centromeric chromatin that mediate chromosome segregation. The inner kinetochore, or constitutive centromere-associated network (CCAN), assembles onto centromeres defined by centromere protein A (CENP-A) nucleosomes (CENP-ANuc), and acts as a platform for the regulated assembly of the microtubule-binding outer kinetochore. Recent cryo-EM work revealed structural conservation of CCAN, from the repeating human regional centromeres to the point centromere of budding yeast. Centromere recognition is determined mainly through engagement of duplex DNA proximal to the CENP-A nucleosome by a DNA-binding CENP-LN channel located at the core of CCAN. Additional DNA interactions formed by other CCAN modules create an enclosed DNA-binding chamber. This configuration explains how kinetochores maintain their tight grip on centromeric DNA to withstand the forces of chromosome segregation. Defining the higher-order architecture of complete kinetochore assemblies with implications for understanding the 3D organisation of regional centromeres and mechanisms of kinetochore dynamics, including how kinetochores sense and respond to tension, are important future directions.
Collapse
Affiliation(s)
- Stanislau Yatskevich
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom. https://twitter.com/StanislauY
| | - David Barford
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom.
| | - Kyle W Muir
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, United Kingdom. https://twitter.com/centromuir
| |
Collapse
|
50
|
Dendooven T, Zhang Z, Yang J, McLaughlin SH, Schwab J, Scheres SHW, Yatskevich S, Barford D. Cryo-EM structure of the complete inner kinetochore of the budding yeast point centromere. SCIENCE ADVANCES 2023; 9:eadg7480. [PMID: 37506202 PMCID: PMC10381965 DOI: 10.1126/sciadv.adg7480] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023]
Abstract
The point centromere of budding yeast specifies assembly of the large kinetochore complex to mediate chromatid segregation. Kinetochores comprise the centromere-associated inner kinetochore (CCAN) complex and the microtubule-binding outer kinetochore KNL1-MIS12-NDC80 (KMN) network. The budding yeast inner kinetochore also contains the DNA binding centromere-binding factor 1 (CBF1) and CBF3 complexes. We determined the cryo-electron microscopy structure of the yeast inner kinetochore assembled onto the centromere-specific centromere protein A nucleosomes (CENP-ANuc). This revealed a central CENP-ANuc with extensively unwrapped DNA ends. These free DNA duplexes bind two CCAN protomers, one of which entraps DNA topologically, positioned on the centromere DNA element I (CDEI) motif by CBF1. The two CCAN protomers are linked through CBF3 forming an arch-like configuration. With a structural mechanism for how CENP-ANuc can also be linked to KMN involving only CENP-QU, we present a model for inner kinetochore assembly onto a point centromere and how it organizes the outer kinetochore for chromosome attachment to the mitotic spindle.
Collapse
Affiliation(s)
| | | | - Jing Yang
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | | | | | | | | | | |
Collapse
|