1
|
Haykal MM, Rodrigues-Ferreira S, El Botty R, Sourd L, Marangoni E, Varin M, Denis A, Nahmias C. Targeting WEE1 kinase as a therapeutic strategy in ATIP3-deficient breast cancers. Cancer Lett 2025; 620:217665. [PMID: 40127815 DOI: 10.1016/j.canlet.2025.217665] [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: 01/06/2025] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
ATIP3-deficient breast cancers represent a subset of aggressive tumors with limited therapeutic options and poor prognosis. Here, we screened a panel of cell cycle kinase inhibitors to identify novel targets for these tumors. We show that loss of ATIP3 sensitizes breast cancer cells to WEE1 inhibition, resulting in aberrant mitoses characterized by detachment of centromere proteins from DNA and chromosome pulverization. This phenotype arises from excessive replication stress and DNA damage in S-phase, combined with premature mitotic entry driven by untimely CDK1 activation. Mechanistically, we identify DNA2 helicase/nuclease as a key mediator of chromosome pulverization. Importantly, the heightened sensitivity of ATIP3-deficient cells to WEE1 inhibition provides a strong rationale for clinical exploration of WEE1-targeted therapies. Furthermore, combining WEE1 and PKMYT1 inhibitors enhances therapeutic efficacy, offering a promising strategy for personalized treatment in ATIP3-deficient breast cancers.
Collapse
Affiliation(s)
- Maria M Haykal
- Institut Gustave Roussy, Inserm U981, Biomarqueurs prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, 94800, Villejuif, France; Université Paris-Saclay, 91400, Orsay, France
| | - Sylvie Rodrigues-Ferreira
- Institut Gustave Roussy, Inserm U981, Biomarqueurs prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, 94800, Villejuif, France; Université Paris-Saclay, 91400, Orsay, France; Inovarion, 75005, Paris, France
| | - Rania El Botty
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | - Laura Sourd
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | - Elisabetta Marangoni
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | | | | | - Clara Nahmias
- Institut Gustave Roussy, Inserm U981, Biomarqueurs prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, 94800, Villejuif, France; Université Paris-Saclay, 91400, Orsay, France.
| |
Collapse
|
2
|
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
|
3
|
Zerbib J, Bloomberg A, Ben-David U. Targeting vulnerabilities of aneuploid cells for cancer therapy. Trends Cancer 2025:S2405-8033(25)00097-4. [PMID: 40368673 DOI: 10.1016/j.trecan.2025.04.005] [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: 02/24/2025] [Revised: 04/04/2025] [Accepted: 04/04/2025] [Indexed: 05/16/2025]
Abstract
Aneuploidy is a common feature of cancer that drives tumor evolution, but it also creates cellular vulnerabilities that might be exploited therapeutically. Recent advances in genomic technologies and experimental models have uncovered diverse cellular consequences of aneuploidy, revealing dependencies on mitotic regulation, DNA replication and repair, proteostasis, metabolism, and immune interactions. Harnessing aneuploidy for precision oncology requires the combination of genomic, functional, and clinical studies that will enable translation of our improved understanding of aneuploidy to targeted therapies. In this review we discuss approaches to targeting both highly aneuploid cells and cells with specific common aneuploidies, summarize the biological underpinning of these aneuploidy-induced vulnerabilities, and explore their therapeutic implications.
Collapse
Affiliation(s)
- Johanna Zerbib
- Department of Human Molecular Genetics and Biochemistry, Gray Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Amit Bloomberg
- Department of Human Molecular Genetics and Biochemistry, Gray Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Gray Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
4
|
Ya A, Deng C, Godek KM. Cell competition eliminates aneuploid human pluripotent stem cells. Stem Cell Reports 2025:102506. [PMID: 40409259 DOI: 10.1016/j.stemcr.2025.102506] [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: 05/23/2024] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/25/2025] Open
Abstract
Human pluripotent stem cells (hPSCs) maintain diploid populations for generations despite frequent mitotic errors that cause aneuploidy or chromosome imbalances. Consequently, aneuploid hPSC propagation must be prevented to sustain genome stability, but how this is achieved is unknown. Surprisingly, we find that, unlike somatic cells, uniformly aneuploid hPSC populations with heterogeneous abnormal karyotypes proliferate. Instead, in mosaic populations, cell-non-autonomous competition between neighboring diploid and aneuploid hPSCs eliminates less fit aneuploid cells, regardless of specific chromosome imbalances. Aneuploid hPSCs with lower MYC or higher p53 levels relative to diploid neighbors are outcompeted but conversely gain an advantage when MYC and p53 relative abundance switches. Thus, MYC- and p53-driven cell competition preserves hPSC genome integrity despite their low mitotic fidelity and intrinsic capacity to proliferate with an aneuploid genome. These findings have important implications for using hPSCs in regenerative medicine and for how diploid human embryos form during development despite the prevalence of aneuploidy.
Collapse
Affiliation(s)
- Amanda Ya
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Chenhui Deng
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Kristina M Godek
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.
| |
Collapse
|
5
|
Bolanos-Garcia VM. Mps1 kinase functions in mitotic spindle assembly and error correction. Trends Biochem Sci 2025; 50:438-453. [PMID: 40082122 DOI: 10.1016/j.tibs.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 02/12/2025] [Accepted: 02/21/2025] [Indexed: 03/16/2025]
Abstract
The protein kinase Mps1 (also known as TTK) is a central component of the mitotic spindle assembly checkpoint (SAC), an essential self-monitoring system of the eukaryotic cell cycle that ensures accurate chromosome segregation by delaying the onset of anaphase until all chromosomes are properly bioriented on the mitotic spindle. Mps1 kinase is an important upstream regulator of the SAC and its recruitment to kinetochores critical for initiating SAC signaling. This review discusses the current understanding of Mps1 essential functions in the SAC, the emerging details of Mps1 role in error correction to safeguard genome stability, and the therapeutic potential of Mps1 inhibition for the treatment of cancer types associated with aberrant SAC signaling and chromosome segregation defects.
Collapse
Affiliation(s)
- Victor M Bolanos-Garcia
- Faculty of Health and Life Sciences, Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK.
| |
Collapse
|
6
|
Leça N, Barbosa F, Rodriguez-Calado S, Esposito Verza A, Moura M, Pedroso PD, Pinto I, Artes E, Bange T, Sunkel CE, Barisic M, Maresca TJ, Conde C. Proximity-based activation of AURORA A by MPS1 potentiates error correction. Curr Biol 2025; 35:1935-1947.e8. [PMID: 40203828 PMCID: PMC12014372 DOI: 10.1016/j.cub.2025.03.018] [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: 06/11/2024] [Revised: 01/29/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025]
Abstract
Faithful cell division relies on mitotic chromosomes becoming bioriented with each pair of sister kinetochores bound to microtubules oriented toward opposing spindle poles. Erroneous kinetochore-microtubule attachments often form during early mitosis but are destabilized through the phosphorylation of outer kinetochore proteins by centromeric AURORA B kinase (ABK) and centrosomal AURORA A kinase (AAK), thus allowing for re-establishment of attachments until biorientation is achieved.1,2,3,4,5,6,7,8,9 MPS1-mediated phosphorylation of NDC80 has also been shown to directly weaken the kinetochore-microtubule interface in yeast.10 In human cells, MPS1 has been proposed to transiently accumulate at end-on attached kinetochores11 and phosphorylate SKA3 to promote microtubule release.12 Whether MPS1 directly targets NDC80 and/or promotes the activity of AURORA kinases in metazoans remains unclear. Here, we report a novel mechanism involving communication between kinetochores and centrosomes, wherein MPS1 acts upstream of AAK to promote error correction. MPS1 on pole-proximal kinetochores phosphorylates the C-lobe of AAK, thereby increasing its activation at centrosomes. This proximity-based activation ensures the establishment of a robust AAK activity gradient that locally destabilizes mal-oriented kinetochores near spindle poles. Accordingly, MPS1 depletion from Drosophila cells causes severe chromosome misalignment and erroneous kinetochore-microtubule attachments, which can be rescued by tethering either MPS1 or constitutively active AAK mutants to centrosomes. Proximity-based activation of AAK by MPS1 also occurs in human cells to promote AAK-mediated phosphorylation of the NDC80 N-terminal tail. These findings uncover an MPS1-AAK crosstalk that is required for efficient error correction, showcasing the ability of kinetochores to modulate centrosome outputs to ensure proper chromosome segregation.
Collapse
Affiliation(s)
- Nelson Leça
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Francisca Barbosa
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Sergi Rodriguez-Calado
- Cell Division and Cytoskeleton, Danish Cancer Institute, Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - Arianna Esposito Verza
- Department of Mechanistic Cell Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227 Dortmund, Germany
| | - Margarida Moura
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Paulo D Pedroso
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Inês Pinto
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Elena Artes
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestrasse 31, 80336 Munich, Germany
| | - Tanja Bange
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Goethestrasse 31, 80336 Munich, Germany
| | - Claudio E Sunkel
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Departamento de Biologia Molecular, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Marin Barisic
- Cell Division and Cytoskeleton, Danish Cancer Institute, Strandboulevarden 49, 2100 Copenhagen, Denmark; Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Thomas J Maresca
- Biology Department, University of Massachusetts, 611 North Pleasant Street, Amherst, MA 01003-9297, USA; Molecular and Cellular Biology Graduate Program, University of Massachusetts, 611 North Pleasant Street, Amherst, MA 01003-9297, USA.
| | - Carlos Conde
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Departamento de Biologia Molecular, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| |
Collapse
|
7
|
Eykelenboom JK, Gierliński M, Yue Z, Tanaka TU. Nuclear exclusion of condensin I in prophase coordinates mitotic chromosome reorganization to ensure complete sister chromatid resolution. Curr Biol 2025; 35:1562-1575.e7. [PMID: 40107266 DOI: 10.1016/j.cub.2025.02.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 12/18/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
During early mitosis, chromosomes transition from their unfolded interphase state to the distinct rod-shaped structures characteristic of mitosis. This process allows correct segregation of replicated sister chromatids to the opposite spindle poles during anaphase. Two protein complexes, named condensin I and condensin II, facilitate mitotic chromosome organization. Condensin II is important for achieving sister chromatid separation (resolution), while condensin I is required for chromosome condensation (folding). Although sister chromatid resolution occurs earlier than chromosome folding, it is not yet clear how these events are coordinated through time or whether this is important for correct chromosome segregation. In this study, we tested the hypothesis that temporal control is achieved through differential localization of the two condensin complexes; i.e., while condensin II localizes in the nucleus, condensin I is excluded from the nucleus in interphase and prophase. We engineered the localization of condensin I to the nucleus and monitored sister chromatid resolution and chromosome folding by real-time imaging. We found that localization of condensin I to the nucleus led to precocious chromosome folding during prophase, with similar timing to sister chromatid resolution. Furthermore, this change led to incomplete sister chromatid resolution in prometaphase/metaphase and frequent chromosome missegregation in anaphase, in which most missegregated chromosomes consisted of lagging chromosomes involving both sister chromatids. We conclude that, in a physiological context, the exclusion of condensin I from the nucleus during prophase delays chromosome folding and allows condensin II to complete sister chromatid resolution, which ensures correct chromosome segregation later in mitosis.
Collapse
Affiliation(s)
- John K Eykelenboom
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK.
| | - Marek Gierliński
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK; Data Analysis Group, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Zuojun Yue
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Tomoyuki U Tanaka
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK.
| |
Collapse
|
8
|
Cearlock A, Mysliwiec H, Agarsheva M, Krzyspiak J, Ozair MZ, Brivanlou AH, Yang M. Exploring and validating the marmoset as a primate model for chromosomal instability in early development. Mol Hum Reprod 2025; 31:gaaf012. [PMID: 40193493 DOI: 10.1093/molehr/gaaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
Aneuploidy in embryos poses a major barrier to successful human reproduction, contributing to nearly 50% of early miscarriages. Despite its high prevalence in human embryos, the molecular mechanisms regulating aneuploid cell fate during development remain poorly understood. This knowledge gap persists due to ethical constraints in human embryo research and the limitations of existing animal models. In this study, we identified the New World primate marmoset (Callithrix jacchus) as a suitable model for investigating aneuploidy. By calling copy number variants from single-cell RNA-sequencing data of marmoset embryonic cells, we identified heterogeneous aneuploidy, indicating chromosomal instability (CIN) in marmoset preimplantation embryos. Furthermore, marmoset aneuploidy displayed lineage-specific behavior during gastruloid differentiation, similar to humans, suggesting a conserved regulatory mechanism in lineage specification. To develop a more pluripotent cell line to study early specification, we established an efficient approach for generating naïve-like marmoset pluripotent stem cells (cjPSCs). These cells resemble preimplantation epiblast-like cells and exhibit inherent CIN. Transcriptome analysis identified potential pathways contributing to aneuploidy during early embryogenesis, including the downregulation of cell cycle checkpoint signaling and the upregulation of autophagy pathways. Additionally, we found no significant effect of spontaneously occurring aneuploidy in cjPSCs on blastoid formation, suggesting that the consequences of aneuploidy become evident only after gastrulation, with preimplantation lineages exhibiting a higher tolerance for genomic instability. Unexpectedly, aneuploidy enhanced cavity formation during blastoid development, suggesting a potential role in facilitating efficient trophectoderm differentiation. Our findings validate the marmoset as a valuable model for studying CIN during early primate development and provide insight into the mechanisms underlying the prevalence of aneuploidy in primates. Naïve-like cjPSCs recapitulate key phenotypic traits of early embryonic cells, providing a robust system for studying post-implantation aneuploid cell fates in vivo and serving as a foundation for future research in this field.
Collapse
Affiliation(s)
- Andrew Cearlock
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Hubert Mysliwiec
- Laboratory of Synthetic Embryology, The Rockefeller University, New York, NY, USA
| | - Margarita Agarsheva
- Laboratory of Synthetic Embryology, The Rockefeller University, New York, NY, USA
| | - Joanna Krzyspiak
- Laboratory of Synthetic Embryology, The Rockefeller University, New York, NY, USA
| | - Mohammad Zeeshan Ozair
- Laboratory of Synthetic Embryology, The Rockefeller University, New York, NY, USA
- Department of Radiation Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York, NY, USA
| | - Ali H Brivanlou
- Laboratory of Synthetic Embryology, The Rockefeller University, New York, NY, USA
| | - Min Yang
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Washington National Primate Center, Seattle, WA, USA
| |
Collapse
|
9
|
Smarduch S, Moreno-Velasquez SD, Ilic D, Dadsena S, Morant R, Ciprinidis A, Pereira G, Binder M, García-Sáez AJ, Acebrón SP. A novel biosensor for the spatiotemporal analysis of STING activation during innate immune responses to dsDNA. EMBO J 2025; 44:2157-2182. [PMID: 39984755 PMCID: PMC11962129 DOI: 10.1038/s44318-025-00370-y] [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: 05/27/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 02/23/2025] Open
Abstract
The cGAS-STING signalling pathway has a central role in the innate immune response to extrinsic and intrinsic sources of cytoplasmic dsDNA. At the core of this pathway is cGAS-dependent production of the intra- and extra-cellular messenger cGAMP, which activates STING and leads to IRF3-dependent expression of cytokines and interferons. Despite its relevance to viral and bacterial infections, cell death, and genome instability, the lack of specific live-cell reporters has precluded spatiotemporal analyses of cGAS-STING signalling. Here, we generate a fluorescent biosensor termed SIRF (STING-IRF3), which reports on the functional interaction between activated STING and IRF3 at the Golgi. We show that cells harbouring SIRF react in a time- and concentration-dependent manner both to STING agonists and to microenvironmental cGAMP. We demonstrate that the new biosensor is suitable for single-cell characterisation of immune responses to HSV-1 infection, mtDNA release upon apoptosis, or other sources of cytoplasmic dsDNA. Furthermore, our results indicate that STING signalling is not activated by ruptured micronuclei, suggesting that other cytosolic pattern recognition receptors underlie the interferon responses to chromosomal instability.
Collapse
Affiliation(s)
- Steve Smarduch
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | | | - Doroteja Ilic
- Division of Virus-associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Shashank Dadsena
- Institute of Genetics, CECAD, University of Cologne, Cologne, Germany
| | - Ryan Morant
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Anja Ciprinidis
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Gislene Pereira
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
- Molecular Biology of Centrosome and Cilia, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Marco Binder
- Division of Virus-associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ana J García-Sáez
- Institute of Genetics, CECAD, University of Cologne, Cologne, Germany
- Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Sergio P Acebrón
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany.
- IKERBASQUE, Basque Foundation of Science, Bilbao, Spain.
- University of the Basque Country (UPV/EHU), Leioa, Spain.
| |
Collapse
|
10
|
DiPeso L, Pendyala S, Huang HZ, Fowler DM, Hatch EM. Image-based identification and isolation of micronucleated cells to dissect cellular consequences. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.05.04.539483. [PMID: 37205341 PMCID: PMC10187275 DOI: 10.1101/2023.05.04.539483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Recent advances in isolating cells based on visual phenotypes have transformed our ability to identify the mechanisms and consequences of complex traits. Micronucleus (MN) formation is a frequent outcome of genome instability, triggers extensive changes in genome structure and signaling coincident with MN rupture, and is almost exclusively defined by visual analysis. Automated MN detection in microscopy images has proved challenging, limiting discovery of the mechanisms and consequences of MN. In this study we describe two new MN segmentation modules: a rapid model for classifying micronucleated cells and their rupture status (VCS MN), and a robust model for accurate MN segmentation (MNFinder) from a broad range of cell lines. As proof-of-concept, we define the transcriptome of non-transformed human cells with intact or ruptured MN after chromosome missegregation by combining VCS MN with photoactivation-based cell isolation and RNASeq. Surprisingly, we find that neither MN formation nor rupture triggers a strong unique transcriptional response. Instead, transcriptional changes appear correlated with small increases in aneuploidy in these cell classes. Our MN segmentation modules overcome a significant challenge with reproducible MN quantification, and, joined with visual cell sorting, enable the application of powerful functional genomics assays to a wide-range of questions in MN biology.
Collapse
Affiliation(s)
- Lucian DiPeso
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular & Cellular Biology, University of Washington, Seattle, WA, USA
| | | | - Heather Z Huang
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Emily M Hatch
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
11
|
Zheng S, Raz L, Zhou L, Cohen-Sharir Y, Tian R, Ippolito MR, Gianotti S, Saad R, Wardenaar R, Broekhuis M, Suarez Peredo Rodriguez M, Wobben S, van den Brink A, Bakker P, Santaguida S, Foijer F, Ben-David U. High CDC20 levels increase sensitivity of cancer cells to MPS1 inhibitors. EMBO Rep 2025; 26:1036-1061. [PMID: 39838194 PMCID: PMC11850905 DOI: 10.1038/s44319-024-00363-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/13/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Spindle assembly checkpoint (SAC) inhibitors are a recently developed class of drugs, which perturb chromosome segregation during cell division, induce chromosomal instability (CIN), and eventually lead to cell death. The molecular features that determine cellular sensitivity to these drugs are not fully understood. We recently reported that aneuploid cancer cells are preferentially sensitive to SAC inhibition. Here we report that sensitivity to SAC inhibition by MPS1 inhibitors is largely driven by the expression of CDC20, a main mitotic activator of the anaphase-promoting complex (APC/C), and that the effect of CDC20 is larger than that of the APC/C itself. Mechanistically, we discovered that CDC20 depletion prolongs metaphase duration, diminishes mitotic errors, and reduces sensitivity to SAC inhibition. We found that aneuploid cells express higher basal levels of CDC20, which shortens the duration of metaphase and leads to multiple mitotic errors, resulting in increased long-term sensitivity to the additional CIN induced by SAC inhibition. Our findings propose high CDC20 expression as a molecular feature associated with the sensitivity to SAC inhibition therapy and as a potential aneuploidy-induced cellular vulnerability.
Collapse
Affiliation(s)
- Siqi Zheng
- European Research Institute for the Biology of Ageing, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands
| | - Linoy Raz
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lin Zhou
- European Research Institute for the Biology of Ageing, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands
| | - Yael Cohen-Sharir
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ruifang Tian
- European Research Institute for the Biology of Ageing, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands
| | | | - Sara Gianotti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, 20141, Italy
| | - Ron Saad
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rene Wardenaar
- European Research Institute for the Biology of Ageing, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands
- Functional Genomics Center, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands
| | - Mathilde Broekhuis
- Functional Genomics Center, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands
| | - Maria Suarez Peredo Rodriguez
- European Research Institute for the Biology of Ageing, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands
| | - Soraya Wobben
- European Research Institute for the Biology of Ageing, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands
| | - Anouk van den Brink
- European Research Institute for the Biology of Ageing, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands
| | - Petra Bakker
- European Research Institute for the Biology of Ageing, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands
| | - Stefano Santaguida
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, 20141, Italy
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands.
- Functional Genomics Center, University of Groningen, 1, Antonius Deusinglaan, 9713 AV, Groningen, The Netherlands.
| | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
12
|
Kong N, Chen K, Chanboonyasitt P, Jiang H, Wong K, Ma H, Chan Y. The interplay of the translocase activity and protein recruitment function of PICH in ultrafine anaphase bridge resolution and genomic stability. Nucleic Acids Res 2025; 53:gkae1249. [PMID: 39704103 PMCID: PMC11797016 DOI: 10.1093/nar/gkae1249] [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: 07/04/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024] Open
Abstract
Incomplete sister centromere decatenation results in centromeric ultrafine anaphase bridges (UFBs). PICH (PLK1-interacting checkpoint helicase), a DNA translocase, plays a crucial role in UFB resolution by recruiting UFB-binding proteins and stimulating topoisomerase IIα. However, the involvement of distinct PICH functions in UFB resolution remains ambiguous. Here, we demonstrate that PICH depletion in non-transformed diploid cells induces DNA damage, micronuclei formation, p53 activation, G1-phase delay and cell death. Whole-genome sequencing reveals that segregation defects induced by PICH depletion cause chromosomal rearrangements, including translocations and inversions, emphasizing its significance in preserving genomic integrity. Furthermore, a PICH mutant that impairs UFB recruitment of BLM and RIF1 partially inhibits UFB resolution while a translocase-inactive mutant (PICHK128A) fails to resolve UFBs. Notably, expression of PICHK128A inhibits single-stranded UFB formation and induces hypocondensed chromosomes. We propose that PICH's translocase activity plays a dual role in promoting UFB resolution by facilitating the generation of single-stranded UFBs and stimulating topoisomerase IIα.
Collapse
Affiliation(s)
- Nannan Kong
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Kun Chen
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Primrose Chanboonyasitt
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Huadong Jiang
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Ka Yan Wong
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hoi Tang Ma
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Liver Research, The University of Hong Kong, 5 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Ying Wai Chan
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| |
Collapse
|
13
|
Heinzle C, Höfler A, Yu J, Heid P, Kremer N, Schunk R, Stengel F, Bange T, Boland A, Mayer TU. Positively charged specificity site in cyclin B1 is essential for mitotic fidelity. Nat Commun 2025; 16:853. [PMID: 39833154 PMCID: PMC11747444 DOI: 10.1038/s41467-024-55669-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: 03/23/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
Phosphorylation of substrates by cyclin-dependent kinases (CDKs) is the driving force of cell cycle progression. Several CDK-activating cyclins are involved, yet how they contribute to substrate specificity is still poorly understood. Here, we discover that a positively charged pocket in cyclin B1, which is exclusively conserved within B-type cyclins and binds phosphorylated serine- or threonine-residues, is essential for correct execution of mitosis. HeLa cells expressing pocket mutant cyclin B1 are strongly delayed in anaphase onset due to multiple defects in mitotic spindle function and timely activation of the E3 ligase APC/C. Pocket integrity is essential for APC/C phosphorylation particularly at non-consensus CDK1 sites and full in vitro ubiquitylation activity. Our results support a model in which cyclin B1's pocket facilitates sequential substrate phosphorylations involving initial priming events that assist subsequent pocket-dependent phosphorylations even at non-consensus CDK1 motifs.
Collapse
Affiliation(s)
- Christian Heinzle
- Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Anna Höfler
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Jun Yu
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Peter Heid
- Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Nora Kremer
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, LMU, Munich, Germany
| | - Rebecca Schunk
- Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Florian Stengel
- Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Tanja Bange
- Institute of Medical Psychology and Biomedical Center (BMC), Faculty of Medicine, LMU, Munich, Germany
| | - Andreas Boland
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland.
| | - Thomas U Mayer
- Department of Biology, University of Konstanz, Konstanz, Germany.
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
14
|
Martis ASA, Soundararajan L, Shetty P, Moin S, Vanje T, Jai Sankar Y, Parveen S. Chromosome number alterations cause apoptosis and cellular hypertrophy in induced pluripotent stem cell models of embryonic epiblast cells. Biol Open 2025; 14:BIO061814. [PMID: 39851179 PMCID: PMC11789280 DOI: 10.1242/bio.061814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/26/2025] Open
Abstract
Chromosomal aneuploidies are a major cause of developmental failure and pregnancy loss. To investigate the possible consequences of aneuploidy on early embryonic development in vitro, we focused on primed pluripotent stem cells that are relatable to the epiblast of post-implantation embryos in vivo. We used human induced pluripotent stem cells (iPSCs) as an epiblast model and altered chromosome numbers by treating with reversine, a small-molecule inhibitor of monopolar spindle 1 kinase (MSP1) that inactivates the spindle assembly checkpoint, which has been strongly implicated in chromosome mis-segregation and aneuploidy generation. Upon reversine treatment, we obtained cells with varied chromosomal content that retained pluripotency and potential to differentiate into cells of three germ lineages. However, these cells displayed lagging chromosomes, increased micronuclei content, high p53 expression and excessive apoptotic activity. Cell proliferation was not affected. Prolonged in vitro culture of these cells resulted in a selective pool of cells with supernumerary chromosomes, which exhibited cellular hypertrophy, enlarged nuclei, and overproduction of total RNAs and proteins. We conclude that increased DNA damage responses, apoptosis, and improper cellular mass and functions are possible mechanisms that contribute to abnormal epiblast development.
Collapse
Affiliation(s)
- Althea Stella Anil Martis
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Loshini Soundararajan
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Pallavi Shetty
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Syed Moin
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Tejashree Vanje
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Yogeshwaran Jai Sankar
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| | - Shagufta Parveen
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
15
|
Zhang Y, Su M, Chen Y, Cui L, Xia W, Xu R, Xue D, Zhang X, Feng X. EHMT2-mediated R-loop formation promotes the malignant progression of prostate cancer via activating Aurora B. Clin Transl Med 2025; 15:e70164. [PMID: 39763034 PMCID: PMC11705492 DOI: 10.1002/ctm2.70164] [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: 08/11/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Chromosomal instability (CIN), a hallmark of cancer, is commonly linked to poor prognosis in high-grade prostate cancer (PCa). Paradoxically, excessively high levels of CIN may impair cancer cell viability. Consequently, understanding how tumours adapt to CIN is critical for identifying novel therapeutic targets. METHODS Bioinformatic analyses were conducted to identify genes overexpressed in PCa tissues using The Cancer Genome Atlas (TCGA) and GEO datasets. Western blotting and immunohistochemistry assays were applied to determine the expression levels of euchromatic histone lysine methyltransferase 2 (EHMT2), pT232-Aurora B and Cullin 3 (CUL3). The proliferation of cells was measured through CCK-8 tests, clonogenesis and subcutaneous xenografts of human PCa cells in BALB/c nude mice. Live cell imaging, immunofluorescence (IF) and flow cytometry were used to confirm the role of EHMT2 in PCa cell mitosis. Co-immunoprecipitation, Western blotting and IF assays further elucidated the underlying molecular mechanisms. RESULTS EHMT2 was highly expressed in metastatic PCa tissues exhibiting elevated CIN and was strongly associated with adverse clinical outcomes in patients with PCa. Silencing EHMT2 impaired cell division, inducing G2/M-phase arrest and mitotic catastrophe in PCa cells. Mechanistically, EHMT2 is indispensable to ensure the full activation of Aurora B through centromeric R-loop-driven ATR-CHK1 pathway, with EHMT2 protein expression peaking during the G2/M-phase. Moreover, CUL3 was identified as a binding partner of EHMT2, mediating its polyubiquitination and destabilising its protein levels. CONCLUSIONS This study reveals a CUL3-EHMT2-Aurora B regulatory axis that safeguards accurate chromosome segregation in PCa cells, supporting the potential therapeutic application of EHMT2 inhibitors. KEY POINTS Euchromatic histone lysine methyltransferase 2 (EHMT2) is overexpressed in advanced prostate cancer, restraining catastrophic chromosomal instability (CIN) and enhancing cell fitness. EHMT2 functions via the centromeric R-loop-driven ATR-CHK1-Aurora B pathway to promote chromosomal stability. EHMT2 confers enzalutamide resistance via activating Aurora B. Cullin 3 (CUL3) promotes EHMT2 destabilisation via deubiquitination.
Collapse
Affiliation(s)
- Yuyang Zhang
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Institute of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityHefeiAnhuiChina
| | - Mingqin Su
- Department of PathologyThe Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical UniversityHefeiAnhuiChina
| | - Yiming Chen
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| | - Li Cui
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| | - Wei Xia
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| | - Renfang Xu
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| | - Dong Xue
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| | - Xiansheng Zhang
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Institute of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityHefeiAnhuiChina
| | - Xingliang Feng
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| |
Collapse
|
16
|
Szmyd R, Casolin S, French L, Manjón AG, Walter M, Cavalli L, Nelson CB, Page SG, Dhawan A, Hau E, Pickett HA, Gee HE, Cesare AJ. Homologous recombination promotes non-immunogenic mitotic cell death upon DNA damage. Nat Cell Biol 2025; 27:59-72. [PMID: 39805921 PMCID: PMC11735404 DOI: 10.1038/s41556-024-01557-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: 09/06/2023] [Accepted: 10/14/2024] [Indexed: 01/16/2025]
Abstract
Double-strand breaks (DSBs) can initiate mitotic catastrophe, a complex oncosuppressive phenomenon characterized by cell death during or after cell division. Here we unveil how cell cycle-regulated DSB repair guides disparate cell death outcomes through single-cell analysis of extended live imaging. Following DSB induction in S or G2, passage of unresolved homologous recombination intermediates into mitosis promotes non-immunogenic intrinsic apoptosis in the immediate attempt at cell division. Conversely, non-homologous end joining, microhomology-mediated end joining and single-strand annealing cooperate to enable damaged G1 cells to complete the first cell cycle with an aberrant cell division at the cost of delayed extrinsic lethality and interferon production. Targeting non-homologous end joining, microhomology-mediated end joining or single-strand annealing promotes mitotic death, while suppressing mitotic death enhances interferon production. Together the data indicate that a temporal repair hierarchy, coupled with cumulative DSB load, serves as a reliable predictor of mitotic catastrophe outcomes following genome damage. In this pathway, homologous recombination suppresses interferon production by promoting mitotic lethality.
Collapse
Affiliation(s)
- Radoslaw Szmyd
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Sienna Casolin
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Lucy French
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Anna G Manjón
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Melanie Walter
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Léa Cavalli
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Christopher B Nelson
- Telomere Length Regulation Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Scott G Page
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Andrew Dhawan
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eric Hau
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
- Westmead Clinical School, University of Sydney, Westmead, New South Wales, Australia
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Hilda A Pickett
- Telomere Length Regulation Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Harriet E Gee
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia.
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia.
- Westmead Clinical School, University of Sydney, Westmead, New South Wales, Australia.
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia.
| | - Anthony J Cesare
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia.
| |
Collapse
|
17
|
Black EM, Ramírez Parrado CA, Trier I, Li W, Joo YK, Pichurin J, Liu Y, Kabeche L. Chk2 sustains PLK1 activity in mitosis to ensure proper chromosome segregation. Nat Commun 2024; 15:10782. [PMID: 39737931 PMCID: PMC11685634 DOI: 10.1038/s41467-024-54922-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Polo-like kinase 1 (PLK1) protects against genome instability by ensuring timely and accurate mitotic cell division, and its activity is tightly regulated throughout the cell cycle. Although the pathways that initially activate PLK1 in G2 are well-characterized, the factors that directly regulate mitotic PLK1 remain poorly understood. Here, we identify that human PLK1 activity is sustained by the DNA damage response kinase Checkpoint kinase 2 (Chk2) in mitosis. Chk2 directly phosphorylates PLK1 T210, a residue on its T-loop whose phosphorylation is essential for full PLK1 kinase activity. Loss of Chk2-dependent PLK1 activity causes increased mitotic errors, including chromosome misalignment, chromosome missegregation, and cytokinetic defects. Moreover, Chk2 deficiency increases sensitivity to PLK1 inhibitors, suggesting that Chk2 status may be an informative biomarker for PLK1 inhibitor efficacy. This work demonstrates that Chk2 sustains mitotic PLK1 activity and protects genome stability through discrete functions in interphase DNA damage repair and mitotic chromosome segregation.
Collapse
Affiliation(s)
- Elizabeth M Black
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Carlos Andrés Ramírez Parrado
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Isabelle Trier
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Wenxue Li
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
- Department of Pharmacology, Yale University, New Haven, CT, 06511, USA
| | - Yoon Ki Joo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Jennifer Pichurin
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Yansheng Liu
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
- Department of Pharmacology, Yale University, New Haven, CT, 06511, USA
| | - Lilian Kabeche
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA.
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA.
| |
Collapse
|
18
|
Regin M, Lei Y, Couvreu De Deckersberg E, Janssens C, Huyghebaert A, Guns Y, Verdyck P, Verheyen G, Van de Velde H, Sermon K, Spits C. Complex aneuploidy triggers autophagy and p53-mediated apoptosis and impairs the second lineage segregation in human preimplantation embryos. eLife 2024; 12:RP88916. [PMID: 39652462 PMCID: PMC11627504 DOI: 10.7554/elife.88916] [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] [Indexed: 12/12/2024] Open
Abstract
About 70% of human cleavage stage embryos show chromosomal mosaicism, falling to 20% in blastocysts. Chromosomally mosaic human blastocysts can implant and lead to healthy new-borns with normal karyotypes. Studies in mouse embryos and human gastruloids showed that aneuploid cells are eliminated from the epiblast by p53-mediated apoptosis while being tolerated in the trophectoderm. These observations suggest a selective loss of aneuploid cells from human embryos, but the underlying mechanisms are not yet fully understood. Here, we investigated the cellular consequences of aneuploidy in a total of 125 human blastocysts. RNA-sequencing of trophectoderm cells showed activated p53 pathway and apoptosis proportionate to the level of chromosomal imbalance. Immunostaining corroborated that aneuploidy triggers proteotoxic stress, autophagy, p53-signaling, and apoptosis independent from DNA damage. Total cell numbers were lower in aneuploid embryos, due to a decline both in trophectoderm and in epiblast/primitive endoderm cell numbers. While lower cell numbers in trophectoderm may be attributed to apoptosis, aneuploidy impaired the second lineage segregation, particularly primitive endoderm formation. This might be reinforced by retention of NANOG. Our findings might explain why fully aneuploid embryos fail to further develop and we hypothesize that the same mechanisms lead to the removal of aneuploid cells from mosaic embryos.
Collapse
Affiliation(s)
- Marius Regin
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Yingnan Lei
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Edouard Couvreu De Deckersberg
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Charlotte Janssens
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Anfien Huyghebaert
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Yves Guns
- Brussels Health Campus, Brussels IVF, Universitair Ziekenhuis Brussel (UZ Brussel)BrusselsBelgium
| | - Pieter Verdyck
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
- Brussels Health Campus, Medical Genetics, Universitair Ziekenhuis Brussel (UZ Brussel)BrusselsBelgium
| | - Greta Verheyen
- Brussels Health Campus, Brussels IVF, Universitair Ziekenhuis Brussel (UZ Brussel)BrusselsBelgium
| | - Hilde Van de Velde
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
- Brussels Health Campus, Brussels IVF, Universitair Ziekenhuis Brussel (UZ Brussel)BrusselsBelgium
| | - Karen Sermon
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| | - Claudia Spits
- Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics Reproduction and Development, Vrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
19
|
Kapanidou M, Curtis NL, Diaz-Minguez SS, Agudo-Alvarez S, Rus Sanchez A, Mayah A, Agena R, Brennan P, Morales P, Benito-Arenas R, Bastida A, Bolanos-Garcia VM. Targeting APC/C Ubiquitin E3-Ligase Activation with Pyrimidinethylcarbamate Apcin Analogues for the Treatment of Breast Cancer. Biomolecules 2024; 14:1439. [PMID: 39595615 PMCID: PMC11591962 DOI: 10.3390/biom14111439] [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: 09/25/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Activation of the ubiquitin ligase APC/C by the protein Cdc20 is an essential requirement for proper cell division in higher organisms, including humans. APC/C is the ultimate effector of the Spindle Assembly Checkpoint (SAC), the signalling system that monitors the proper attachment of chromosomes to microtubules during cell division. Defects in this process result in genome instability and cancer. Interfering with APC/C substrate ubiquitylation in cancer cells delays mitotic exit, which induces cell death. Therefore, impairing APC/C function represents an opportunity for the treatment of cancer and malignancies associated with SAC dysregulation. In this study, we report a new class of pyrimidinethylcarbamate apcin analogues that interfere with APC/C activity in 2D and 3D breast cancer cells. The new pyrimidinethylcarbamate apcin analogues exhibited higher cytotoxicity than apcin in all breast cancer cell subtypes investigated, with much lower cytotoxicity observed in fibroblasts and RPE-1 cells. Further molecular rationalisation of apcin and its derivatives was conducted using molecular docking studies. These structural modifications selected from the in silico studies provide a rational basis for the development of more potent chemotypes to treat highly aggressive breast cancer and possibly other aggressive tumour types of diverse tissue origins.
Collapse
Affiliation(s)
- Maria Kapanidou
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; (M.K.); (R.A.)
| | - Natalie L. Curtis
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; (M.K.); (R.A.)
| | - Sandra S. Diaz-Minguez
- Instituto de Química Orgánica, Consejo Superior de Investigaciones Científicas (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain (R.B.-A.)
| | - Sandra Agudo-Alvarez
- Instituto de Química Orgánica, Consejo Superior de Investigaciones Científicas (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain (R.B.-A.)
| | - Alfredo Rus Sanchez
- Instituto de Química Orgánica, Consejo Superior de Investigaciones Científicas (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain (R.B.-A.)
| | - Ammar Mayah
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; (M.K.); (R.A.)
| | - Rosette Agena
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; (M.K.); (R.A.)
- Department of Bioingeniería, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Paul Brennan
- Nuffield Department of Medicine (NDM), Old Road Campus, University of Oxford, Oxford OX3 7BN, UK;
| | - Paula Morales
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain;
| | - Raul Benito-Arenas
- Instituto de Química Orgánica, Consejo Superior de Investigaciones Científicas (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain (R.B.-A.)
| | - Agatha Bastida
- Instituto de Química Orgánica, Consejo Superior de Investigaciones Científicas (CSIC), Juan de la Cierva 3, 28006 Madrid, Spain (R.B.-A.)
| | - Victor M. Bolanos-Garcia
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; (M.K.); (R.A.)
| |
Collapse
|
20
|
Sflakidou E, Adhikari B, Siokatas C, Wolf E, Sarli V. Development of 2-Aminoadenine-Based Proteolysis-Targeting Chimeras (PROTACs) as Novel Potent Degraders of Monopolar Spindle 1 and Aurora Kinases. ACS Pharmacol Transl Sci 2024; 7:3488-3501. [PMID: 39539259 PMCID: PMC11555526 DOI: 10.1021/acsptsci.4c00405] [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: 07/10/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Monopolar spindle 1 (Mps1, also known as TTK) and Aurora kinase (AURK) A and B are critical regulators of mitosis and have been linked to the progression of various cancers. Here, we report the design, synthesis, and biological evaluation of a series of PROTACs (proteolysis-targeting chimeras) targeting TTK and AURKs. We synthesized various degrader molecules based on four different 2-aminoadenine-based ligands, recruiting either cereblon or VHL as the E3-ligase. Our research showed that the nature of the linker and modification of the ligand significantly influence the target specificity and degradation efficacy. Notably, compound 19, among the most potent degraders, demonstrated robust proteasome-mediated degradation of TTK with D max of 66.5% and DC50 value (6 h) of 17.7 nM as compared to its structurally akin inhibitor control, 23. The cytotoxicity of most of the synthesized chimeras against acute myeloid leukemia cell line MV4-11 was lower than that of the corresponding parent inhibitors. However, we could also identify degraders such as 15 and 26 that induce potent AURKA degradation and display comparable antiproliferative activities to their parent compound SF1. Compound 15 degrades AURKA with low DC50 value of 2.05 nM, which is 77-fold and 21-fold more selective toward AURKB and TTK and has an EC50 value of 39 nM against cancer MV4-11 cells. Overall, the observations we made with the degrader molecules we developed can further aid in the design and development of optimized TTK or AURK degraders for cancer therapy.
Collapse
Affiliation(s)
- Eleni Sflakidou
- Department
of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Bikash Adhikari
- Cancer
Systems Biology Group, Chair of Biochemistry and Molecular Biology,
Theodor Boveri Institute, University of
Würzburg, 97074 Würzburg, Germany
- Institute
of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | - Christos Siokatas
- Department
of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Elmar Wolf
- Cancer
Systems Biology Group, Chair of Biochemistry and Molecular Biology,
Theodor Boveri Institute, University of
Würzburg, 97074 Würzburg, Germany
- Institute
of Biochemistry, University of Kiel, 24118 Kiel, Germany
| | - Vasiliki Sarli
- Department
of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| |
Collapse
|
21
|
Yaguchi K, Saito D, Menon T, Matsura A, Hosono M, Mizutani T, Kotani T, Nair S, Uehara R. Haploidy-linked cell proliferation defects limit larval growth in zebrafish. Open Biol 2024; 14:240126. [PMID: 39378986 PMCID: PMC11461072 DOI: 10.1098/rsob.240126] [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: 05/15/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 10/10/2024] Open
Abstract
Haploid larvae in non-mammalian vertebrates are lethal, with characteristic organ growth retardation collectively called 'haploid syndrome'. In contrast to mammals, whose haploid intolerance is attributed to imprinting misregulation, the cellular principle of haploidy-linked defects in non-mammalian vertebrates remains unknown. Here, we investigated cellular defects that disrupt the ontogeny of gynogenetic haploid zebrafish larvae. Unlike diploid control larvae, haploid larvae manifested unscheduled cell death at the organogenesis stage, attributed to haploidy-linked p53 upregulation. Moreover, we found that haploid larvae specifically suffered the gradual aggravation of mitotic spindle monopolarization during 1-3 days post-fertilization, causing spindle assembly checkpoint-mediated mitotic arrest throughout the entire body. High-resolution imaging revealed that this mitotic defect accompanied the haploidy-linked centrosome loss occurring concomitantly with the gradual decrease in larval cell size. Either resolution of mitotic arrest or depletion of p53 partially improved organ growth in haploid larvae. Based on these results, we propose that haploidy-linked mitotic defects and cell death are parts of critical cellular causes shared among vertebrates that limit the larval growth in the haploid state, contributing to an evolutionary constraint on allowable ploidy status in the vertebrate life cycle.
Collapse
Affiliation(s)
- Kan Yaguchi
- Graduate School of Life Science, Hokkaido University, Kita 21, Nishi 11, Kita-Ku, Sapporo001-0021, Japan
- Faculty of Advanced Life Science, Hokkaido University, Kita 21, Nishi 11, Kita-Ku, Sapporo001-0021, Japan
| | - Daiki Saito
- Graduate School of Life Science, Hokkaido University, Kita 21, Nishi 11, Kita-Ku, Sapporo001-0021, Japan
| | - Triveni Menon
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Akira Matsura
- Graduate School of Life Science, Hokkaido University, Kita 21, Nishi 11, Kita-Ku, Sapporo001-0021, Japan
| | - Miyu Hosono
- Graduate School of Life Science, Hokkaido University, Kita 21, Nishi 11, Kita-Ku, Sapporo001-0021, Japan
| | - Takeomi Mizutani
- Department of Life Science and Technology, Faculty of Engineering, Hokkai-Gakuen University, Minami 26, Nishi 11, Chuo-ku, Sapporo064-0926, Japan
| | - Tomoya Kotani
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Kita 10, Nishi 8, Kita-Ku, Sapporo060-0810, Japan
| | - Sreelaja Nair
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai400076, India
| | - Ryota Uehara
- Graduate School of Life Science, Hokkaido University, Kita 21, Nishi 11, Kita-Ku, Sapporo001-0021, Japan
- Faculty of Advanced Life Science, Hokkaido University, Kita 21, Nishi 11, Kita-Ku, Sapporo001-0021, Japan
| |
Collapse
|
22
|
Campos G, Nel-Themaat L. Blastocoel fluid as an alternative source of DNA for minimally invasive PGT and biomarker of embryo competence. Reprod Biomed Online 2024; 49:104322. [PMID: 39121560 DOI: 10.1016/j.rbmo.2024.104322] [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: 02/16/2024] [Revised: 04/26/2024] [Accepted: 06/05/2024] [Indexed: 08/12/2024]
Abstract
The discovery of DNA in blastocoel fluid (BF-DNA) generated new perspectives in the potential development of simpler and safer alternative non-invasive tests in reproductive genetics. Short DNA fragments of apoptotic origin, together with specific expression patterns of pro- and anti-apoptotic genes in the blastocoel fluid of euploid and aneuploid embryos, suggest a self-correction mechanism to preferentially eliminate aneuploid cells, and purge defective and non-viable cells. The correlation of blastocoel fluid content with the genetic status of the whole embryo, and therefore its potential use in minimally invasive preimplantation genetic testing (miPGT), or as an indicator of embryo potential, remains uncertain and needs to be determined. The limited amount and compromised integrity of BF-DNA, with likely apoptotic origination, constrains its amplification, leading to low concordance and reproducibility rates for both aneuploidy screening and monogenic testing. While embryo genotyping constitutes a more ambitious goal, the presence of analysable DNA after amplification in blastocoel fluid may be used as a clinical biomarker of embryo competency to select the most viable embryo(s) for transfer, and potentially improve the implantation rate. Although blastocentesis remains a promising area for future research, several technical and methodological limitations are currently constraining its consideration for clinical practice.
Collapse
Affiliation(s)
- Gerard Campos
- Geisinger Medical Centre, Women's Health Fertility Clinic, Danville, Pennsylvania, USA; Girexx Fertility Clinics, Barcelona, Spain.
| | - Liesl Nel-Themaat
- Stanford Fertility and Reproductive Health Services, Stanford Medicine Children's Health, Sunnyvale, California, USA
| |
Collapse
|
23
|
Cosper PF, Paracha M, Jones KM, Hrycyniak L, Henderson L, Bryan A, Eyzaguirre D, McCunn E, Boulanger E, Wan J, Nickel KP, Horner V, Hu R, Harari PM, Kimple RJ, Weaver BA. Chromosomal instability increases radiation sensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612942. [PMID: 39345631 PMCID: PMC11429890 DOI: 10.1101/2024.09.13.612942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Continuous chromosome missegregation over successive mitotic divisions, known as chromosomal instability (CIN), is common in cancer. Increasing CIN above a maximally tolerated threshold leads to cell death due to loss of essential chromosomes. Here, we show in two tissue contexts that otherwise isogenic cancer cells with higher levels of CIN are more sensitive to ionizing radiation, which itself induces CIN. CIN also sensitizes HPV-positive and HPV-negative head and neck cancer patient derived xenograft (PDX) tumors to radiation. Moreover, laryngeal cancers with higher CIN prior to treatment show improved response to radiation therapy. In addition, we reveal a novel mechanism of radiosensitization by docetaxel, a microtubule stabilizing drug commonly used in combination with radiation. Docetaxel causes cell death by inducing CIN due to abnormal multipolar spindles rather than causing mitotic arrest, as previously assumed. Docetaxel-induced CIN, rather than mitotic arrest, is responsible for the enhanced radiation sensitivity observed in vitro and in vivo, challenging the mechanistic dogma of the last 40 years. These results implicate CIN as a potential biomarker and inducer of radiation response, which could provide valuable cancer therapeutic opportunities. Statement of Significance Cancer cells and laryngeal tumors with higher chromosome missegregation rates are more sensitive to radiation therapy, supporting chromosomal instability as a promising biomarker of radiation response.
Collapse
Affiliation(s)
- Pippa F. Cosper
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Maha Paracha
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kathryn M. Jones
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Laura Hrycyniak
- Molecular and Cellular Pharmacology Graduate Training Program, University of Wisconsin, Madison, WI 53705, USA
| | - Les Henderson
- Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, WI
| | - Ava Bryan
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Diego Eyzaguirre
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Emily McCunn
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Elizabeth Boulanger
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jun Wan
- Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kwangok P. Nickel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Vanessa Horner
- Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, WI
| | - Rong Hu
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Paul M. Harari
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Randall J. Kimple
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Beth A. Weaver
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
24
|
Deng C, Lan YC, Chen GY, Ekeabu CS, Chung M, Lampson MA, Chenoweth DM. Conditional Localization Pharmacology Manipulates the Cell Cycle with Spatiotemporal Precision. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612697. [PMID: 39314350 PMCID: PMC11419097 DOI: 10.1101/2024.09.12.612697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Traditional pharmacology has limited control of drug activity and localization in space and time. Herein, we described an approach for kinase regulation using conditional localization pharmacology (CLP), where an inactive caged inhibitor is localized to a site of interest in a dormant state using intracellular protein tethering. The activity of the inhibitor can be regulated with spatial and temporal precision in a live cellular environment using light. As a proof of concept, a photocaged MPS1 kinase inhibitor (reversine) bearing a Halo-tag ligand tether was designed to manipulate the cell cycle. We demonstrate that this new caged reversine halo probe (CRH) strategy is capable of efficient localization and exceptional spatiotemporal control over spindle assembly checkpoint (SAC) silencing and mitotic exit.
Collapse
|
25
|
Zerbib J, Ippolito MR, Eliezer Y, De Feudis G, Reuveni E, Savir Kadmon A, Martin S, Viganò S, Leor G, Berstler J, Muenzner J, Mülleder M, Campagnolo EM, Shulman ED, Chang T, Rubolino C, Laue K, Cohen-Sharir Y, Scorzoni S, Taglietti S, Ratti A, Stossel C, Golan T, Nicassio F, Ruppin E, Ralser M, Vazquez F, Ben-David U, Santaguida S. Human aneuploid cells depend on the RAF/MEK/ERK pathway for overcoming increased DNA damage. Nat Commun 2024; 15:7772. [PMID: 39251587 PMCID: PMC11385192 DOI: 10.1038/s41467-024-52176-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: 06/12/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024] Open
Abstract
Aneuploidy is a hallmark of human cancer, yet the molecular mechanisms to cope with aneuploidy-induced cellular stresses remain largely unknown. Here, we induce chromosome mis-segregation in non-transformed RPE1-hTERT cells and derive multiple stable clones with various degrees of aneuploidy. We perform a systematic genomic, transcriptomic and proteomic profiling of 6 isogenic clones, using whole-exome DNA, mRNA and miRNA sequencing, as well as proteomics. Concomitantly, we functionally interrogate their cellular vulnerabilities, using genome-wide CRISPR/Cas9 and large-scale drug screens. Aneuploid clones activate the DNA damage response and are more resistant to further DNA damage induction. Aneuploid cells also exhibit elevated RAF/MEK/ERK pathway activity and are more sensitive to clinically-relevant drugs targeting this pathway, and in particular to CRAF inhibition. Importantly, CRAF and MEK inhibition sensitize aneuploid cells to DNA damage-inducing chemotherapies and to PARP inhibitors. We validate these results in human cancer cell lines. Moreover, resistance of cancer patients to olaparib is associated with high levels of RAF/MEK/ERK signaling, specifically in highly-aneuploid tumors. Overall, our study provides a comprehensive resource for genetically-matched karyotypically-stable cells of various aneuploidy states, and reveals a therapeutically-relevant cellular dependency of aneuploid cells.
Collapse
Affiliation(s)
- Johanna Zerbib
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Marica Rosaria Ippolito
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Yonatan Eliezer
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Giuseppina De Feudis
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Eli Reuveni
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anouk Savir Kadmon
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sara Martin
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Sonia Viganò
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Gil Leor
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Julia Muenzner
- Charité Universitätsmedizin Berlin, Department of Biochemistry, Berlin, Germany
| | - Michael Mülleder
- Charité Universitätsmedizin Berlin, Core Facility High-Throughput Mass Spectrometry, Berlin, Germany
| | - Emma M Campagnolo
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eldad D Shulman
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tiangen Chang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carmela Rubolino
- Center for Genomic Science of IIT@SEMM, Fondazione Instituto Italiano di Technologia, Milan, Italy
| | - Kathrin Laue
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Cohen-Sharir
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Simone Scorzoni
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Silvia Taglietti
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Alice Ratti
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Chani Stossel
- Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Talia Golan
- Oncology Institute, Sheba Medical Center, Tel Hashomer, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Francesco Nicassio
- Center for Genomic Science of IIT@SEMM, Fondazione Instituto Italiano di Technologia, Milan, Italy
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Markus Ralser
- Charité Universitätsmedizin Berlin, Department of Biochemistry, Berlin, Germany
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | | | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Stefano Santaguida
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
26
|
Cheng SY, Yi ZY, Zhang CH, Sun QY, Qian WP, Li J. Vinorelbine administration impedes the timely progression of meiotic maturation and induces aneuploidy in mouse oocytes. Reprod Toxicol 2024; 128:108634. [PMID: 38851359 DOI: 10.1016/j.reprotox.2024.108634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Vinorelbine is a commonly used drug to treat various malignancies, such as breast cancer, non-small cell lung cancer, and metastatic pleural mesothelioma. Its side effects include severe neutropenia, local phlebitis, gastrointestinal reactions, and neurotoxicity. In view of the scarcity of research on vinorelbine's reproductive toxicity, this study evaluated the impact of vinorelbine ditartrate, a commonly used form of vinorelbine, on oocyte maturation in vitro. Our investigation revealed that vinorelbine ditartrate had no effect on oocyte meiotic resumption. However, it did reduce the rate of first polar body extrusion, suggesting that it could significantly impede the meiotic maturation of oocytes. Vinorelbine ditartrate exposure was found to disturb the regular spindle assembly and chromosome alignment, leading to the continuous activation of the spindle assembly checkpoint (SAC) and a delayed activation of the anaphase-promoting complex/cyclosome (APC/C), ultimately causing aneuploidy in oocytes. Consequently, the administration of vinorelbine is likely to result in oocyte aneuploidy, which can be helpful in providing a drug reference and fertility guidance in a clinical context.
Collapse
Affiliation(s)
- Si-Yu Cheng
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zi-Yun Yi
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Chun-Hui Zhang
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.
| | - Wei-Ping Qian
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Jian Li
- Department of Reproductive Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
27
|
Martin S, Scorzoni S, Cordone S, Mazzagatti A, Beznoussenko GV, Gunn AL, Di Bona M, Eliezer Y, Leor G, Ben-Yishay T, Loffreda A, Cancila V, Rainone MC, Ippolito MR, Martis V, Bedin F, Garrè M, Vaites LP, Vasapolli P, Polo S, Parazzoli D, Tripodo C, Mironov AA, Cuomo A, Ben-David U, Bakhoum SF, Hatch EM, Ly P, Santaguida S. A p62-dependent rheostat dictates micronuclei catastrophe and chromosome rearrangements. Science 2024; 385:eadj7446. [PMID: 39208097 PMCID: PMC11664475 DOI: 10.1126/science.adj7446] [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: 07/25/2023] [Revised: 04/29/2024] [Accepted: 07/05/2024] [Indexed: 09/04/2024]
Abstract
Chromosomal instability (CIN) generates micronuclei-aberrant extranuclear structures that catalyze the acquisition of complex chromosomal rearrangements present in cancer. Micronuclei are characterized by persistent DNA damage and catastrophic nuclear envelope collapse, which exposes DNA to the cytoplasm. We found that the autophagic receptor p62/SQSTM1 modulates micronuclear stability, influencing chromosome fragmentation and rearrangements. Mechanistically, proximity of micronuclei to mitochondria led to oxidation-driven homo-oligomerization of p62, limiting endosomal sorting complex required for transport (ESCRT)-dependent micronuclear envelope repair by triggering autophagic degradation. We also found that p62 levels correlate with increased chromothripsis across human cancer cell lines and with increased CIN in colorectal tumors. Thus, p62 acts as a regulator of micronuclei and may serve as a prognostic marker for tumors with high CIN.
Collapse
Affiliation(s)
- Sara Martin
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Simone Scorzoni
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Cordone
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Alice Mazzagatti
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Amanda L. Gunn
- Division of Basic Sciences and Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Melody Di Bona
- Human Oncology and Pathogenesis Program and Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yonatan Eliezer
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gil Leor
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tal Ben-Yishay
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Alessia Loffreda
- Experimental Imaging Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care “G. D’Alessandro,” University of Palermo, Palermo, Italy
| | - Maria Chiara Rainone
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Marica Rosaria Ippolito
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Valentino Martis
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Fabio Bedin
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | | | - Paolo Vasapolli
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Simona Polo
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Parazzoli
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Claudio Tripodo
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care “G. D’Alessandro,” University of Palermo, Palermo, Italy
| | | | - Alessandro Cuomo
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Samuel F. Bakhoum
- Human Oncology and Pathogenesis Program and Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emily M. Hatch
- Division of Basic Sciences and Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter Ly
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stefano Santaguida
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
28
|
Johnson BA, Liu AZ, Bi T, Dong Y, Li T, Zhou D, Narkar A, Wu Y, Sun SX, Larman TC, Zhu J, Li R. Simple aneuploidy evades p53 surveillance and promotes niche factor-independent growth in human intestinal organoids. Mol Biol Cell 2024; 35:br15. [PMID: 38985518 PMCID: PMC11321050 DOI: 10.1091/mbc.e24-04-0166] [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: 04/15/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
Aneuploidy is nearly ubiquitous in tumor genomes, but the role of aneuploidy in the early stages of cancer evolution remains unclear. Here, by inducing heterogeneous aneuploidy in non-transformed human colon organoids (colonoids), we investigated how the effects of aneuploidy on cell growth and differentiation may promote malignant transformation. Previous work implicated p53 activation as a downstream response to aneuploidy induction. We found that simple aneuploidy, characterized by 1-3 gained or lost chromosomes, resulted in little or modest p53 activation and cell cycle arrest when compared with more complex aneuploid cells. Single-cell RNA sequencing analysis revealed that the degree of p53 activation was strongly correlated with karyotype complexity. Single-cell tracking showed that cells could continue to divide despite the observation of one to a few lagging chromosomes. Unexpectedly, colonoids with simple aneuploidy exhibited impaired differentiation after niche factor withdrawal. These findings demonstrate that simple aneuploid cells can escape p53 surveillance and may contribute to niche factor-independent growth of cancer-initiating colon stem cells.
Collapse
Affiliation(s)
- Blake A. Johnson
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Albert Z. Liu
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Tianhao Bi
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Yi Dong
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Taibo Li
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218
| | - Dingjingyu Zhou
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Akshay Narkar
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Yufei Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD 21218
| | - Sean X. Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD 21218
| | - Tatianna C. Larman
- Department of Pathology, Division of Gastrointestinal/Liver Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Jin Zhu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Rong Li
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
| |
Collapse
|
29
|
Sanchez-Vasquez E, Bronner ME, Zernicka-Goetz M. HIF1A contributes to the survival of aneuploid and mosaic pre-implantation embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.04.556218. [PMID: 39071426 PMCID: PMC11275769 DOI: 10.1101/2023.09.04.556218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Human fertility is suboptimal, partly due to error-prone divisions in early cleavage-stages that result in aneuploidy. Most human pre-implantation are mosaics of euploid and aneuploid cells, however, mosaic embryos with a low proportion of aneuploid cells have a similar likelihood of developing to term as fully euploid embryos. How embryos manage aneuploidy during development is poorly understood. This knowledge is crucial for improving fertility treatments and reducing developmental defects. To explore these mechanisms, we established a new mouse model of chromosome mosaicism to study the fate of aneuploid cells during pre-implantation development. We previously used the Mps1 inhibitor reversine to generate aneuploidy in embryos. Here, we found that treatment with the more specific Mps1 inhibitor AZ3146 induced chromosome segregation defects in pre-implantation embryos, similar to reversine. However, AZ3146-treated embryos showed a higher developmental potential than reversine-treated embryos. Unlike reversine-treated embryos, AZ3146-treated embryos exhibited transient upregulation of Hypoxia Inducible-Factor-1A (HIF1A) and lacked p53 upregulation. Pre-implantation embryos develop in a hypoxic environment in vivo, and hypoxia exposure in vitro reduced DNA damage in response to Mps1 inhibition and increased the proportion of euploid cells in the mosaic epiblast. Inhibiting HIF1A in mosaic embryos also decreased the proportion of aneuploid cells in mosaic embryos. Our work illuminates potential strategies to improve the developmental potential of mosaic embryos.
Collapse
Affiliation(s)
| | - Marianne E. Bronner
- Division of Biology 139-74, California Institute of Technology, Pasadena, CA 91125, USA
| | - Magdalena Zernicka-Goetz
- Division of Biology 139-74, California Institute of Technology, Pasadena, CA 91125, USA
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
30
|
Bastianello G, Kidiyoor GR, Lowndes C, Li Q, Bonnal R, Godwin J, Iannelli F, Drufuca L, Bason R, Orsenigo F, Parazzoli D, Pavani M, Cancila V, Piccolo S, Scita G, Ciliberto A, Tripodo C, Pagani M, Foiani M. Mechanical stress during confined migration causes aberrant mitoses and c-MYC amplification. Proc Natl Acad Sci U S A 2024; 121:e2404551121. [PMID: 38990945 PMCID: PMC11260125 DOI: 10.1073/pnas.2404551121] [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: 03/04/2024] [Accepted: 06/07/2024] [Indexed: 07/13/2024] Open
Abstract
Confined cell migration hampers genome integrity and activates the ATR and ATM mechano-transduction pathways. We investigated whether the mechanical stress generated by metastatic interstitial migration contributes to the enhanced chromosomal instability observed in metastatic tumor cells. We employed live cell imaging, micro-fluidic approaches, and scRNA-seq to follow the fate of tumor cells experiencing confined migration. We found that, despite functional ATR, ATM, and spindle assembly checkpoint (SAC) pathways, tumor cells dividing across constriction frequently exhibited altered spindle pole organization, chromosome mis-segregations, micronuclei formation, chromosome fragility, high gene copy number variation, and transcriptional de-regulation and up-regulation of c-MYC oncogenic transcriptional signature via c-MYC locus amplifications. In vivo tumor settings showed that malignant cells populating metastatic foci or infiltrating the interstitial stroma gave rise to cells expressing high levels of c-MYC. Altogether, our data suggest that mechanical stress during metastatic migration contributes to override the checkpoint controls and boosts genotoxic and oncogenic events. Our findings may explain why cancer aneuploidy often does not correlate with mutations in SAC genes and why c-MYC amplification is strongly linked to metastatic tumors.
Collapse
Affiliation(s)
- Giulia Bastianello
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Università degli Studi di Milano, Milan20122, Italy
| | - Gururaj Rao Kidiyoor
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Conor Lowndes
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Qingsen Li
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Raoul Bonnal
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Jeffrey Godwin
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Fabio Iannelli
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | | | - Ramona Bason
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Fabrizio Orsenigo
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Dario Parazzoli
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Mattia Pavani
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Science, University of Palermo School of Medicine, Palermo90133, Italy
| | - Stefano Piccolo
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Department of Molecular Medicine, University of Padua, Padua35123, Italy
| | - Giorgio Scita
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Università degli Studi di Milano, Milan20122, Italy
| | - Andrea Ciliberto
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
| | - Claudio Tripodo
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Tumor Immunology Unit, Department of Health Science, University of Palermo School of Medicine, Palermo90133, Italy
| | - Massimiliano Pagani
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Università degli Studi di Milano, Milan20122, Italy
| | - Marco Foiani
- Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano20139, Italy
- Istituto di Genetica Molecolare, Centro Nazionale Ricerca, Pavia27100, Italy
- Cancer Science Institute of Singapore, National University of Singapore, Singapore117599, Singapore
| |
Collapse
|
31
|
Hosea R, Duan W, Meliala ITS, Li W, Wei M, Hillary S, Zhao H, Miyagishi M, Wu S, Kasim V. YY2/BUB3 Axis promotes SAC Hyperactivation and Inhibits Colorectal Cancer Progression via Regulating Chromosomal Instability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308690. [PMID: 38682484 PMCID: PMC11234461 DOI: 10.1002/advs.202308690] [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: 11/13/2023] [Revised: 03/08/2024] [Indexed: 05/01/2024]
Abstract
Spindle assembly checkpoint (SAC) is a crucial safeguard mechanism of mitosis fidelity that ensures equal division of duplicated chromosomes to the two progeny cells. Impaired SAC can lead to chromosomal instability (CIN), a well-recognized hallmark of cancer that facilitates tumor progression; paradoxically, high CIN levels are associated with better therapeutic response and prognosis. However, the mechanism by which CIN determines tumor cell survival and therapeutic response remains poorly understood. Here, using a cross-omics approach, YY2 is identified as a mitotic regulator that promotes SAC activity by activating the transcription of budding uninhibited by benzimidazole 3 (BUB3), a component of SAC. While both conditions induce CIN, a defect in YY2/SAC activity enhances mitosis and tumor growth. Meanwhile, hyperactivation of SAC mediated by YY2/BUB3 triggers a delay in mitosis and suppresses growth. Furthermore, it is revealed that YY2/BUB3-mediated excessive CIN causes higher cell death rates and drug sensitivity, whereas residual tumor cells that survived DNA damage-based therapy have moderate CIN and increased drug resistance. These results provide insights into the role of SAC activity and CIN levels in influencing tumor cell survival and drug response, as well as suggest a novel anti-tumor therapeutic strategy that combines SAC activity modulators and DNA-damage agents.
Collapse
Affiliation(s)
- Rendy Hosea
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Wei Duan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Ian Timothy Sembiring Meliala
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Wenfang Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Mankun Wei
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Sharon Hillary
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
| | - Hezhao Zhao
- Department of Gastrointestinal Surgery, Chongqing University Cancer HospitalChongqing UniversityChongqing400030P. R. China
| | - Makoto Miyagishi
- Life Science Innovation, School of Integrative and Global MajorsUniversity of TsukubaTsukubaIbaraki305‐0006Japan
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer HospitalChongqing UniversityChongqing400030P. R. China
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of BioengineeringChongqing UniversityChongqing400045P. R. China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of BioengineeringChongqing UniversityChongqing400044P. R. China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer HospitalChongqing UniversityChongqing400030P. R. China
| |
Collapse
|
32
|
Ha G, Dieterle P, Shen H, Amir A, Needleman DJ. Measuring and modeling the dynamics of mitotic error correction. Proc Natl Acad Sci U S A 2024; 121:e2323009121. [PMID: 38875144 PMCID: PMC11194551 DOI: 10.1073/pnas.2323009121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/11/2024] [Indexed: 06/16/2024] Open
Abstract
Error correction is central to many biological systems and is critical for protein function and cell health. During mitosis, error correction is required for the faithful inheritance of genetic material. When functioning properly, the mitotic spindle segregates an equal number of chromosomes to daughter cells with high fidelity. Over the course of spindle assembly, many initially erroneous attachments between kinetochores and microtubules are fixed through the process of error correction. Despite the importance of chromosome segregation errors in cancer and other diseases, there is a lack of methods to characterize the dynamics of error correction and how it can go wrong. Here, we present an experimental method and analysis framework to quantify chromosome segregation error correction in human tissue culture cells with live cell confocal imaging, timed premature anaphase, and automated counting of kinetochores after cell division. We find that errors decrease exponentially over time during spindle assembly. A coarse-grained model, in which errors are corrected in a chromosome-autonomous manner at a constant rate, can quantitatively explain both the measured error correction dynamics and the distribution of anaphase onset times. We further validated our model using perturbations that destabilized microtubules and changed the initial configuration of chromosomal attachments. Taken together, this work provides a quantitative framework for understanding the dynamics of mitotic error correction.
Collapse
Affiliation(s)
- Gloria Ha
- Department of Systems Biology, Harvard Medical School, Boston, MA02115
| | - Paul Dieterle
- Department of Physics, Harvard University, Cambridge, MA02138
| | - Hao Shen
- Reverie Labs, Cambridge, MA02139
| | - Ariel Amir
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot7610001, Israel
| | - Daniel J. Needleman
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA02138
- Center for Computational Biology, Flatiron Institute, New York, NY10010
| |
Collapse
|
33
|
Leça N, Barbosa F, Rodriguez-Calado S, Moura M, Pedroso PD, Pinto I, Verza AE, Bange T, Sunkel CE, Barisic M, Maresca TJ, Conde C. Proximity-based activation of AURORA A by MPS1 potentiates error correction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598300. [PMID: 38948877 PMCID: PMC11213139 DOI: 10.1101/2024.06.11.598300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Faithfull cell division relies on mitotic chromosomes becoming bioriented with each pair of sister kinetochores bound to microtubules oriented toward opposing spindle poles. Erroneous kinetochore-microtubule attachments often form during early mitosis, but are destabilized through the phosphorylation of outer kinetochore proteins by centromeric AURORA B kinase (ABK) and centrosomal AURORA A kinase (AAK), thus allowing for re-establishment of attachments until biorientation is achieved. MPS1-mediated phosphorylation of NDC80 has also been shown to directly weaken the kinetochore-microtubule interface in yeast. In human cells, MPS1 has been proposed to transiently accumulate at end-on attached kinetochores and phosphorylate SKA3 to promote microtubule release. Whether MPS1 directly targets NDC80 and/or promotes the activity of AURORA kinases in metazoans remains unclear. Here, we report a novel mechanism involving communication between kinetochores and centrosomes, wherein MPS1 acts upstream of AAK to promote error correction. MPS1 on pole-proximal kinetochores phosphorylates the C-lobe of AAK thereby increasing its activation at centrosomes. This proximity-based activation ensures the establishment of a robust AAK activity gradient that locally destabilizes mal-oriented kinetochores near spindle poles. Accordingly, MPS1 depletion from Drosophila cells causes severe chromosome misalignment and erroneous kinetochore-microtubule attachments, which can be rescued by tethering either MPS1 or constitutively active AAK mutants to centrosomes. Proximity-based activation of AAK by MPS1 also occurs in human cells to promote AAK-mediated phosphorylation of the NDC80 N-terminal tail. These findings uncover an MPS1-AAK cross-talk that is required for efficient error correction, showcasing the ability of kinetochores to modulate centrosome outputs to ensure proper chromosome segregation.
Collapse
|
34
|
Takaki T, Millar R, Hiley CT, Boulton SJ. Micronuclei induced by radiation, replication stress, or chromosome segregation errors do not activate cGAS-STING. Mol Cell 2024; 84:2203-2213.e5. [PMID: 38749421 DOI: 10.1016/j.molcel.2024.04.017] [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: 01/02/2024] [Revised: 03/15/2024] [Accepted: 04/23/2024] [Indexed: 06/09/2024]
Abstract
The cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway plays a pivotal role in innate immune responses to viral infection and inhibition of autoimmunity. Recent studies have suggested that micronuclei formed by genotoxic stress can activate innate immune signaling via the cGAS-STING pathway. Here, we investigated cGAS localization, activation, and downstream signaling from micronuclei induced by ionizing radiation, replication stress, and chromosome segregation errors. Although cGAS localized to ruptured micronuclei via binding to self-DNA, we failed to observe cGAS activation; cGAMP production; downstream phosphorylation of STING, TBK1, or IRF3; nuclear accumulation of IRF3; or expression of interferon-stimulated genes. Failure to activate the cGAS-STING pathway was observed across primary and immortalized cell lines, which retained the ability to activate the cGAS-STING pathway in response to dsDNA or modified vaccinia virus infection. We provide evidence that micronuclei formed by genotoxic insults contain histone-bound self-DNA, which we show is inhibitory to cGAS activation in cells.
Collapse
Affiliation(s)
- Tohru Takaki
- DSB Repair Metabolism Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Rhona Millar
- DSB Repair Metabolism Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Cancer Research UK Radnet City of London Centre, UCL Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Crispin T Hiley
- Cancer Research UK Radnet City of London Centre, UCL Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Simon J Boulton
- DSB Repair Metabolism Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Artios Pharma Ltd., Babraham Research Campus, Meditrina Building, Cambridge CB22 3AT, UK.
| |
Collapse
|
35
|
Zahm JA, Harrison SC. A communication hub for phosphoregulation of kinetochore-microtubule attachment. Curr Biol 2024; 34:2308-2318.e6. [PMID: 38776904 PMCID: PMC11847324 DOI: 10.1016/j.cub.2024.04.067] [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: 11/22/2023] [Revised: 04/06/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
The Mps1 and Aurora B kinases regulate and monitor kinetochore attachment to spindle microtubules during cell division, ultimately ensuring accurate chromosome segregation. In yeast, the critical spindle attachment components are the Ndc80 and Dam1 complexes (Ndc80c and DASH/Dam1c, respectively). Ndc80c is a 600-Å-long heterotetramer that binds microtubules through a globular "head" at one end and centromere-proximal kinetochore components through a globular knob at the other end. Dam1c is a heterodecamer that forms a ring of 16-17 protomers around the shaft of the single kinetochore microtubule in point-centromere yeast. The ring coordinates the approximately eight Ndc80c rods per kinetochore. In published work, we showed that a site on the globular "head" of Ndc80c, including residues from both Ndc80 and Nuf2, binds a bipartite segment in the long C-terminal extension of Dam1. Results reported here show, both by in vitro binding experiments and by crystal structure determination, that the same site binds a conserved segment in the long N-terminal extension of Mps1. It also binds, less tightly, a conserved segment in the N-terminal extension of Ipl1 (yeast Aurora B). Together with results from experiments in yeast cells and from biochemical assays reported in two accompanying papers, the structures and graded affinities identify a communication hub for ensuring uniform bipolar attachment and for signaling anaphase onset.
Collapse
Affiliation(s)
- Jacob A Zahm
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Stephen C Harrison
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA.
| |
Collapse
|
36
|
Parnell EJ, Jenson EE, Miller MP. A conserved site on Ndc80 complex facilitates dynamic recruitment of Mps1 to yeast kinetochores to promote accurate chromosome segregation. Curr Biol 2024; 34:2294-2307.e4. [PMID: 38776906 PMCID: PMC11178286 DOI: 10.1016/j.cub.2024.04.054] [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: 11/09/2023] [Revised: 03/27/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
Accurate chromosome segregation relies on kinetochores carrying out multiple functions, including establishing and maintaining microtubule attachments, forming precise bi-oriented attachments between sister chromatids, and activating the spindle assembly checkpoint. Central to these processes is the highly conserved Ndc80 complex. This kinetochore subcomplex interacts directly with microtubules but also serves as a critical platform for recruiting kinetochore-associated factors and as a key substrate for error correction kinases. The precise manner in which these kinetochore factors interact and regulate each other's function remains unknown, considerably hindering our understanding of how Ndc80 complex-dependent processes function together to orchestrate accurate chromosome segregation. Here, we aimed to uncover the role of Nuf2's CH domain, a component of the Ndc80 complex, in ensuring these processes. Through extensive mutational analysis, we identified a conserved interaction domain composed of two segments in Nuf2's CH domain that form the binding site for Mps1 within the yeast Ndc80 complex. Interestingly, this site also associates with the Dam1 complex, suggesting Mps1 recruitment may be subject to regulation by competitive binding with other factors. Mutants disrupting this "interaction hub" exhibit defects in spindle assembly checkpoint function and severe chromosome segregation errors. Significantly, specifically restoring Mps1-Ndc80 complex association rescues these defects. Our findings shed light on the intricate regulation of Ndc80 complex-dependent functions and highlight the essential role of Mps1 in kinetochore bi-orientation and accurate chromosome segregation.
Collapse
Affiliation(s)
- Emily J Parnell
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Erin E Jenson
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Matthew P Miller
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.
| |
Collapse
|
37
|
Lacefield S. Chromosome segregation: Mps1 and Dam1 battle to bind a shared interaction site at the kinetochore. Curr Biol 2024; 34:R530-R533. [PMID: 38834024 PMCID: PMC11932353 DOI: 10.1016/j.cub.2024.04.070] [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: 06/06/2024]
Abstract
The attachment of kinetochores to spindle microtubules is highly regulated to ensure proper chromosome segregation. Three new studies identify an interaction hub at the kinetochore that integrates kinetochore attachment state with spindle checkpoint activity and kinetochore assembly.
Collapse
Affiliation(s)
- Soni Lacefield
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
| |
Collapse
|
38
|
Chen X, Zhong Y, Wang S, Xu S, Chen J, Cheng X, Yang X. Reversine inhibits proliferation and induces apoptosis of human osteosarcoma cells through targeting MEK1. J Bone Oncol 2024; 46:100601. [PMID: 38706714 PMCID: PMC11063522 DOI: 10.1016/j.jbo.2024.100601] [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: 11/19/2023] [Revised: 03/03/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024] Open
Abstract
Reversine, or 2-(4-morpholinoanilino)-6-cyclohexylaminopurine, is a 2,6-disubstituted purine derivative. This small molecule shows anti-tumor potential by playing a central role in the inhibition of several kinases related to cell cycle regulation and cytokinesis. In this study, systematic review demonstrated the feasibility and pharmacological mechanism of anti-tumor effect of reversine. Firstly, we grafted MNNG/HOS, U-2 OS, MG-63 osteosarcoma cell aggregates onto chicken embryonic chorioallantoic membrane (CAM) to examine the tumor volume of these grafts after reversine treatment. Following culture, reversine inhibited the growth of osteosarcoma cell aggregates on CAM significantly. In vitro experiment, reversine suppressed osteosarcoma cell viability, colony formation, proliferation, and induced apoptosis and cell cycle arrest at G0-G1 phase. Scratch wound assay demonstrated that reversine restrained cell migration. Reversine increased the protein expression of E-cadherin. The mRNA expression of Rac1, RhoA, CDC42, PTK2, PXN, N-cadherin, Vimentin in MNNG/HOS, U-2 OS and MG-63 cells were suppressed and PTEN increased after reversine treatment. Network pharmacology prediction, molecular docking and systematic review revealed MEK1 can be used as an effective target for reversine to inhibit osteosarcoma. Western blot results show the regulation of MEK1 and ERK1/2 by reversine was not consistent in different osteosarcoma cell lines, but we found that reversine significantly inhibited the protein expression of MEK1 in MNNG/HOS, U-2 OS and MG-63. All these suggested that reversine can exert its anti-tumor effect by targeting the expression of MEK1.
Collapse
Affiliation(s)
- Xianlong Chen
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Yeyin Zhong
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Simiao Wang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
| | - Shujie Xu
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Junyuan Chen
- Center for Bone, Joint and Sports Medicine, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Xin Cheng
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Xuesong Yang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou 510632, China
- Clinical Research Center, Clifford Hospital, Guangzhou 511495, China
| |
Collapse
|
39
|
Hayes BH, Wang M, Zhu H, Phan SH, Dooling LJ, Andrechak JC, Chang AH, Tobin MP, Ontko NM, Marchena T, Discher DE. Chromosomal instability induced in cancer can enhance macrophage-initiated immune responses that include anti-tumor IgG. eLife 2024; 12:RP88054. [PMID: 38805560 PMCID: PMC11132682 DOI: 10.7554/elife.88054] [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] [Indexed: 05/30/2024] Open
Abstract
Solid tumors generally exhibit chromosome copy number variation, which is typically caused by chromosomal instability (CIN) in mitosis. The resulting aneuploidy can drive evolution and associates with poor prognosis in various cancer types as well as poor response to T-cell checkpoint blockade in melanoma. Macrophages and the SIRPα-CD47 checkpoint are understudied in such contexts. Here, CIN is induced in poorly immunogenic B16F10 mouse melanoma cells using spindle assembly checkpoint MPS1 inhibitors that generate persistent micronuclei and diverse aneuploidy while skewing macrophages toward a tumoricidal 'M1-like' phenotype based on markers and short-term anti-tumor studies. Mice bearing CIN-afflicted tumors with wild-type CD47 levels succumb similar to controls, but long-term survival is maximized by SIRPα blockade on adoptively transferred myeloid cells plus anti-tumor monoclonal IgG. Such cells are the initiating effector cells, and survivors make de novo anti-cancer IgG that not only promote phagocytosis of CD47-null cells but also suppress tumor growth. CIN does not affect the IgG response, but pairing CIN with maximal macrophage anti-cancer activity increases durable cures that possess a vaccination-like response against recurrence.
Collapse
Affiliation(s)
- Brandon H Hayes
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| | - Mai Wang
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Hui Zhu
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Steven H Phan
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Lawrence J Dooling
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Jason C Andrechak
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| | - Alexander H Chang
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Michael P Tobin
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| | - Nicholas M Ontko
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Tristan Marchena
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Dennis E Discher
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
40
|
Ya A, Deng C, Godek KM. Cell Competition Eliminates Aneuploid Human Pluripotent Stem Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593217. [PMID: 38766106 PMCID: PMC11100710 DOI: 10.1101/2024.05.08.593217] [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: 05/22/2024]
Abstract
Human pluripotent stem cells (hPSCs) maintain diploid populations for generations despite a persistently high rate of mitotic errors that cause aneuploidy, or chromosome imbalances. Consequently, to maintain genome stability, aneuploidy must inhibit hPSC proliferation, but the mechanisms are unknown. Here, we surprisingly find that homogeneous aneuploid populations of hPSCs proliferate unlike aneuploid non-transformed somatic cells. Instead, in mosaic populations, cell non-autonomous competition between neighboring diploid and aneuploid hPSCs eliminates less fit aneuploid cells. Aneuploid hPSCs with lower Myc or higher p53 levels relative to diploid neighbors are outcompeted but conversely gain a selective advantage when Myc and p53 relative abundance switches. Thus, although hPSCs frequently missegregate chromosomes and inherently tolerate aneuploidy, Myc- and p53-driven cell competition preserves their genome integrity. These findings have important implications for the use of hPSCs in regenerative medicine and for how diploid human embryos are established despite the prevalence of aneuploidy during early development.
Collapse
Affiliation(s)
- Amanda Ya
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Chenhui Deng
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Kristina M. Godek
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
- Lead contact
| |
Collapse
|
41
|
Watson EV, Lee JJK, Gulhan DC, Melloni GEM, Venev SV, Magesh RY, Frederick A, Chiba K, Wooten EC, Naxerova K, Dekker J, Park PJ, Elledge SJ. Chromosome evolution screens recapitulate tissue-specific tumor aneuploidy patterns. Nat Genet 2024; 56:900-912. [PMID: 38388848 PMCID: PMC11096114 DOI: 10.1038/s41588-024-01665-2] [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: 03/16/2022] [Accepted: 01/16/2024] [Indexed: 02/24/2024]
Abstract
Whole chromosome and arm-level copy number alterations occur at high frequencies in tumors, but their selective advantages, if any, are poorly understood. Here, utilizing unbiased whole chromosome genetic screens combined with in vitro evolution to generate arm- and subarm-level events, we iteratively selected the fittest karyotypes from aneuploidized human renal and mammary epithelial cells. Proliferation-based karyotype selection in these epithelial lines modeled tissue-specific tumor aneuploidy patterns in patient cohorts in the absence of driver mutations. Hi-C-based translocation mapping revealed that arm-level events usually emerged in multiples of two via centromeric translocations and occurred more frequently in tetraploids than diploids, contributing to the increased diversity in evolving tetraploid populations. Isogenic clonal lineages enabled elucidation of pro-tumorigenic mechanisms associated with common copy number alterations, revealing Notch signaling potentiation as a driver of 1q gain in breast cancer. We propose that intrinsic, tissue-specific proliferative effects underlie tumor copy number patterns in cancer.
Collapse
Affiliation(s)
- Emma V Watson
- Department of Genetics, Harvard Medical School and Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jake June-Koo Lee
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Doga C Gulhan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Giorgio E M Melloni
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Sergey V Venev
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Rayna Y Magesh
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Abdulrazak Frederick
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kunitoshi Chiba
- Department of Genetics, Harvard Medical School and Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
| | - Eric C Wooten
- Department of Genetics, Harvard Medical School and Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
| | - Kamila Naxerova
- Department of Genetics, Harvard Medical School and Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Job Dekker
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
| | - Stephen J Elledge
- Department of Genetics, Harvard Medical School and Department of Medicine, Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
42
|
Guscott MA, McClelland SE. Experimental evolution of cancer chromosomal changes. Nat Genet 2024; 56:743-745. [PMID: 38684897 DOI: 10.1038/s41588-024-01742-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Affiliation(s)
- Molly A Guscott
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | | |
Collapse
|
43
|
Kelley ME, Carlini L, Kornakov N, Aher A, Khodjakov A, Kapoor TM. Spastin regulates anaphase chromosome separation distance and microtubule-containing nuclear tunnels. Mol Biol Cell 2024; 35:ar48. [PMID: 38335450 PMCID: PMC11064660 DOI: 10.1091/mbc.e24-01-0031-t] [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: 01/24/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Nuclear envelope reassembly during the final stages of each mitosis depends on disassembling spindle microtubules without disrupting chromosome separation. This process involves the transient recruitment of the ESCRT-III complex and spastin, a microtubule-severing AAA (ATPases associated with diverse cellular activities) mechanoenzyme, to late-anaphase chromosomes. However, dissecting mechanisms underlying these rapid processes, which can be completed within minutes, has been difficult. Here, we combine fast-acting chemical inhibitors with live-cell imaging and find that spindle microtubules, along with spastin activity, regulate the number and lifetimes of spastin foci at anaphase chromosomes. Unexpectedly, spastin inhibition impedes chromosome separation, but does not alter the anaphase localization dynamics of CHMP4B, an ESCRT-III protein, or increase γ-H2AX foci, a DNA damage marker. We show spastin inhibition increases the frequency of lamin-lined nuclear microtunnels that can include microtubules penetrating the nucleus. Our findings suggest failure to sever spindle microtubules impedes chromosome separation, yet reforming nuclear envelopes can topologically accommodate persistent microtubules ensuring nuclear DNA is not damaged or exposed to cytoplasm.
Collapse
Affiliation(s)
- Megan E. Kelley
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY 10065
| | - Lina Carlini
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY 10065
| | - Nikolay Kornakov
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY 10065
| | - Amol Aher
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY 10065
| | - Alexey Khodjakov
- Wadsworth Center, New York State Department of Health, Albany, NY 12237
| | - Tarun M. Kapoor
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY 10065
| |
Collapse
|
44
|
Sun SM, Zhao BW, Li YY, Liu HY, Xu YH, Yang XM, Guo JN, Ouyang YC, Weng CJ, Guan YC, Sun QY, Wang ZB. Loss of UBE2S causes meiosis I arrest with normal spindle assembly checkpoint dynamics in mouse oocytes. Development 2024; 151:dev202285. [PMID: 38546043 DOI: 10.1242/dev.202285] [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: 08/29/2023] [Accepted: 01/25/2024] [Indexed: 04/04/2024]
Abstract
The timely degradation of proteins that regulate the cell cycle is essential for oocyte maturation. Oocytes are equipped to degrade proteins via the ubiquitin-proteasome system. In meiosis, anaphase promoting complex/cyclosome (APC/C), an E3 ubiquitin-ligase, is responsible for the degradation of proteins. Ubiquitin-conjugating enzyme E2 S (UBE2S), an E2 ubiquitin-conjugating enzyme, delivers ubiquitin to APC/C. APC/C has been extensively studied, but the functions of UBE2S in oocyte maturation and mouse fertility are not clear. In this study, we used Ube2s knockout mice to explore the role of UBE2S in mouse oocytes. Ube2s-deleted oocytes were characterized by meiosis I arrest with normal spindle assembly and spindle assembly checkpoint dynamics. However, the absence of UBE2S affected the activity of APC/C. Cyclin B1 and securin are two substrates of APC/C, and their levels were consistently high, resulting in the failure of homologous chromosome separation. Unexpectedly, the oocytes arrested in meiosis I could be fertilized and the embryos could become implanted normally, but died before embryonic day 10.5. In conclusion, our findings reveal an indispensable regulatory role of UBE2S in mouse oocyte meiosis and female fertility.
Collapse
Affiliation(s)
- Si-Min Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Bing-Wang Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yuan-Yuan Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Hong-Yang Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yuan-Hong Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xue-Mei Yang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Jia-Ni Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chang-Jiang Weng
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yi-Chun Guan
- Center for Reproductive Medicine, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Qing-Yuan Sun
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
45
|
Di Tommaso E, Giunta S. Dynamic interplay between human alpha-satellite DNA structure and centromere functions. Semin Cell Dev Biol 2024; 156:130-140. [PMID: 37926668 DOI: 10.1016/j.semcdb.2023.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/04/2023] [Accepted: 10/10/2023] [Indexed: 11/07/2023]
Abstract
Maintenance of genome stability relies on functional centromeres for correct chromosome segregation and faithful inheritance of the genetic information. The human centromere is the primary constriction within mitotic chromosomes made up of repetitive alpha-satellite DNA hierarchically organized in megabase-long arrays of near-identical higher order repeats (HORs). Centromeres are epigenetically specified by the presence of the centromere-specific histone H3 variant, CENP-A, which enables the assembly of the kinetochore for microtubule attachment. Notably, centromeric DNA is faithfully inherited as intact haplotypes from the parents to the offspring without intervening recombination, yet, outside of meiosis, centromeres are akin to common fragile sites (CFSs), manifesting crossing-overs and ongoing sequence instability. Consequences of DNA changes within the centromere are just starting to emerge, with unclear effects on intra- and inter-generational inheritance driven by centromere's essential role in kinetochore assembly. Here, we review evidence of meiotic selection operating to mitigate centromere drive, as well as recent reports on centromere damage, recombination and repair during the mitotic cell division. We propose an antagonistic pleiotropy interpretation to reconcile centromere DNA instability as both driver of aneuploidy that underlies degenerative diseases, while also potentially necessary for the maintenance of homogenized HORs for centromere function. We attempt to provide a framework for this conceptual leap taking into consideration the structural interface of centromere-kinetochore interaction and present case scenarios for its malfunctioning. Finally, we offer an integrated working model to connect DNA instability, chromatin, and structural changes with functional consequences on chromosome integrity.
Collapse
Affiliation(s)
- Elena Di Tommaso
- Laboratory of Genome Evolution, Department of Biology & Biotechnology Charles Darwin, Sapienza University of Rome, Rome 00185, Italy
| | - Simona Giunta
- Laboratory of Genome Evolution, Department of Biology & Biotechnology Charles Darwin, Sapienza University of Rome, Rome 00185, Italy.
| |
Collapse
|
46
|
Li S, Kasciukovic T, Tanaka TU. Kinetochore-microtubule error correction for biorientation: lessons from yeast. Biochem Soc Trans 2024; 52:29-39. [PMID: 38305688 PMCID: PMC10903472 DOI: 10.1042/bst20221261] [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: 11/23/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/03/2024]
Abstract
Accurate chromosome segregation in mitosis relies on sister kinetochores forming stable attachments to microtubules (MTs) extending from opposite spindle poles and establishing biorientation. To achieve this, erroneous kinetochore-MT interactions must be resolved through a process called error correction, which dissolves improper kinetochore-MT attachment and allows new interactions until biorientation is achieved. The Aurora B kinase plays key roles in driving error correction by phosphorylating Dam1 and Ndc80 complexes, while Mps1 kinase, Stu2 MT polymerase and phosphatases also regulate this process. Once biorientation is formed, tension is applied to kinetochore-MT interaction, stabilizing it. In this review article, we discuss the mechanisms of kinetochore-MT interaction, error correction and biorientation. We focus mainly on recent insights from budding yeast, where the attachment of a single MT to a single kinetochore during biorientation simplifies the analysis of error correction mechanisms.
Collapse
Affiliation(s)
- Shuyu Li
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Taciana Kasciukovic
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| | - Tomoyuki U. Tanaka
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K
| |
Collapse
|
47
|
Horakova A, Konecna M, Anger M. Chromosome Division in Early Embryos-Is Everything under Control? And Is the Cell Size Important? Int J Mol Sci 2024; 25:2101. [PMID: 38396778 PMCID: PMC10889803 DOI: 10.3390/ijms25042101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Chromosome segregation in female germ cells and early embryonic blastomeres is known to be highly prone to errors. The resulting aneuploidy is therefore the most frequent cause of termination of early development and embryo loss in mammals. And in specific cases, when the aneuploidy is actually compatible with embryonic and fetal development, it leads to severe developmental disorders. The main surveillance mechanism, which is essential for the fidelity of chromosome segregation, is the Spindle Assembly Checkpoint (SAC). And although all eukaryotic cells carry genes required for SAC, it is not clear whether this pathway is active in all cell types, including blastomeres of early embryos. In this review, we will summarize and discuss the recent progress in our understanding of the mechanisms controlling chromosome segregation and how they might work in embryos and mammalian embryos in particular. Our conclusion from the current literature is that the early mammalian embryos show limited capabilities to react to chromosome segregation defects, which might, at least partially, explain the widespread problem of aneuploidy during the early development in mammals.
Collapse
Affiliation(s)
- Adela Horakova
- Department of Genetics and Reproductive Biotechnologies, Veterinary Research Institute, 621 00 Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Science, 277 21 Libechov, Czech Republic
- Faculty of Science, Masaryk University, 602 00 Brno, Czech Republic
| | - Marketa Konecna
- Department of Genetics and Reproductive Biotechnologies, Veterinary Research Institute, 621 00 Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Science, 277 21 Libechov, Czech Republic
- Faculty of Science, Masaryk University, 602 00 Brno, Czech Republic
| | - Martin Anger
- Department of Genetics and Reproductive Biotechnologies, Veterinary Research Institute, 621 00 Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Science, 277 21 Libechov, Czech Republic
| |
Collapse
|
48
|
Miyamoto H, Kobayashi H, Kishima N, Yamazaki K, Hamamichi S, Uno N, Abe S, Hiramuki Y, Kazuki K, Tomizuka K, Kazuki Y. Rapid human genomic DNA cloning into mouse artificial chromosome via direct chromosome transfer from human iPSC and CRISPR/Cas9-mediated translocation. Nucleic Acids Res 2024; 52:1498-1511. [PMID: 38180813 PMCID: PMC10853801 DOI: 10.1093/nar/gkad1218] [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: 08/29/2023] [Revised: 11/26/2023] [Accepted: 01/03/2024] [Indexed: 01/07/2024] Open
Abstract
A 'genomically' humanized animal stably maintains and functionally expresses the genes on human chromosome fragment (hCF; <24 Mb) loaded onto mouse artificial chromosome (MAC); however, cloning of hCF onto the MAC (hCF-MAC) requires a complex process that involves multiple steps of chromosome engineering through various cells via chromosome transfer and Cre-loxP chromosome translocation. Here, we aimed to develop a strategy to rapidly construct the hCF-MAC by employing three alternative techniques: (i) application of human induced pluripotent stem cells (hiPSCs) as chromosome donors for microcell-mediated chromosome transfer (MMCT), (ii) combination of paclitaxel (PTX) and reversine (Rev) as micronucleation inducers and (iii) CRISPR/Cas9 genome editing for site-specific translocations. We achieved a direct transfer of human chromosome 6 or 21 as a model from hiPSCs as alternative human chromosome donors into CHO cells containing MAC. MMCT was performed with less toxicity through induction of micronucleation by PTX and Rev. Furthermore, chromosome translocation was induced by simultaneous cleavage between human chromosome and MAC by using CRISPR/Cas9, resulting in the generation of hCF-MAC containing CHO clones without Cre-loxP recombination and drug selection. Our strategy facilitates rapid chromosome cloning and also contributes to the functional genomic analyses of human chromosomes.
Collapse
Affiliation(s)
- Hitomaru Miyamoto
- Department of Chromosome Biomedical Engineering, Integrated Medical Sciences, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Hiroaki Kobayashi
- Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Nanami Kishima
- Department of Chromosome Biomedical Engineering, Integrated Medical Sciences, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Kyotaro Yamazaki
- Chromosome Engineering Research Group, The Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - Shusei Hamamichi
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Narumi Uno
- Laboratory of Bioengineering, Faculty of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Satoshi Abe
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Yosuke Hiramuki
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Kanako Kazuki
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Kazuma Tomizuka
- Laboratory of Bioengineering, Faculty of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yasuhiro Kazuki
- Department of Chromosome Biomedical Engineering, Integrated Medical Sciences, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
- Department of Chromosome Biomedical Engineering, School of Life Science, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
- Chromosome Engineering Research Group, The Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| |
Collapse
|
49
|
Zhang Y, Young R, Garvanska DH, Song C, Zhai Y, Wang Y, Jiang H, Fang J, Nilsson J, Alfieri C, Zhang G. Functional analysis of Cdc20 reveals a critical role of CRY box in mitotic checkpoint signaling. Commun Biol 2024; 7:164. [PMID: 38337031 PMCID: PMC10858191 DOI: 10.1038/s42003-024-05859-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Accurate mitosis is coordinated by the spindle assembly checkpoint (SAC) through the mitotic checkpoint complex (MCC), which inhibits the anaphase-promoting complex or cyclosome (APC/C). As an essential regulator, Cdc20 promotes mitotic exit through activating APC/C and monitors kinetochore-microtubule attachment through activating SAC. Cdc20 requires multiple interactions with APC/C and MCC subunits to elicit these functions. Functionally assessing these interactions within cells requires efficient depletion of endogenous Cdc20, which is highly difficult to achieve by RNA interference (RNAi). Here we generated Cdc20 RNAi-sensitive cell lines which display a penetrant metaphase arrest by a single RNAi treatment. In this null background, we accurately measured the contribution of each known motif of Cdc20 on APC/C and SAC activation. The CRY box, a previously identified degron, was found critical for SAC by promoting MCC formation and its interaction with APC/C. These data reveal additional regulation within the SAC and establish a novel method to interrogate Cdc20.
Collapse
Affiliation(s)
- Yuqing Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Rose Young
- Chester Beatty Laboratories, Structural Biology Division, Institute of Cancer Research, London, UK
| | | | - Chunlin Song
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yujing Zhai
- School of Public Health, Qingdao University, Qingdao, China
| | - Ying Wang
- School of Public Health, Qingdao University, Qingdao, China
| | - Hongfei Jiang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jing Fang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jakob Nilsson
- The NNF Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Claudio Alfieri
- Chester Beatty Laboratories, Structural Biology Division, Institute of Cancer Research, London, UK.
| | - Gang Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.
| |
Collapse
|
50
|
Sun Y, Chen Z, Liu G, Chen X, Shi Z, Feng H, Yu L, Li G, Ding K, Huang H, Zhang Z, Xu S. Discovery of a potent and selective covalent threonine tyrosine kinase (TTK) inhibitor. Bioorg Chem 2024; 143:107053. [PMID: 38159497 DOI: 10.1016/j.bioorg.2023.107053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/07/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Threonine tyrosine kinase (TTK) is a critical component of the spindle assembly checkpoint and plays a pivotal role in mitosis. TTK has been identified as a potential therapeutic target for human cancers. Here, we describe our design, synthesis and evaluation of a class of covalent TTK inhibitors, exemplified by 16 (SYL1073). Compound 16 potently inhibits TTK kinase with an IC50 of 0.016 μM and displays improved selectivity in a panel of kinases. Mass spectrometry analysis reveals that 16 covalently binds to the C604 cysteine residue in the hinge region of the TTK kinase domain. Furthermore, 16 achieves strong potency in inhibiting the growth of various human cancer cell lines, outperforming its relative reversible inhibitor, and eliciting robust downstream effects. Taken together, compound 16 provides a valuable lead compound for further optimization toward the development of drug for treatment of human cancers.
Collapse
Affiliation(s)
- Yaoliang Sun
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhiwen Chen
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Guobin Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoai Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zihan Shi
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Science, 19 Yuquan Road, Beijing 100049, China
| | - Huixu Feng
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lei Yu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Guodong Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - He Huang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Zhang Zhang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Shilin Xu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; University of Chinese Academy of Science, 19 Yuquan Road, Beijing 100049, China.
| |
Collapse
|