1
|
Hou Y, Li S, Hou Q, Wang R, Xu X, Li Z, Ma L, Liu Q, Shen Y, Zheng H. Vitamin K2 mitigates cognitive, and motor impairments induced by multiple surgery with anesthesia and analgesia in neonatal stage. Biochem Biophys Res Commun 2025; 765:151784. [PMID: 40273626 DOI: 10.1016/j.bbrc.2025.151784] [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: 01/18/2025] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025]
Abstract
Anesthesia and analgesia are essential components for surgical procedures. While the neurotoxic effects of multiple anesthesia exposures during brain development are well established, the combined impact of multiple surgery with anesthesia and analgesia exposures on neurodevelopmental remains unknown. In this study, neonatal mice underwent multiple surgery with fentanyl and sevoflurane (MSFS) exposures on postnatal days 6, 8, and 10, resulting in attention deficit hyperactivity disorder (ADHD)-like hyperactivity, impulsive behavior, cognitive impairment, and fine motor dysfunction in adulthood. Additionally, MSFS exposures inhibited neurogenesis in the nucleus accumbens (NAc) by reducing neural stem cells (NSCs) proliferation and differentiation into neurons and astrocytes. Pre-administration of Vitamin K2 (VK2) 30 min before each MSFS procedure significantly mitigated the behavioral impairments and restored neurogenesis. RNA-sequencing revealed that MSFS treatment induced 75 up-regulated and 140 down-regulated differentially expressed genes (DEGs) in the NAc, while VK2 pre-administration resulted in 149 up-regulated and 56 down-regulated DEGs. Among these, 32 DEGs were down-regulated by MSFS but restored by VK2 and 12 DEGs were up-regulated by MSFS but down-regulated by VK2. To identify key regulatory genes modulated by VK2, we performed protein-protein interaction analysis using CytoHubba, which revealed 10 hub genes-DLGAP5, TPX2, KIF20B, PLK1, SGO1, GTSE1, ASPM, CDCA2, BUB1B, and NUSAP1-with critical roles in cell cycle, cell division and NSCs pathways. The expression of hub genes was validated by RT-qPCR and immunofluorescence staining. These findings suggest that MSFS-induces ADHD-like behaviors, cognitive impairment, fine motor dysfunction, impaired neurogenesis and altering genes expression involved in cell cycle, cell division and NSCs pathways, which are rescued by VK2. This study presents the clinically MSFS model for investigating neurodevelopmental toxicity and highlights VK2's potential as a neuroprotective agent in pediatric involving multiple surgery with anesthesia and analgesia.
Collapse
Affiliation(s)
- Yu Hou
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuai Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qi Hou
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Runjia Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, China
| | - Xiyuan Xu
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhichao Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Linhui Ma
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qidong Liu
- School of Medicine, Tongji University, Shanghai 200331, China.
| | - Yuan Shen
- Shanghai Tongji Hospital, Tongji University, Shanghai, 200065, China.
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
2
|
Saade M, Martí E. Early spinal cord development: from neural tube formation to neurogenesis. Nat Rev Neurosci 2025; 26:195-213. [PMID: 39915695 DOI: 10.1038/s41583-025-00906-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 03/26/2025]
Abstract
As one of the simplest and most evolutionarily conserved parts of the vertebrate nervous system, the spinal cord serves as a key model for understanding the principles of nervous system construction. During embryonic development, the spinal cord originates from a population of bipotent stem cells termed neuromesodermal progenitors, which are organized within a transient embryonic structure known as the neural tube. Neural tube morphogenesis differs along its anterior-to-posterior axis: most of the neural tube (including the regions that will develop into the brain and the anterior spinal cord) forms via the bending and dorsal fusion of the neural groove, but the establishment of the posterior region of the neural tube involves de novo formation of a lumen within a solid medullary cord. The early spinal cord primordium consists of highly polarized neural progenitor cells organized into a pseudostratified epithelium. Tight regulation of the cell division modes of these progenitors drives the embryonic growth of the neural tube and initiates primary neurogenesis. A rich history of observational and functional studies across various vertebrate models has advanced our understanding of the cellular events underlying spinal cord development, and these foundational studies are beginning to inform our knowledge of human spinal cord development.
Collapse
Affiliation(s)
- Murielle Saade
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| | - Elisa Martí
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| |
Collapse
|
3
|
Gonzalez-Padilla D, Eagles NJ, Cano M, Pertea G, Jaffe AE, Maynard KR, Hancock DB, Handa JT, Martinowich K, Collado-Torres L. Molecular impact of nicotine and smoking exposure on the developing and adult mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.05.622149. [PMID: 39574597 PMCID: PMC11580964 DOI: 10.1101/2024.11.05.622149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Maternal smoking during pregnancy (MSDP) is associated with significant cognitive and behavioral effects on offspring. While neurodevelopmental outcomes have been studied for prenatal exposure to nicotine, the main psychoactive component of cigarette smoke, its contribution to MSDP effects has never been explored. Comparing the effects of these substances on molecular signaling in the prenatal and adult brain may provide insights into nicotinic and broader tobacco consequences that are developmental-stage specific or age-independent. Pregnant mice were administered nicotine or exposed to chronic cigarette smoke, and RNA-sequencing was performed on frontal cortices of postnatal day 0 pups born to these mice, as well as on frontal cortices and blood of the adult dams. We identified 1,010 and 4,165 differentially expressed genes (DEGs) in nicotine and smoking-exposed pup brains, respectively (FDR<0.05, Ns = 19 nicotine-exposed vs 23 vehicle-exposed; 46 smoking-exposed vs 49 controls). Prenatal nicotine exposure (PNE) alone was related to dopaminergic synapses and long-term synaptic depression, whereas MSDP was associated with the SNARE complex and vesicle transport. Both substances affected SMN-Sm protein complexes and postsynaptic endosomes. Analyses at the transcript, exon, and exon-exon junction levels supported gene level results and revealed additional smoking-affected processes. No DEGs at FDR<0.05 were found in adult mouse brain for any substance (12 nicotine-administered vs 11 vehicle-administered; 12 smoking-exposed vs 12 controls), nor in adult blood (12 smoking-exposed vs 12 controls), and only 3% and 6.41% of the DEGs in smoking-exposed pup brain replicated in smoking-exposed blood and human prenatal brain, respectively. Together, these results demonstrate variable but overlapping molecular effects of PNE and MSDP on the developing brain, and attenuated effects of both smoking and nicotine on adult versus fetal brain.
Collapse
|
4
|
Kiyomitsu A, Nishimura T, Hwang SJ, Ansai S, Kanemaki MT, Tanaka M, Kiyomitsu T. Ran-GTP assembles a specialized spindle structure for accurate chromosome segregation in medaka early embryos. Nat Commun 2024; 15:981. [PMID: 38302485 PMCID: PMC10834446 DOI: 10.1038/s41467-024-45251-w] [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: 07/09/2023] [Accepted: 01/19/2024] [Indexed: 02/03/2024] Open
Abstract
Despite drastic cellular changes during cleavage, a mitotic spindle assembles in each blastomere to accurately segregate duplicated chromosomes. Mechanisms of mitotic spindle assembly have been extensively studied using small somatic cells. However, mechanisms of spindle assembly in large vertebrate embryos remain little understood. Here, we establish functional assay systems in medaka (Oryzias latipes) embryos by combining CRISPR knock-in with auxin-inducible degron technology. Live imaging reveals several unexpected features of microtubule organization and centrosome positioning that achieve rapid, accurate cleavage. Importantly, Ran-GTP assembles a dense microtubule network at the metaphase spindle center that is essential for chromosome segregation in early embryos. This unique spindle structure is remodeled into a typical short, somatic-like spindle after blastula stages, when Ran-GTP becomes dispensable for chromosome segregation. We propose that despite the presence of centrosomes, the chromosome-derived Ran-GTP pathway has essential roles in functional spindle assembly in large, rapidly dividing vertebrate early embryos, similar to acentrosomal spindle assembly in oocytes.
Collapse
Affiliation(s)
- Ai Kiyomitsu
- Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0495, Japan
| | - Toshiya Nishimura
- Division of Biological Science, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
- Hokkaido University Fisheries Sciences, 3-1-1, Minato-cho, Hakodate, Hokkaido, 041-8611, Japan
| | - Shiang Jyi Hwang
- Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0495, Japan
| | - Satoshi Ansai
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, 980-8577, Japan
- Laboratory of Genome Editing Breeding, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masato T Kanemaki
- Department of Chromosome Science, National Institute of Genetics, Research Organization of Information and Systems (ROIS), and Graduate Institute for Advanced Studies, SOKENDAI, Yata 1111, Mishima, Shizuoka, 411-8540, Japan
- Department of Biological Science, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Minoru Tanaka
- Division of Biological Science, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Tomomi Kiyomitsu
- Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0495, Japan.
| |
Collapse
|
5
|
Zekušić M, Bujić Mihica M, Skoko M, Vukušić K, Risteski P, Martinčić J, Tolić IM, Bendelja K, Ramić S, Dolenec T, Vrgoč Zimić I, Puljić D, Petric Vicković I, Iveković R, Batarilo I, Prosenc Zmrzljak U, Hoffmeister A, Vučemilo T. New characterization and safety evaluation of human limbal stem cells used in clinical application: fidelity of mitotic process and mitotic spindle morphologies. Stem Cell Res Ther 2023; 14:368. [PMID: 38093301 PMCID: PMC10720168 DOI: 10.1186/s13287-023-03586-z] [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: 08/08/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Limbal stem cells (LSCs) are crucial for the regeneration of the corneal epithelium in patients with limbal stem cell deficiency (LSCD). Thus, LSCs during cultivation in vitro should be in highly homogeneous amounts, while potency and expression of stemness without tumorigenesis would be desirable. Therefore, further characterization and safety evaluation of engineered limbal grafts is required to provide safe and high-quality therapeutic applications. METHODS After in vitro expansion, LSCs undergo laboratory characterization in a single-cell suspension, cell culture, and in limbal grafts before transplantation. Using a clinically applicable protocol, the data collected on LSCs at passage 1 were summarized, including: identity (cell size, morphology); potency (yield, viability, population doubling time, colony-forming efficiency); expression of putative stem cell markers through flow cytometry, immunofluorescence, and immunohistochemistry. Then, mitotic chromosome stability and normal mitotic outcomes were explored by using live-cell imaging. Finally, impurities, bacterial endotoxins and sterility were determined. RESULTS Expression of the stemness marker p63 in single-cell suspension and in cell culture showed high values by different methods. Limbal grafts showed p63-positive cells (78.7 ± 9.4%), Ki67 proliferation (41.7 ± 15.9%), while CK3 was negative. Impurity with 3T3 feeder cells and endotoxins was minimized. We presented mitotic spindles with a length of 11.40 ± 0.54 m and a spindle width of 8.05 ± 0.55 m as new characterization in LSC culture. Additionally, live-cell imaging of LSCs (n = 873) was performed, and only a small fraction < 2.5% of aberrant interphase cells was observed; 2.12 ± 2.10% of mitotic spindles exhibited a multipolar phenotype during metaphase, and 3.84 ± 3.77% of anaphase cells had a DNA signal present within the spindle midzone, indicating a chromosome bridge or lagging chromosome phenotype. CONCLUSION This manuscript provides, for the first time, detailed characterization of the parameters of fidelity of the mitotic process and mitotic spindle morphologies of LSCs used in a direct clinical application. Our data show that p63-positive CK3-negative LSCs grown in vitro for clinical purposes undergo mitotic processes with extremely high fidelity, suggesting high karyotype stability. This finding confirms LSCs as a high-quality and safe therapy for eye regeneration in humans.
Collapse
Affiliation(s)
- Marija Zekušić
- Department of Transfusion and Regenerative Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Marina Bujić Mihica
- Department of Transfusion and Regenerative Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia.
| | - Marija Skoko
- Department of Transfusion and Regenerative Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Kruno Vukušić
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Patrik Risteski
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Jelena Martinčić
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Iva M Tolić
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Krešo Bendelja
- Center for Research and Knowledge Transfer in Biotechnology, Laboratory of Immunology, University of Zagreb, Zagreb, Croatia
| | - Snježana Ramić
- Department of Oncological Pathology and Clinical Cytology 'Ljudevit Jurak', University Hospital Center Sestre Milosrdnice, Zagreb, Croatia
| | - Tamara Dolenec
- Department of Transfusion and Regenerative Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Ivana Vrgoč Zimić
- Department of Transfusion and Regenerative Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Dominik Puljić
- Department of Transfusion and Regenerative Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Ivanka Petric Vicković
- Clinical Department of Ophthalmology, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Renata Iveković
- Clinical Department of Ophthalmology, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Ivanka Batarilo
- Department of Microbiology, Croatian Institute of Transfusion Medicine, Zagreb, Croatia
| | - Uršula Prosenc Zmrzljak
- Molecular Biology Department, BIA Separations CRO, Labena d.O.O, Ljubljana, Slovenia
- Labena d.o.o, Zagreb, Croatia
| | | | - Tiha Vučemilo
- Department of Transfusion and Regenerative Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| |
Collapse
|
6
|
Dell'Amico C, Angulo Salavarria MM, Takeo Y, Saotome I, Dell'Anno MT, Galimberti M, Pellegrino E, Cattaneo E, Louvi A, Onorati M. Microcephaly-associated protein WDR62 shuttles from the Golgi apparatus to the spindle poles in human neural progenitors. eLife 2023; 12:e81716. [PMID: 37272619 PMCID: PMC10241521 DOI: 10.7554/elife.81716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 04/17/2023] [Indexed: 06/06/2023] Open
Abstract
WDR62 is a spindle pole-associated scaffold protein with pleiotropic functions. Recessive mutations in WDR62 cause structural brain abnormalities and account for the second most common cause of autosomal recessive primary microcephaly (MCPH), indicating WDR62 as a critical hub for human brain development. Here, we investigated WDR62 function in corticogenesis through the analysis of a C-terminal truncating mutation (D955AfsX112). Using induced Pluripotent Stem Cells (iPSCs) obtained from a patient and his unaffected parent, as well as isogenic corrected lines, we generated 2D and 3D models of human neurodevelopment, including neuroepithelial stem cells, cerebro-cortical progenitors, terminally differentiated neurons, and cerebral organoids. We report that WDR62 localizes to the Golgi apparatus during interphase in cultured cells and human fetal brain tissue, and translocates to the mitotic spindle poles in a microtubule-dependent manner. Moreover, we demonstrate that WDR62 dysfunction impairs mitotic progression and results in alterations of the neurogenic trajectories of iPSC neuroderivatives. In summary, impairment of WDR62 localization and function results in severe neurodevelopmental abnormalities, thus delineating new mechanisms in the etiology of MCPH.
Collapse
Affiliation(s)
- Claudia Dell'Amico
- Department of Biology, Unit of Cell and Developmental Biology, University of PisaPisaItaly
| | | | - Yutaka Takeo
- Departments of Neurosurgery and Neuroscience, Yale School of MedicineNew HavenUnited States
| | - Ichiko Saotome
- Departments of Neurosurgery and Neuroscience, Yale School of MedicineNew HavenUnited States
| | | | - Maura Galimberti
- Dipartimento di Bioscienze, Università degli Studi di MilanoMilanItaly
- INGM, Istituto Nazionale Genetica MolecolareMilanItaly
| | - Enrica Pellegrino
- Department of Biology, Unit of Cell and Developmental Biology, University of PisaPisaItaly
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Elena Cattaneo
- Dipartimento di Bioscienze, Università degli Studi di MilanoMilanItaly
- INGM, Istituto Nazionale Genetica MolecolareMilanItaly
| | - Angeliki Louvi
- Departments of Neurosurgery and Neuroscience, Yale School of MedicineNew HavenUnited States
| | - Marco Onorati
- Department of Biology, Unit of Cell and Developmental Biology, University of PisaPisaItaly
| |
Collapse
|
7
|
Wimmer R, Baffet AD. The microtubule cytoskeleton of radial glial progenitor cells. Curr Opin Neurobiol 2023; 80:102709. [PMID: 37003105 DOI: 10.1016/j.conb.2023.102709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/14/2023] [Accepted: 02/23/2023] [Indexed: 04/01/2023]
Abstract
A high number of genetic mutations associated with cortical malformations are found in genes coding for microtubule-related factors. This has stimulated research to understand how the various microtubule-based processes are regulated to build a functional cerebral cortex. Here, we focus our review on the radial glial progenitor cells, the stem cells of the developing neocortex, summarizing research mostly performed in rodents and humans. We highlight how the centrosomal and acentrosomal microtubule networks are organized during interphase to support polarized transport and proper attachment of the apical and basal processes. We describe the molecular mechanism for interkinetic nuclear migration (INM), a microtubule-dependent oscillation of the nucleus. Finally, we describe how the mitotic spindle is built to ensure proper chromosome segregation, with a strong focus on factors mutated in microcephaly.
Collapse
Affiliation(s)
- Ryszard Wimmer
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France. https://twitter.com/RyWim
| | - Alexandre D Baffet
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France; Institut national de la santé et de la recherche médicale (INSERM), France.
| |
Collapse
|
8
|
Kaplow IM, Lawler AJ, Schäffer DE, Srinivasan C, Sestili HH, Wirthlin ME, Phan BN, Prasad K, Brown AR, Zhang X, Foley K, Genereux DP, Zoonomia Consortium, Karlsson EK, Lindblad-Toh K, Meyer WK, Pfenning AR. Relating enhancer genetic variation across mammals to complex phenotypes using machine learning. Science 2023; 380:eabm7993. [PMID: 37104615 PMCID: PMC10322212 DOI: 10.1126/science.abm7993] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/23/2023] [Indexed: 04/29/2023]
Abstract
Protein-coding differences between species often fail to explain phenotypic diversity, suggesting the involvement of genomic elements that regulate gene expression such as enhancers. Identifying associations between enhancers and phenotypes is challenging because enhancer activity can be tissue-dependent and functionally conserved despite low sequence conservation. We developed the Tissue-Aware Conservation Inference Toolkit (TACIT) to associate candidate enhancers with species' phenotypes using predictions from machine learning models trained on specific tissues. Applying TACIT to associate motor cortex and parvalbumin-positive interneuron enhancers with neurological phenotypes revealed dozens of enhancer-phenotype associations, including brain size-associated enhancers that interact with genes implicated in microcephaly or macrocephaly. TACIT provides a foundation for identifying enhancers associated with the evolution of any convergently evolved phenotype in any large group of species with aligned genomes.
Collapse
Affiliation(s)
- Irene M. Kaplow
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Alyssa J. Lawler
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Biology, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Daniel E. Schäffer
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Chaitanya Srinivasan
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Heather H. Sestili
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Morgan E. Wirthlin
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA
| | - BaDoi N. Phan
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kavya Prasad
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Ashley R. Brown
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Xiaomeng Zhang
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Kathleen Foley
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, USA
| | - Diane P. Genereux
- Broad Institute, Cambridge, MA, USA
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Elinor K. Karlsson
- Broad Institute, Cambridge, MA, USA
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kerstin Lindblad-Toh
- Broad Institute, Cambridge, MA, USA
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Wynn K. Meyer
- Department of Biological Sciences, Lehigh University, Bethlehem, PA, USA
| | - Andreas R. Pfenning
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Biology, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
9
|
Bernhard SV, Gemble S, Basto R, Storchova Z. Experimental Approaches to Generate and Isolate Human Tetraploid Cells. Methods Mol Biol 2023; 2545:391-399. [PMID: 36720824 DOI: 10.1007/978-1-0716-2561-3_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cancer cells are frequently affected by large-scale chromosome copy number changes, such as polyploidy or whole chromosome aneuploidy, and thus understanding the consequences of these changes is important for cancer research. In the past, it has been difficult to study the consequences of large-scale genomic changes, especially in pure isogenic populations. Here, we describe two methods to generate tetraploid cells induced either by cytokinesis failure or mitotic slippage. These treatments result in mixed population of diploids and tetraploids that can be analyzed directly. Alternatively, tetraploid populations can be established by single cell clone selection or by fluorescence activated cell sorting. These methods enable to analyze and compare the consequences of whole-genome doubling between the parental cell line, freshly arising tetraploid cells, and post-tetraploid aneuploid clones.
Collapse
Affiliation(s)
| | - Simon Gemble
- Institut Curie, PSL Research University, CNRS, UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Renata Basto
- Institut Curie, PSL Research University, CNRS, UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Zuzana Storchova
- Department of Molecular Genetics, Paul Ehrlich Strasse 24, Kaiserslautern, Germany.
| |
Collapse
|
10
|
Morretton J, Simon A, Herbette A, Barbazan J, Pérez‐González C, Cosson C, Mboup B, Latouche A, Popova T, Kieffer Y, Macé A, Gestraud P, Bataillon G, Becette V, Meseure D, Nicolas A, Mariani O, Vincent‐Salomon A, Stern M, Mechta‐Grigoriou F, Roman Roman S, Vignjevic DM, Rouzier R, Sastre‐Garau X, Goundiam O, Basto R. A catalog of numerical centrosome defects in epithelial ovarian cancers. EMBO Mol Med 2022; 14:e15670. [PMID: 36069081 PMCID: PMC9449595 DOI: 10.15252/emmm.202215670] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Centrosome amplification, the presence of more than two centrosomes in a cell is a common feature of most human cancer cell lines. However, little is known about centrosome numbers in human cancers and whether amplification or other numerical aberrations are frequently present. To address this question, we have analyzed a large cohort of primary human epithelial ovarian cancers (EOCs) from 100 patients. We found that rigorous quantitation of centrosome number in tumor samples was extremely challenging due to tumor heterogeneity and extensive tissue disorganization. Interestingly, even if centrosome clusters could be identified, the incidence of centrosome amplification was not comparable to what has been described in cultured cancer cells. Surprisingly, centrosome loss events where a few or many nuclei were not associated with centrosomes were clearly noticed and overall more frequent than centrosome amplification. Our findings highlight the difficulty of characterizing centrosome numbers in human tumors, while revealing a novel paradigm of centrosome number defects in EOCs.
Collapse
Affiliation(s)
- Jean‐Philippe Morretton
- Biology of Centrosomes and Genetic Instability, Institut CuriePSL Research University, CNRS UMR 144ParisFrance
| | - Anthony Simon
- Biology of Centrosomes and Genetic Instability, Institut CuriePSL Research University, CNRS UMR 144ParisFrance
| | - Aurélie Herbette
- Department of Translational Research, Institut CuriePSL UniversityParis Cedex 05France
| | - Jorge Barbazan
- Migration and Invasion Laboratory, Institut CuriePSL Research University, CNRS UMR 144ParisFrance
| | - Carlos Pérez‐González
- Migration and Invasion Laboratory, Institut CuriePSL Research University, CNRS UMR 144ParisFrance
| | - Camille Cosson
- Department of Translational Research, Institut CuriePSL UniversityParis Cedex 05France
| | - Bassirou Mboup
- Statistical Methods for Precision MedicineINSERM U900, Institut CurieSaint‐CloudFrance
| | - Aurélien Latouche
- Statistical Methods for Precision MedicineINSERM U900, Institut CurieSaint‐CloudFrance
| | - Tatiana Popova
- DNA Repair & Uveal Melanoma (D.R.U.M.), INSERM U830, Institut CuriePSL Research UniversityParis Cedex 05France
| | - Yann Kieffer
- Stress and Cancer Laboratory, INSERM U830, Institut Curie, Team Ligue Nationale Contre le CancerPSL Research UniversityParisFrance
| | - Anne‐Sophie Macé
- Cell and Tissue Imaging Facility (PICT‐IBiSA), Institut CuriePSL Research University, Centre National de la Recherche ScientifiqueParisFrance
| | - Pierre Gestraud
- Bioinformatics and Computational Systems Biology of Cancer, Mines Paristech, INSERM U900, Institut CuriePSL UniversityParis Cedex 05France
| | | | | | - Didier Meseure
- Department of PathologyInstitut CurieParis Cedex 05France
| | - André Nicolas
- Department of PathologyInstitut CurieParis Cedex 05France
| | - Odette Mariani
- Department of PathologyInstitut CurieParis Cedex 05France
- Biological Resource Center, Department of Pathology, Institut CuriePSL Research UniversityParisFrance
| | | | - Marc‐Henri Stern
- DNA Repair & Uveal Melanoma (D.R.U.M.), INSERM U830, Institut CuriePSL Research UniversityParis Cedex 05France
| | - Fatima Mechta‐Grigoriou
- Stress and Cancer Laboratory, INSERM U830, Institut Curie, Team Ligue Nationale Contre le CancerPSL Research UniversityParisFrance
| | - Sergio Roman Roman
- Department of Translational Research, Institut CuriePSL UniversityParis Cedex 05France
| | - Danijela Matic Vignjevic
- Migration and Invasion Laboratory, Institut CuriePSL Research University, CNRS UMR 144ParisFrance
| | - Roman Rouzier
- Statistical Methods for Precision MedicineINSERM U900, Institut CurieSaint‐CloudFrance
- Department of SurgeryInstitut CurieSaint‐CloudFrance
- UFR Simone Veil – SantéUniversité Versailles Saint Quentin, Université Paris SaclayMontigny le BretonneuxFrance
| | - Xavier Sastre‐Garau
- Department of PathologyInstitut CurieParis Cedex 05France
- Present address:
Laboratory of PathologyIntercommunal Hospital Center of CreteilCreteil CedexFrance
| | - Oumou Goundiam
- Department of Translational Research, Institut CuriePSL UniversityParis Cedex 05France
| | - Renata Basto
- Biology of Centrosomes and Genetic Instability, Institut CuriePSL Research University, CNRS UMR 144ParisFrance
| |
Collapse
|
11
|
Tátrai P, Gergely F. Centrosome function is critical during terminal erythroid differentiation. EMBO J 2022; 41:e108739. [PMID: 35678476 PMCID: PMC9289712 DOI: 10.15252/embj.2021108739] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 05/03/2022] [Accepted: 05/25/2022] [Indexed: 11/26/2022] Open
Abstract
Red blood cells are produced by terminal erythroid differentiation, which involves the dramatic morphological transformation of erythroblasts into enucleated reticulocytes. Microtubules are important for enucleation, but it is not known if the centrosome, a key microtubule-organizing center, is required as well. Mice lacking the conserved centrosome component, CDK5RAP2, are likely to have defective erythroid differentiation because they develop macrocytic anemia. Here, we show that fetal liver-derived, CDK5RAP2-deficient erythroid progenitors generate fewer and larger reticulocytes, hence recapitulating features of macrocytic anemia. In erythroblasts, but not in embryonic fibroblasts, loss of CDK5RAP2 or pharmacological depletion of centrosomes leads to highly aberrant spindle morphologies. Consistent with such cells exiting mitosis without chromosome segregation, tetraploidy is frequent in late-stage erythroblasts, thereby giving rise to fewer but larger reticulocytes than normal. Our results define a critical role for CDK5RAP2 and centrosomes in spindle formation specifically during blood production. We propose that disruption of centrosome and spindle function could contribute to the emergence of macrocytic anemias, for instance, due to nutritional deficiency or exposure to chemotherapy.
Collapse
Affiliation(s)
- Péter Tátrai
- Cancer Research UK Cambridge InstituteLi Ka Shing CentreUniversity of CambridgeCambridgeUK
- Present address:
Solvo BiotechnologyBudapestHungary
| | - Fanni Gergely
- Cancer Research UK Cambridge InstituteLi Ka Shing CentreUniversity of CambridgeCambridgeUK
- Department of BiochemistryUniversity of OxfordOxfordUK
| |
Collapse
|
12
|
Dang H, Martin‐Villalba A, Schiebel E. Centrosome linker protein C-Nap1 maintains stem cells in mouse testes. EMBO Rep 2022; 23:e53805. [PMID: 35599622 PMCID: PMC9253759 DOI: 10.15252/embr.202153805] [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: 08/13/2021] [Revised: 04/13/2022] [Accepted: 04/27/2022] [Indexed: 11/27/2022] Open
Abstract
The centrosome linker component C-Nap1 (encoded by CEP250) anchors filaments to centrioles that provide centrosome cohesion by connecting the two centrosomes of an interphase cell into a single microtubule organizing unit. The role of the centrosome linker during development of an animal remains enigmatic. Here, we show that male CEP250-/- mice are sterile because sperm production is abolished. Premature centrosome separation means that germ stem cells in CEP250-/- mice fail to establish an E-cadherin polarity mark and are unable to maintain the older mother centrosome on the basal site of the seminiferous tubules. This failure prompts premature stem cell differentiation in expense of germ stem cell expansion. The concomitant induction of apoptosis triggers the complete depletion of germ stem cells and consequently infertility. Our study reveals a role for centrosome cohesion in asymmetric cell division, stem cell maintenance, and fertility.
Collapse
Affiliation(s)
- Hairuo Dang
- Zentrum für Molekulare Biologie der Universität HeidelbergDeutsches Krebsforschungszentrum‐ZMBH AllianzUniversität HeidelbergHeidelbergGermany
- Heidelberg Biosciences International Graduate School (HBIGS)Universität HeidelbergHeidelbergGermany
| | - Ana Martin‐Villalba
- Deutsches Krebsforschungszentrum‐ZMBH AllianzUniversität HeidelbergHeidelbergGermany
| | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität HeidelbergDeutsches Krebsforschungszentrum‐ZMBH AllianzUniversität HeidelbergHeidelbergGermany
| |
Collapse
|
13
|
Casas Gimeno G, Paridaen JTML. The Symmetry of Neural Stem Cell and Progenitor Divisions in the Vertebrate Brain. Front Cell Dev Biol 2022; 10:885269. [PMID: 35693936 PMCID: PMC9174586 DOI: 10.3389/fcell.2022.885269] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/20/2022] [Indexed: 12/23/2022] Open
Abstract
Robust brain development requires the tight coordination between tissue growth, neuronal differentiation and stem cell maintenance. To achieve this, neural stem cells need to balance symmetric proliferative and terminal divisions with asymmetric divisions. In recent years, the unequal distribution of certain cellular components in mitosis has emerged as a key mechanism to regulate the symmetry of division, and the determination of equal and unequal sister cell fates. Examples of such components include polarity proteins, signaling components, and cellular structures such as endosomes and centrosomes. In several types of neural stem cells, these factors show specific patterns of inheritance that correlate to specific cell fates, albeit the underlying mechanism and the potential causal relationship is not always understood. Here, we review these examples of cellular neural stem and progenitor cell asymmetries and will discuss how they fit into our current understanding of neural stem cell function in neurogenesis in developing and adult brains. We will focus mainly on the vertebrate brain, though we will incorporate relevant examples from invertebrate organisms as well. In particular, we will highlight recent advances in our understanding of the complexities related cellular asymmetries in determining division mode outcomes, and how these mechanisms are spatiotemporally regulated to match the different needs for proliferation and differentiation as the brain forms.
Collapse
|
14
|
Spindle motility skews division site determination during asymmetric cell division in Physcomitrella. Nat Commun 2022; 13:2488. [PMID: 35513464 PMCID: PMC9072379 DOI: 10.1038/s41467-022-30239-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 04/21/2022] [Indexed: 01/09/2023] Open
Abstract
Asymmetric cell division (ACD) underlies the development of multicellular organisms. In animal ACD, the cell division site is determined by active spindle-positioning mechanisms. In contrast, it is considered that the division site in plants is determined prior to mitosis by the microtubule-actin belt known as the preprophase band (PPB) and that the localization of the mitotic spindle is typically static and does not govern the division plane. However, in some plant species, ACD occurs in the absence of PPB. Here, we isolate a hypomorphic mutant of the conserved microtubule-associated protein TPX2 in the moss Physcomitrium patens (Physcomitrella) and observe spindle motility during PPB-independent cell division. This defect compromises the position of the division site and produces inverted daughter cell sizes in the first ACD of gametophore (leafy shoot) development. The phenotype is rescued by restoring endogenous TPX2 function and, unexpectedly, by depolymerizing actin filaments. Thus, we identify an active spindle-positioning mechanism that, reminiscent of acentrosomal ACD in animals, involves microtubules and actin filaments, and sets the division site in plants.
Collapse
|
15
|
Kristofova M, Ori A, Wang ZQ. Multifaceted Microcephaly-Related Gene MCPH1. Cells 2022; 11:cells11020275. [PMID: 35053391 PMCID: PMC8774270 DOI: 10.3390/cells11020275] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
MCPH1, or BRIT1, is often mutated in human primary microcephaly type 1, a neurodevelopmental disorder characterized by a smaller brain size at birth, due to its dysfunction in regulating the proliferation and self-renewal of neuroprogenitor cells. In the last 20 years or so, genetic and cellular studies have identified MCPH1 as a multifaceted protein in various cellular functions, including DNA damage signaling and repair, the regulation of chromosome condensation, cell-cycle progression, centrosome activity and the metabolism. Yet, genetic and animal model studies have revealed an unpredicted essential function of MPCH1 in gonad development and tumorigenesis, although the underlying mechanism remains elusive. These studies have begun to shed light on the role of MPCH1 in controlling various pathobiological processes of the disorder. Here, we summarize the biological functions of MCPH1, and lessons learnt from cellular and mouse models of MCPH1.
Collapse
Affiliation(s)
- Martina Kristofova
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
| | - Alessandro Ori
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
| | - Zhao-Qi Wang
- Leibniz Institute on Aging—Fritz Lipmann Institute (FLI), Beutenbergstrasse 11, 07745 Jena, Germany; (M.K.); (A.O.)
- Faculty of Biological Sciences, Friedrich-Schiller University of Jena, Bachstrasse 18k, 07743 Jena, Germany
- Correspondence: ; Tel.: +49-3641-656415; Fax: +49-3641-656335
| |
Collapse
|
16
|
Wilsch-Bräuninger M, Huttner WB. Primary Cilia and Centrosomes in Neocortex Development. Front Neurosci 2021; 15:755867. [PMID: 34744618 PMCID: PMC8566538 DOI: 10.3389/fnins.2021.755867] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022] Open
Abstract
During mammalian brain development, neural stem and progenitor cells generate the neurons for the six-layered neocortex. The proliferative capacity of the different types of progenitor cells within the germinal zones of the developing neocortex is a major determinant for the number of neurons generated. Furthermore, the various modes of progenitor cell divisions, for which the orientation of the mitotic spindle of progenitor cells has a pivotal role, are a key parameter to ensure the appropriate size and proper cytoarchitecture of the neocortex. Here, we review the roles of primary cilia and centrosomes of progenitor cells in these processes during neocortical development. We specifically focus on the apical progenitor cells in the ventricular zone. In particular, we address the alternating, dual role of the mother centriole (i) as a component of one of the spindle poles during mitosis, and (ii) as the basal body of the primary cilium in interphase, which is pivotal for the fate of apical progenitor cells and their proliferative capacity. We also discuss the interactions of these organelles with the microtubule and actin cytoskeleton, and with junctional complexes. Centriolar appendages have a specific role in this interaction with the cell cortex and the plasma membrane. Another topic of this review is the specific molecular composition of the ciliary membrane and the membrane vesicle traffic to the primary cilium of apical progenitors, which underlie the ciliary signaling during neocortical development; this signaling itself, however, is not covered in depth here. We also discuss the recently emerging evidence regarding the composition and roles of primary cilia and centrosomes in basal progenitors, a class of progenitors thought to be of particular importance for neocortex expansion in development and evolution. While the tight interplay between primary cilia and centrosomes makes it difficult to allocate independent roles to either organelle, mutations in genes encoding ciliary and/or centrosome proteins indicate that both are necessary for the formation of a properly sized and functioning neocortex during development. Human neocortical malformations, like microcephaly, underpin the importance of primary cilia/centrosome-related processes in neocortical development and provide fundamental insight into the underlying mechanisms involved.
Collapse
Affiliation(s)
| | - Wieland B Huttner
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
17
|
Huang J, Liang Z, Guan C, Hua S, Jiang K. WDR62 regulates spindle dynamics as an adaptor protein between TPX2/Aurora A and katanin. J Cell Biol 2021; 220:212395. [PMID: 34137789 PMCID: PMC8240853 DOI: 10.1083/jcb.202007167] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 04/12/2021] [Accepted: 05/18/2021] [Indexed: 12/21/2022] Open
Abstract
WDR62 is a microcephaly-related, microtubule (MT)-associated protein (MAP) that localizes to the spindle pole and regulates spindle organization, but the underlying mechanisms remain elusive. Here, we show that WDR62 regulates spindle dynamics by recruiting katanin to the spindle pole and further reveal a TPX2–Aurora A–WDR62–katanin axis in cells. By combining cellular and in vitro experiments, we demonstrate that WDR62 shows preference for curved segments of dynamic GDP-MTs, as well as GMPCPP- and paclitaxel-stabilized MTs, suggesting that it recognizes extended MT lattice. Consistent with this property, WDR62 alone is inefficient in recruiting katanin to GDP-MTs, while WDR62 complexed with TPX2/Aurora A can potently promote katanin-mediated severing of GDP-MTs in vitro. In addition, the MT-binding affinity of WDR62 is autoinhibited through JNK phosphorylation-induced intramolecular interaction. We propose that WDR62 is an atypical MAP and functions as an adaptor protein between its recruiting factor TPX2/Aurora A and the effector katanin to orchestrate the regulation of spindle dynamics.
Collapse
Affiliation(s)
- Junjie Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Zhuobi Liang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Cuirong Guan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Shasha Hua
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Kai Jiang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
18
|
Guerreiro A, De Sousa F, Liaudet N, Ivanova D, Eskat A, Meraldi P. WDR62 localizes katanin at spindle poles to ensure synchronous chromosome segregation. J Cell Biol 2021; 220:212394. [PMID: 34137788 PMCID: PMC8240857 DOI: 10.1083/jcb.202007171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 04/12/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
Mutations in the WDR62 gene cause primary microcephaly, a pathological condition often associated with defective cell division that results in severe brain developmental defects. The precise function and localization of WDR62 within the mitotic spindle is, however, still under debate, as it has been proposed to act either at centrosomes or on the mitotic spindle. Here we explored the cellular functions of WDR62 in human epithelial cell lines using both short-term siRNA protein depletions and long-term CRISPR/Cas9 gene knockouts. We demonstrate that WDR62 localizes at spindle poles, promoting the recruitment of the microtubule-severing enzyme katanin. Depletion or loss of WDR62 stabilizes spindle microtubules due to insufficient microtubule minus-end depolymerization but does not affect plus-end microtubule dynamics. During chromosome segregation, WDR62 and katanin promote efficient poleward microtubule flux and favor the synchronicity of poleward movements in anaphase to prevent lagging chromosomes. We speculate that these lagging chromosomes might be linked to developmental defects in primary microcephaly.
Collapse
Affiliation(s)
- Amanda Guerreiro
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Filipe De Sousa
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Radiation Oncology Division, Geneva University Hospitals, Geneva, Switzerland
| | - Nicolas Liaudet
- Bioimaging Facility, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Daria Ivanova
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Anja Eskat
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Translational Research Centre in Onco-hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
19
|
Polverino F, Naso FD, Asteriti IA, Palmerini V, Singh D, Valente D, Bird AW, Rosa A, Mapelli M, Guarguaglini G. The Aurora-A/TPX2 Axis Directs Spindle Orientation in Adherent Human Cells by Regulating NuMA and Microtubule Stability. Curr Biol 2020; 31:658-667.e5. [PMID: 33275894 DOI: 10.1016/j.cub.2020.10.096] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/16/2020] [Accepted: 10/30/2020] [Indexed: 12/17/2022]
Abstract
Mitotic spindle orientation is a crucial process that defines the axis of cell division, contributing to daughter cell positioning and fate, and hence to tissue morphogenesis and homeostasis.1,2 The trimeric NuMA/LGN/Gαi complex, the major determinant of spindle orientation, exerts pulling forces on the spindle poles by anchoring astral microtubules (MTs) and dynein motors to the cell cortex.3,4 Mitotic kinases contribute to correct spindle orientation by regulating nuclear mitotic apparatus protein (NuMA) localization,5-7 among which the Aurora-A centrosomal kinase regulates NuMA targeting to the cell cortex in metaphase.8,9 Aurora-A and its activator targeting protein for Xklp2 (TPX2) are frequently overexpressed in cancer,10-12 raising the question as to whether spindle orientation is among the processes downstream the Aurora-A/TPX2 signaling axis altered under pathological conditions. Here, we investigated the role of TPX2 in the Aurora-A- and NuMA-dependent spindle orientation. We show that, in cultured adherent human cells, the interaction with TPX2 is required for Aurora-A to exert this function. We also show that Aurora-A, TPX2, and NuMA are part of a complex at spindle MTs, where TPX2 acts as a platform for Aurora-A regulation of NuMA. Interestingly, excess TPX2 does not influence NuMA localization but induces a "super-alignment" of the spindle axis with respect to the substrate, although an excess of Aurora-A induces spindle misorientation. These opposite effects are both linked to altered MT stability. Overall, our results highlight the importance of TPX2 for spindle orientation and suggest that spindle orientation is differentially sensitive to unbalanced levels of Aurora-A, TPX2, or the Aurora-A/TPX2 complex.
Collapse
Affiliation(s)
- Federica Polverino
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy; Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Adamello 16, 20141 Milan, Italy
| | - Francesco D Naso
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy
| | - Italia A Asteriti
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy
| | - Valentina Palmerini
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Adamello 16, 20141 Milan, Italy
| | - Divya Singh
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227 Dortmund, Germany
| | - Davide Valente
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy
| | - Alexander W Bird
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227 Dortmund, Germany
| | - Alessandro Rosa
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; Department of Biology and Biotechnology "C. Darwin," Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Marina Mapelli
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Via Adamello 16, 20141 Milan, Italy.
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, National Research Council of Italy, c/o Department of Biology and Biotechnology, Sapienza University of Rome, Via degli Apuli 4, 00185 Rome, Italy.
| |
Collapse
|
20
|
Goundiam O, Basto R. Centrosomes in disease: how the same music can sound so different? Curr Opin Struct Biol 2020; 66:74-82. [PMID: 33186811 DOI: 10.1016/j.sbi.2020.09.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/15/2022]
Abstract
Centrosomes are the major microtubule organizing center of animal cells. Centrosomes contribute to timely bipolar spindle assembly during mitosis and participate in the regulation of other processes such as polarity establishment and cell migration. Centrosome numbers are tightly controlled during the cell cycle to ensure that mitosis is initiated with only two centrosomes. Deviations in centrosome number or structure are known to impact cell or tissue homeostasis and can impact different processes as diverse as proliferation, death or disease. Interestingly, defects in centrosome number seem to culminate with common responses, which depend on p53 activation even in different contexts such as development or cancer. p53 is a tumor suppressor gene with essential roles in the maintenance of genetic stability normally stimulated by various cellular stresses. Here, we review current knowledge and discuss how defects in centrosome structure and number can lead to different human pathologies.
Collapse
Affiliation(s)
- Oumou Goundiam
- Department of Translational Research, Institut Curie, PSL University, 26 rue d' Ulm, F-75248 Paris Cedex 05, France
| | - Renata Basto
- Biology of Centrosomes and Genetic Instability Lab, CNRS, Institut Curie, PSL Research University, UMR144, 12 rue Lhomond, 75005 Paris, France.
| |
Collapse
|
21
|
Chippalkatti R, Egger B, Suter B. Mms19 promotes spindle microtubule assembly in Drosophila neural stem cells. PLoS Genet 2020; 16:e1008913. [PMID: 33211700 PMCID: PMC7714366 DOI: 10.1371/journal.pgen.1008913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/03/2020] [Accepted: 10/13/2020] [Indexed: 01/27/2023] Open
Abstract
Mitotic divisions depend on the timely assembly and proper orientation of the mitotic spindle. Malfunctioning of these processes can considerably delay mitosis, thereby compromising tissue growth and homeostasis, and leading to chromosomal instability. Loss of functional Mms19 drastically affects the growth and development of mitotic tissues in Drosophila larvae and we now demonstrate that Mms19 is an important factor that promotes spindle and astral microtubule (MT) growth, and MT stability and bundling. Mms19 function is needed for the coordination of mitotic events and for the rapid progression through mitosis that is characteristic of neural stem cells. Surprisingly, Mms19 performs its mitotic activities through two different pathways. By stimulating the mitotic kinase cascade, it triggers the localization of the MT regulatory complex TACC/Msps (Transforming Acidic Coiled Coil/Minispindles, the homolog of human ch-TOG) to the centrosome. This activity of Mms19 can be rescued by stimulating the mitotic kinase cascade. However, other aspects of the Mms19 phenotypes cannot be rescued in this way, pointing to an additional mechanism of Mms19 action. We provide evidence that Mms19 binds directly to MTs and that this stimulates MT stability and bundling.
Collapse
Affiliation(s)
- Rohan Chippalkatti
- Cell Biology, University of Bern, Berne, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Berne, Switzerland
| | - Boris Egger
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Beat Suter
- Cell Biology, University of Bern, Berne, Switzerland
| |
Collapse
|
22
|
Guilloux G, Gibeaux R. Mechanisms of spindle assembly and size control. Biol Cell 2020; 112:369-382. [PMID: 32762076 DOI: 10.1111/boc.202000065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 01/09/2023]
Abstract
The spindle is crucial for cell division by allowing the faithful segregation of replicated chromosomes to daughter cells. Proper segregation is ensured only if microtubules (MTs) and hundreds of other associated factors interact to assemble this complex structure with the appropriate architecture and size. In this review, we describe the latest view of spindle organisation as well as the molecular gradients and mechanisms underlying MT nucleation and spindle assembly. We then discuss the overlapping physical and molecular constraints that dictate spindle morphology, concluding with a focus on spindle size regulation.
Collapse
Affiliation(s)
- Gabriel Guilloux
- Univ Rennes, CNRS, IGDR [(Institute of Genetics and Development of Rennes)] - UMR 6290, F-35000 Rennes, France
| | - Romain Gibeaux
- Univ Rennes, CNRS, IGDR [(Institute of Genetics and Development of Rennes)] - UMR 6290, F-35000 Rennes, France
| |
Collapse
|
23
|
Stracker TH, Morrison CG, Gergely F. Molecular causes of primary microcephaly and related diseases: a report from the UNIA Workshop. Chromosoma 2020; 129:115-120. [PMID: 32424716 DOI: 10.1007/s00412-020-00737-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/30/2022]
Abstract
The International University of Andalucía (UNIA) Current Trends in Biomedicine Workshop on Molecular Causes of Primary Microcephaly and Related Diseases took place in Baeza, Spain, November 18-20, 2019. This meeting brought together scientists from Europe, the USA and China to discuss recent advances in our molecular and genetic understanding of a group of rare neurodevelopmental diseases characterised by primary microcephaly, a condition in which head circumference is smaller than normal at birth. Microcephaly can be caused by inherited mutations that affect key cellular processes, or environmental exposure to radiation or other toxins. It can also result from viral infection, as exemplified by the recent Zika virus outbreak in South America. Here we summarise a number of the scientific advances presented and topics discussed at the meeting.
Collapse
Affiliation(s)
- Travis H Stracker
- Institute for Research in Biomedicine (IRB Barcelona) and Barcelona Institute of Science and Technology, 08028, Barcelona, Spain.
| | - Ciaran G Morrison
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, Biosciences Building, Dangan, Galway, H91 TK33, Ireland
| | - Fanni Gergely
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| |
Collapse
|
24
|
Prc1-rich kinetochores are required for error-free acentrosomal spindle bipolarization during meiosis I in mouse oocytes. Nat Commun 2020; 11:2652. [PMID: 32461611 PMCID: PMC7253481 DOI: 10.1038/s41467-020-16488-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 05/01/2020] [Indexed: 12/18/2022] Open
Abstract
Acentrosomal meiosis in oocytes represents a gametogenic challenge, requiring spindle bipolarization without predefined bipolar cues. While much is known about the structures that promote acentrosomal microtubule nucleation, less is known about the structures that mediate spindle bipolarization in mammalian oocytes. Here, we show that in mouse oocytes, kinetochores are required for spindle bipolarization in meiosis I. This process is promoted by oocyte-specific, microtubule-independent enrichment of the antiparallel microtubule crosslinker Prc1 at kinetochores via the Ndc80 complex. In contrast, in meiosis II, cytoplasm that contains upregulated factors including Prc1 supports kinetochore-independent pathways for spindle bipolarization. The kinetochore-dependent mode of spindle bipolarization is required for meiosis I to prevent chromosome segregation errors. Human oocytes, where spindle bipolarization is reportedly error prone, exhibit no detectable kinetochore enrichment of Prc1. This study reveals an oocyte-specific function of kinetochores in acentrosomal spindle bipolarization in mice, and provides insights into the error-prone nature of human oocytes. Oocyte meiosis must achieve spindle bipolarization without predefined spatial cues. Yoshida et al. demonstrate that spindle bipolarization during meiosis I in mouse oocytes requires kinetochores to prevent chromosome segregation errors, a phenomenon that does not occur in error-prone human oocytes.
Collapse
|
25
|
Ou MY, Ju XC, Cai YJ, Sun XY, Wang JF, Fu XQ, Sun Q, Luo ZG. Heterogeneous nuclear ribonucleoprotein A3 controls mitotic progression of neural progenitors via interaction with cohesin. Development 2020; 147:dev185132. [PMID: 32321712 DOI: 10.1242/dev.185132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 04/03/2020] [Indexed: 01/13/2023]
Abstract
Cortex development is controlled by temporal patterning of neural progenitor (NP) competence with sequential generation of deep and superficial layer neurons, but underlying mechanisms remain elusive. Here, we report a role for heterogeneous nuclear ribonucleoprotein A3 (HNRNPA3) in regulating the division of early cortical NPs that mainly give rise to deep-layer neurons via direct neurogenesis. HNRNPA3 is expressed at high levels in NPs of mouse and human cortex at early stages, with a unique peri-chromosome pattern. Intriguingly, downregulation of HNRNPA3 caused chromosome disarrangement, which hindered normal separation of chromosomes during NP division, leading to mitotic delay. Furthermore, HNRNPA3 is associated with the cohesin-core subunit SMC1A and controls its association with chromosomes, implicating a mechanism for the role of HNRNPA3 in regulating chromosome segregation in dividing NPs. Hnrnpa3-deficient mice exhibited reduced cortical thickness, especially of deep layers. Moreover, downregulation of HNRNPA3 in cultured human cerebral organoids led to marked reduction in NPs and deep-layer neurons. Thus, this study has identified a crucial role for HNRNPA3 in NP division and highlighted the relationship between mitosis progression and early neurogenesis.
Collapse
Affiliation(s)
- Min-Yi Ou
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang-Chun Ju
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yi-Jun Cai
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xin-Yao Sun
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun-Feng Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiu-Qing Fu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qiang Sun
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhen-Ge Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
26
|
Goupil A, Nano M, Letort G, Gemble S, Edwards F, Goundiam O, Gogendeau D, Pennetier C, Basto R. Chromosomes function as a barrier to mitotic spindle bipolarity in polyploid cells. J Cell Biol 2020; 219:133854. [PMID: 32328633 PMCID: PMC7147111 DOI: 10.1083/jcb.201908006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/13/2019] [Accepted: 01/24/2020] [Indexed: 01/22/2023] Open
Abstract
Ploidy variations such as genome doubling are frequent in human tumors and have been associated with genetic instability favoring tumor progression. How polyploid cells deal with increased centrosome numbers and DNA content remains unknown. Using Drosophila neuroblasts and human cancer cells to study mitotic spindle assembly in polyploid cells, we found that most polyploid cells divide in a multipolar manner. We show that even if an initial centrosome clustering step can occur at mitotic entry, the establishment of kinetochore-microtubule attachments leads to spatial chromosome configurations, whereby the final coalescence of supernumerary poles into a bipolar array is inhibited. Using in silico approaches and various spindle and DNA perturbations, we show that chromosomes act as a physical barrier blocking spindle pole coalescence and bipolarity. Importantly, microtubule stabilization suppressed multipolarity by improving both centrosome clustering and pole coalescence. This work identifies inhibitors of bipolar division in polyploid cells and provides a rationale to understand chromosome instability typical of polyploid cancer cells.
Collapse
Affiliation(s)
- Alix Goupil
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Maddalena Nano
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Gaëlle Letort
- Center for Interdisciplinary Research in Biology, Collège de France, UMR7241/U1050, Paris, France
| | - Simon Gemble
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Frances Edwards
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Oumou Goundiam
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France.,Department of Translational Research, Institut Curie, PSL University, Paris, France
| | - Delphine Gogendeau
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Carole Pennetier
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| | - Renata Basto
- Institut Curie, Paris Science et Lettres Research University, Centre National de la Recherche Scientifique, Unité Mixte de Recherche UMR144, Biology of Centrosomes and Genetic Instability Laboratory, Paris, France
| |
Collapse
|
27
|
Marthiens V, Basto R. Centrosomes: The good and the bad for brain development. Biol Cell 2020; 112:153-172. [PMID: 32170757 DOI: 10.1111/boc.201900090] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/25/2020] [Accepted: 03/01/2020] [Indexed: 12/15/2022]
Abstract
Centrosomes nucleate and organise the microtubule cytoskeleton in animal cells. These membraneless organelles are key structures for tissue organisation, polarity and growth. Centrosome dysfunction, defined as deviation in centrosome numbers and/or structural integrity, has major impact on brain size and functionality, as compared with other tissues of the organism. In this review, we discuss the contribution of centrosomes to brain growth during development. We discuss in particular the impact of centrosome dysfunction in Drosophila and mammalian neural stem cell division and fitness, which ultimately underlie brain growth defects.
Collapse
Affiliation(s)
- Véronique Marthiens
- Biology of Centrosomes and Genetic Instability Laboratory, Institut Curie, PSL Research University, CNRS, UMR144, Paris, 75005, France
| | - Renata Basto
- Biology of Centrosomes and Genetic Instability Laboratory, Institut Curie, PSL Research University, CNRS, UMR144, Paris, 75005, France
| |
Collapse
|
28
|
Johnson CA, Ghashghaei HT. Sp2 regulates late neurogenic but not early expansive divisions of neural stem cells underlying population growth in the mouse cortex. Development 2020; 147:dev186056. [PMID: 32001437 PMCID: PMC7044455 DOI: 10.1242/dev.186056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/23/2020] [Indexed: 12/18/2022]
Abstract
Cellular and molecular mechanisms underlying the switch from self-amplification of cortical stem cells to neuronal and glial generation are incompletely understood, despite their importance for neural development. Here, we have investigated the role of the transcription factor specificity protein 2 (Sp2) in expansive and neurogenic divisions of the developing cerebral cortex by combining conditional genetic deletion with the mosaic analysis with double markers (MADM) system in mice. We find that loss of Sp2 in progenitors undergoing neurogenic divisions results in prolonged mitosis due to extension of early mitotic stages. This disruption is correlated with depletion of the populations of upper layer neurons in the cortex. In contrast, early cortical neural stem cells proliferate and expand normally in the absence of Sp2. These results indicate a stage-specific requirement for Sp2 in neural stem and progenitor cells, and reveal mechanistic differences between the early expansive and later neurogenic periods of cortical development.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Caroline A Johnson
- Department of Molecular Biomedical Sciences College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - H Troy Ghashghaei
- Department of Molecular Biomedical Sciences College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
29
|
Nano M, Gemble S, Simon A, Pennetier C, Fraisier V, Marthiens V, Basto R. Cell-Cycle Asynchrony Generates DNA Damage at Mitotic Entry in Polyploid Cells. Curr Biol 2019; 29:3937-3945.e7. [PMID: 31708395 DOI: 10.1016/j.cub.2019.09.041] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/29/2019] [Accepted: 09/16/2019] [Indexed: 01/17/2023]
Abstract
Polyploidy arises from the gain of complete chromosome sets [1], and it is known to promote cancer genome evolution. Recent evidence suggests that a large proportion of human tumors experience whole-genome duplications (WGDs), which might favor the generation of highly abnormal karyotypes within a short time frame, rather than in a stepwise manner [2-6]. However, the molecular mechanisms linking whole-genome duplication to genetic instability remain poorly understood. Using repeated cytokinesis failure to induce polyploidization of Drosophila neural stem cells (NSCs) (also called neuroblasts [NBs]), we investigated the consequences of polyploidy in vivo. Surprisingly, we found that DNA damage is generated in a subset of nuclei of polyploid NBs during mitosis. Importantly, our observations in flies were confirmed in mouse NSCs (mNSCs) and human cancer cells after acute cytokinesis inhibition. Interestingly, DNA damage occurs in nuclei that were not ready to enter mitosis but were forced to do so when exposed to the mitotic environment of neighboring nuclei within the same cell. Additionally, we found that polyploid cells are cell-cycle asynchronous and forcing cell-cycle synchronization was sufficient to lower the levels of DNA damage generated during mitosis. Overall, this work supports a model in which DNA damage at mitotic entry can generate DNA structural abnormalities that might contribute to the onset of genetic instability.
Collapse
Affiliation(s)
- Maddalena Nano
- Biology of Centrosomes and Genetic Instability Lab, Institut Curie, PSL Research University, CNRS UMR144, 12 rue Lhomond, 75005 Paris, France.
| | - Simon Gemble
- Biology of Centrosomes and Genetic Instability Lab, Institut Curie, PSL Research University, CNRS UMR144, 12 rue Lhomond, 75005 Paris, France
| | - Anthony Simon
- Biology of Centrosomes and Genetic Instability Lab, Institut Curie, PSL Research University, CNRS UMR144, 12 rue Lhomond, 75005 Paris, France
| | - Carole Pennetier
- Biology of Centrosomes and Genetic Instability Lab, Institut Curie, PSL Research University, CNRS UMR144, 12 rue Lhomond, 75005 Paris, France
| | - Vincent Fraisier
- Plateforme Imagerie PICT-IBiSA, Institut Curie, PSL Research University, CNRS UMR144, 12 rue Lhomond, 75005 Paris, France
| | - Veronique Marthiens
- Biology of Centrosomes and Genetic Instability Lab, Institut Curie, PSL Research University, CNRS UMR144, 12 rue Lhomond, 75005 Paris, France
| | - Renata Basto
- Biology of Centrosomes and Genetic Instability Lab, Institut Curie, PSL Research University, CNRS UMR144, 12 rue Lhomond, 75005 Paris, France.
| |
Collapse
|
30
|
Centromere Dysfunction Compromises Mitotic Spindle Pole Integrity. Curr Biol 2019; 29:3072-3080.e5. [DOI: 10.1016/j.cub.2019.07.052] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 06/21/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022]
|