1
|
Piano V. Multitasking Proteins: Exploring Noncanonical Functions of Proteins during Mitosis. Biochemistry 2025; 64:2123-2137. [PMID: 40315343 DOI: 10.1021/acs.biochem.5c00083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
This review provides a comprehensive overview of how mitotic cells drive the repurposing of proteins to fulfill mitosis-specific functions. To ensure the successful completion of cell division, the cell strategically reallocates its "workforce" by assigning additional functions to available proteins. Protein repurposing occurs at multiple levels of cellular organization and involves diverse mechanisms. At the protein level, proteins may gain mitosis-specific functions through post-translational modifications. At the structural level, proteins that typically maintain cellular architecture in interphase are co-opted to participate in mitotic spindle formation, chromosome condensation, and kinetochore assembly. Furthermore, the dynamic reorganization of the nuclear envelope and other organelles relies on the temporary reassignment of enzymes, structural proteins, and motor proteins to facilitate these changes. These adaptive mechanisms underscore the remarkable versatility of the cellular proteome in responding to the stringent requirements of mitosis. By leveraging the existing proteome for dual or multiple specialized roles, cells optimize resource usage while maintaining the precision needed to preserve genomic integrity and ensure the survival of the next generation of cells.
Collapse
Affiliation(s)
- Valentina Piano
- Institute of Human Genetics, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital, University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
2
|
Cunningham CE, Vizeacoumar FS, Zhang Y, Kyrylenko L, Both S, Maranda V, Dong H, Price JDW, Gao P, Wagner K, Wu Y, Lazell-Wright M, Ganapathysamy A, Hari R, Bhanumathy KK, Denomy C, Saxena A, Vizeacoumar JP, Morales AM, Khan F, Mosley S, Chen A, Katrii T, Zoller BGE, Rajamanickam K, Walke P, Gong L, Patel H, Elhasasna H, Dahiya R, Abuhussein O, Dmitriev A, Freywald T, Munhoz EP, Ruppin E, Lee JS, Rox K, Koebel M, Hopkins L, Lee CH, Yadav S, Gasparoni G, Walter J, Krishnan A, Datla R, Toosi B, Baker K, Meens J, Cescon DW, Ailles L, Leary SC, Wu Y, Empting M, Kiemer AK, Freywald A, Vizeacoumar FJ. Identification of targetable vulnerabilities of PLK1-overexpressing cancers by synthetic dosage lethality. CELL GENOMICS 2025:100876. [PMID: 40347943 DOI: 10.1016/j.xgen.2025.100876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/12/2025] [Accepted: 04/14/2025] [Indexed: 05/14/2025]
Abstract
Chromosomal instability (CIN) drives tumor heterogeneity, complicating cancer therapy. Although Polo-like kinase 1 (PLK1) overexpression induces CIN, direct inhibition of PLK1 has shown limited clinical benefits. We therefore performed a genome-wide synthetic dosage lethality (SDL) screen to identify effective alternative targets and validated over 100 candidates using in vivo and in vitro secondary CRISPR screens. We employed direct-capture Perturb-seq to assess the transcriptional consequences and viability of each SDL perturbation at a single-cell resolution. This revealed IGF2BP2 as a critical genetic dependency that, when targeted, downregulated PLK1 and significantly restricted tumor growth. Mechanistic analyses showed that IGF2BP2 loss disrupted cellular energy metabolism and mitochondrial ATP production by downregulating PLK1 levels as well as genes associated with oxidative phosphorylation. Consistent with this, pharmacological inhibition of IGF2BP2 severely impacts the viability of PLK1-overexpressing cancer cells addicted to higher metabolic rates. Our work offers a novel therapeutic strategy against PLK1-driven heterogeneous malignancies.
Collapse
Affiliation(s)
- Chelsea E Cunningham
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Frederick S Vizeacoumar
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Yue Zhang
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Liliia Kyrylenko
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Simon Both
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, PharmaScienceHub, 66123 Saarbrücken, Germany
| | - Vincent Maranda
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - He Dong
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Jared D W Price
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Peng Gao
- Global Institute for Food Security, University of Saskatchewan, Saskatoon, SK S7N 4L8, Canada; Agriculture and Agri-Food Canada, Saskatoon Research and Development Centre, 107 Science Place, Saskatoon, SK S7N 0X2, Canada
| | - Konrad Wagner
- Antiviral & Antivirulence Drugs (AVID), Helmholtz Institute for Pharmaceutical Research, Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Yingwen Wu
- Antiviral & Antivirulence Drugs (AVID), Helmholtz Institute for Pharmaceutical Research, Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Mary Lazell-Wright
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | | | - Rithik Hari
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Kalpana K Bhanumathy
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Connor Denomy
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Anjali Saxena
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Jeff P Vizeacoumar
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Alain Morejon Morales
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Faizaan Khan
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Shayla Mosley
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Angie Chen
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Tetiana Katrii
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Ben G E Zoller
- Antiviral & Antivirulence Drugs (AVID), Helmholtz Institute for Pharmaceutical Research, Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Karthic Rajamanickam
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Prachi Walke
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St., Saskatoon, SK S7K 0M7, Canada
| | - Lihui Gong
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Hardikkumar Patel
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Hussain Elhasasna
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Renuka Dahiya
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Omar Abuhussein
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Anton Dmitriev
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Tanya Freywald
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Erika Prando Munhoz
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Eytan Ruppin
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Center for Bioinformatics and Computational Biology and Department of Computer Sciences, University of Maryland, College Park, MD 20742, USA
| | - Joo Sang Lee
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Center for Bioinformatics and Computational Biology and Department of Computer Sciences, University of Maryland, College Park, MD 20742, USA; Department of Precision Medicine, School of Medicine and Department of Artificial Intelligence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Katharina Rox
- Department of Chemical Biology (CBIO), Helmholtz Center for Infection Research (HZI), Inhoffenstrasse 7, 38124 Braunschweig, Germany
| | - Martin Koebel
- Department of Pathology, University of Calgary, Calgary, AB, Canada
| | - Laura Hopkins
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Cheng Han Lee
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Sunil Yadav
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Gilles Gasparoni
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Jörn Walter
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Anand Krishnan
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, and Cameco MS Neuroscience Research Centre, 701 Queen St., Saskatoon, SK S7K 0M7, Canada
| | - Raju Datla
- Global Institute for Food Security, University of Saskatchewan, Saskatoon, SK S7N 4L8, Canada
| | - Behzad Toosi
- Western College of Veterinary Medicine, University of Saskatchewan, Room 2343, 52 Campus Drive, Saskatoon S7N 5B4, Canada
| | - Kristi Baker
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Jalna Meens
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - David W Cescon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Laurie Ailles
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Scot C Leary
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Yuliang Wu
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Martin Empting
- Antiviral & Antivirulence Drugs (AVID), Helmholtz Institute for Pharmaceutical Research, Saarland (HIPS), Helmholtz Centre for Infection Research (HZI) and Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany.
| | - Alexandra K Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, PharmaScienceHub, 66123 Saarbrücken, Germany; Center for Gender-Specific Biology and Medicine (CGBM), 66421 Homburg, Germany.
| | - Andrew Freywald
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada.
| | - Franco J Vizeacoumar
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; Cancer Research, Saskatchewan Cancer Agency, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada.
| |
Collapse
|
3
|
Erickson B, Fedoryshchak R, Fong N, Sheridan R, Larson KY, Saviola AJ, Mouilleron S, Hansen KC, Treisman R, Bentley DL. PP1/PNUTS phosphatase binds the restrictor complex and stimulates RNA Pol II transcription termination. Cell Rep 2025; 44:115564. [PMID: 40244850 DOI: 10.1016/j.celrep.2025.115564] [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: 12/26/2024] [Revised: 02/18/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
The restrictor ZC3H4/WDR82 terminates antisense transcription from bidirectional promoters, but its mechanism is poorly understood. We report that ZC3H4/WDR82 immunoprecipitates with PP1 phosphatase and its nuclear targeting subunit, PP1 phosphatase nuclear targeting subunit (PNUTS), which binds to WDR82. AlphaFold predicts a complex of PP1/PNUTS with the restrictor where both PNUTS and ZC3H4 contact WDR82. A substrate trap, PP1H66K-PNUTS, comprising inactive PP1 fused to the PNUTS C terminus, antagonizes restrictor-mediated termination, whereas PP1WT-PNUTS has less of an effect, suggesting that phosphatase activity is required for termination. One PP1/PNUTS substrate implicated in termination by the restrictor is RNA polymerase II (RNA Pol II) CTD Ser5-P. PP1H66K-PNUTS induces Ser5-P hyperphosphorylation at 5' ends, presumably by inhibiting dephosphorylation. NET-seq analysis suggests that CTD Ser5 dephosphorylation would promote termination by increasing RNA Pol II pausing. Both inhibition of termination and CTD hyperphosphorylation require the WDR82 binding domain of PP1H66K-PNUTS, which mediates restrictor binding. In summary, the PP1/PNUTS phosphatase associated with the restrictor via WDR82 promotes efficient transcription termination.
Collapse
Affiliation(s)
- Benjamin Erickson
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, PO Box 6511, Aurora, CO 80045, USA
| | | | - Nova Fong
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, PO Box 6511, Aurora, CO 80045, USA
| | - Ryan Sheridan
- RNA Bioscience Initiative, Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, PO Box 6511, Aurora, CO 80045, USA
| | - Keira Y Larson
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, PO Box 6511, Aurora, CO 80045, USA
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, PO Box 6511, Aurora, CO 80045, USA
| | | | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, PO Box 6511, Aurora, CO 80045, USA
| | | | - David L Bentley
- RNA Bioscience Initiative, Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, PO Box 6511, Aurora, CO 80045, USA.
| |
Collapse
|
4
|
Ji M, Cui W, Feng Q, Qi J, Wang X, Zhu H, Zhang W, Fu W. NME7 maintains primary cilium assembly, ciliary microtubule stability, and Hedgehog signaling. Life Sci Alliance 2025; 8:e202402933. [PMID: 39824631 PMCID: PMC11742093 DOI: 10.26508/lsa.202402933] [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: 07/08/2024] [Revised: 01/05/2025] [Accepted: 01/06/2025] [Indexed: 01/20/2025] Open
Abstract
NME7 (nucleoside diphosphate kinase 7), a lesser studied member of the non-metastatic expressed (NME) family, has been reported as a potential subunit of the γ-tubulin ring complex (γTuRC). However, its role in the cilium assembly and function remains unclear. Our research demonstrated that NME7 is located at the centrosome, including at the spindle poles during metaphase and at the basal bodies during cilium assembly. Notably, a small fraction of NME7 localizes within the cilium. Detailed analysis of cilium assembly after NME7 knockdown and knockout revealed that NME7 is required for this process. NME7 knockout cells exhibited sensitivity to nocodazole, indicating its role in ciliary microtubule stability. In addition, NME7 deficiency impacted the Hedgehog signaling pathway, evident from reduced smoothened (Smo) fluorescence within primary cilia. This role of NME7 in Hedgehog signaling may depend on its nucleoside diphosphate kinase activity and γTuRC association. In conclusion, these findings enhance our understanding of the γTuRC roles in primary cilia in mammalian cells, highlighting the importance of NME7 in ciliary functions and signaling pathways.
Collapse
Affiliation(s)
- Menghui Ji
- Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Wenjuan Cui
- Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Qian Feng
- Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Jingjin Qi
- Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Xinmin Wang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Hong Zhu
- Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Wenqing Zhang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Wenxiang Fu
- Yunnan Key Laboratory of Cell Metabolism and Diseases, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| |
Collapse
|
5
|
Gao Q, Hofer FW, Filbeck S, Vermeulen BJA, Würtz M, Neuner A, Kaplan C, Zezlina M, Sala C, Shin H, Gruss OJ, Schiebel E, Pfeffer S. Structural mechanisms for centrosomal recruitment and organization of the microtubule nucleator γ-TuRC. Nat Commun 2025; 16:2453. [PMID: 40074789 PMCID: PMC11903878 DOI: 10.1038/s41467-025-57729-2] [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: 06/22/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
The γ-tubulin ring complex (γ-TuRC) acts as a structural template for microtubule formation at centrosomes, associating with two main compartments: the pericentriolar material and the centriole lumen. In the pericentriolar material, the γ-TuRC is involved in microtubule organization, while the function of the centriole lumenal pool remains unclear. The conformational landscape of the γ-TuRC, which is crucial for its activity, and its centrosomal anchoring mechanisms, which determine γ-TuRC activity and turnover, are not understood. Using cryo-electron tomography, we analyze γ-TuRCs in human cells and purified centrosomes. Pericentriolar γ-TuRCs simultaneously associate with the essential adapter NEDD1 and the microcephaly protein CDK5RAP2. NEDD1 forms a tetrameric structure at the γ-TuRC base through interactions with four GCP3/MZT1 modules and GCP5/6-specific extensions, while multiple copies of CDK5RAP2 engage the γ-TuRC in two distinct binding patterns to promote γ-TuRC closure and activation. In the centriole lumen, the microtubule branching factor Augmin tethers a condensed cluster of γ-TuRCs to the centriole wall with defined directional orientation. Centriole-lumenal γ-TuRC-Augmin is protected from degradation during interphase and released in mitosis to aid chromosome alignment. This study provides a unique view on γ-TuRC structure and molecular organization at centrosomes and identifies an important cellular function of centriole-lumenal γ-TuRCs.
Collapse
Affiliation(s)
- Qi Gao
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Florian W Hofer
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Sebastian Filbeck
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Bram J A Vermeulen
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Martin Würtz
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Annett Neuner
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | | | - Maja Zezlina
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Cornelia Sala
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Hyesu Shin
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | | | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany.
| | - Stefan Pfeffer
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany.
| |
Collapse
|
6
|
Li L, Zhang Z, Wang X, Zhao H, Liu L, Xiao Y, Hua S, Chen Y. PRMT5 Maintains Homeostasis of the Intestinal Epithelium by Modulating Cell Proliferation and Survival. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415559. [PMID: 39899687 PMCID: PMC11948081 DOI: 10.1002/advs.202415559] [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: 11/23/2024] [Indexed: 02/05/2025]
Abstract
Intestinal homeostasis is sustained by self-renewal of intestinal stem cells, which continuously divide and produce proliferative transit-amplifying (TA) and progenitor cells. Protein arginine methyltransferases 5 (PRMT5) plays a crucial role in regulating homeostasis of various mammalian tissues. However, its function in intestinal homeostasis remains elusive. In this study, conditional knockout of Prmt5 in the mouse intestinal epithelium leads to a reduction in stem cell population, suppression of cell proliferation, and increased cell apoptosis within the intestinal crypts, accompanied with shortened gut length, decreased mouse body weight, and eventual animal mortality. Additionally, Prmt5 deletion or its enzymatic inhibition in intestinal organoids in vitro also shows resembling cellular phenotypes. Methylome profiling identifies 90 potential Prmt5 substrates, which are involved in RNA-related biological processes and cell division. Consistently, Prmt5 depletion in intestinal organoids leads to aberrant alternative splicing in a subset of genes related to the mitotic cell cycle. Furthermore, Prmt5 loss triggers p53-mediated apoptosis in the intestinal epithelium. Collectively, the findings uncover an indispensable role of PRMT5 in promoting cell proliferation and survival, as well as maintaining stem cells in the gut epithelium.
Collapse
Affiliation(s)
- Leilei Li
- Guangzhou LaboratoryGuangzhou510700China
| | - Zhe Zhang
- Guangzhou LaboratoryGuangzhou510700China
| | - Xu Wang
- Guangzhou LaboratoryGuangzhou510700China
| | | | | | | | - Shan Hua
- Guangzhou LaboratoryGuangzhou510700China
| | - Ye‐Guang Chen
- Guangzhou LaboratoryGuangzhou510700China
- The State Key Laboratory of Membrane BiologyTsinghua‐Peking Center for Life SciencesSchool of Life SciencesTsinghua UniversityBeijing100084China
- School of Basic MedicineJiangxi Medical CollegeNanchang UniversityNanchang330031China
| |
Collapse
|
7
|
Li Z, Zhang W, Zhang Z, Mao G, Qi L, Wang Y, Yang H, Ye H. PICH, A protein that maintains genomic stability, can promote tumor growth. Gene 2025; 935:149074. [PMID: 39491600 DOI: 10.1016/j.gene.2024.149074] [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: 07/26/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Genomic instability is regardedas a hallmark of cancer cells. It can be presented in many ways, among which chromosome instability has received attention. Ultrafine anaphase bridges are a typeof chromatin bridges, the untimely resolution of which can also lead to chromosome instability. PICH can play a role in maintaining chromosome stability by regulating chromosome morphologyand resolving ultrafine anaphase bridges. Recently, PICH has been found to be overexpressed in various cancers. Overexpression of PICH is related to the proliferation of tumors and poor prognosis. In this article, we consider that PICH can maintain genomic stability by regulating appropriate chromosome structure, ensuring proper chromosome segregation, and facilitating replication fork reversal. We summarize how PICH regulates chromosome stability, how PICH resolves Ultrafine anaphase bridges with other proteins, and how PICH promotes tumor progression.
Collapse
Affiliation(s)
- Zeyuan Li
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Wentao Zhang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Zihan Zhang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Guoming Mao
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Linping Qi
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Yubin Wang
- Laboratory Medicine Center Gansu Provincial Natural Science, Lanzhou University Second Hospital, People's Republic of China
| | - Hanteng Yang
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, People's Republic of China.
| | - Huili Ye
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China; Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, People's Republic of China.
| |
Collapse
|
8
|
Zheng A, Vermeulen BJA, Würtz M, Neuner A, Lübbehusen N, Mayer MP, Schiebel E, Pfeffer S. Structural insights into the interplay between microtubule polymerases, γ-tubulin complexes and their receptors. Nat Commun 2025; 16:402. [PMID: 39757296 DOI: 10.1038/s41467-024-55778-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 12/24/2024] [Indexed: 01/07/2025] Open
Abstract
The γ-tubulin ring complex (γ-TuRC) is a structural template for controlled nucleation of microtubules from α/β-tubulin heterodimers. At the cytoplasmic side of the yeast spindle pole body, the CM1-containing receptor protein Spc72 promotes γ-TuRC assembly from seven γ-tubulin small complexes (γ-TuSCs) and recruits the microtubule polymerase Stu2, yet their molecular interplay remains unclear. Here, we determine the cryo-EM structure of the Candida albicans cytoplasmic nucleation unit at 3.6 Å resolution, revealing how the γ-TuRC is assembled and conformationally primed for microtubule nucleation by the dimerised Spc72 CM1 motif. Two coiled-coil regions of Spc72 interact with the conserved C-terminal α-helix of Stu2 and thereby position the α/β-tubulin-binding TOG domains of Stu2 in the vicinity of the microtubule assembly site. Collectively, we reveal the function of CM1 motifs in γ-TuSC oligomerisation and the recruitment of microtubule polymerases to the γ-TuRC.
Collapse
Affiliation(s)
- Anjun Zheng
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Bram J A Vermeulen
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Martin Würtz
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Heidelberg Meyerhofstraße 1, Heidelberg, Germany
| | - Annett Neuner
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Nicole Lübbehusen
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Matthias P Mayer
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany
| | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany.
| | - Stefan Pfeffer
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), Heidelberg, Germany.
| |
Collapse
|
9
|
Xu Y, Muñoz-Hernández H, Krutyhołowa R, Marxer F, Cetin F, Wieczorek M. Partial closure of the γ-tubulin ring complex by CDK5RAP2 activates microtubule nucleation. Dev Cell 2024; 59:3161-3174.e15. [PMID: 39321808 DOI: 10.1016/j.devcel.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 06/04/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
Microtubule nucleation is templated by the γ-tubulin ring complex (γ-TuRC), but its structure deviates from the geometry of α-/β-tubulin in the microtubule, explaining the complex's poor nucleating activity. Several proteins may activate the γ-TuRC, but the mechanisms underlying activation are not known. Here, we determined the structure of the porcine γ-TuRC purified using CDK5RAP2's centrosomin motif 1 (CM1). We identified an unexpected conformation of the γ-TuRC bound to multiple protein modules containing MZT2, GCP2, and CDK5RAP2, resulting in a long-range constriction of the γ-tubulin ring that brings it in closer agreement with the 13-protofilament microtubule. Additional CDK5RAP2 promoted γ-TuRC decoration and stimulated the microtubule-nucleating activities of the porcine γ-TuRC and a reconstituted, CM1-free human complex in single-molecule assays. Our results provide a structural mechanism for the control of microtubule nucleation by CM1 proteins and identify conformational transitions in the γ-TuRC that prime it for microtubule nucleation.
Collapse
Affiliation(s)
- Yixin Xu
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zürich, 8093 Zürich, Switzerland
| | - Hugo Muñoz-Hernández
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zürich, 8093 Zürich, Switzerland
| | - Rościsław Krutyhołowa
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zürich, 8093 Zürich, Switzerland
| | - Florina Marxer
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zürich, 8093 Zürich, Switzerland
| | - Ferdane Cetin
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zürich, 8093 Zürich, Switzerland
| | - Michal Wieczorek
- Department of Biology, Institute of Molecular Biology and Biophysics, ETH Zürich, 8093 Zürich, Switzerland.
| |
Collapse
|
10
|
Sun C, Fan E, Huang L, Zhang Z. Performance of radiomics in preoperative determination of malignant potential and Ki-67 expression levels in gastrointestinal stromal tumors: a systematic review and meta-analysis. Acta Radiol 2024; 65:1307-1318. [PMID: 39411915 DOI: 10.1177/02841851241285958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2024]
Abstract
Empirical evidence for radiomics predicting the malignant potential and Ki-67 expression in gastrointestinal stromal tumors (GISTs) is lacking. The aim of this review article was to explore the preoperative discriminative performance of radiomics in assessing the malignant potential, mitotic index, and Ki-67 expression levels of GISTs. We systematically searched PubMed, EMBASE, Web of Science, and the Cochrane Library. The search was conducted up to 30 September 2023. Quality assessment was performed using the Radiomics Quality Score (RQS). A total of 35 original studies were included in the analysis. Among them, 26 studies focused on determining malignant potential, three studies on mitotic index discrimination, and six studies on Ki-67 discrimination. In the validation set, the sensitivity, specificity, and area under the receiver operating characteristic curve (AUC) of radiomics in the determination of high malignant potential were 0.74 (95% CI=0.69-0.78), 0.90 (95% CI=0.83-0.94), and 0.81 (95% CI=0.14-0.99), respectively. For moderately to highly malignant potential, the sensitivity, specificity, and AUC were 0.86 (95% CI=0.83-0.88), 0.73 (95% CI=0.67-0.78), and 0.88 (95% CI=0.27-0.99), respectively. Regarding the determination of high mitotic index, the sensitivity, specificity, and AUC of radiomics were 0.86 (95% CI=0.83-0.88), 0.73 (95% CI=0.67-0.78), and 0.88 (95% CI=0.27-0.99), respectively. When determining high Ki-67 expression, the combined sensitivity, specificity, and AUC were 0.74 (95% CI=0.65-0.81), 0.81 (95% CI=0.74-0.86), and 0.84 (95% CI=0.61-0.95), respectively. Radiomics demonstrates promising discriminative performance in the preoperative assessment of malignant potential, mitotic index, and Ki-67 expression levels in GISTs.
Collapse
Affiliation(s)
- Chengyu Sun
- Department of Colorectal Surgery, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Enguo Fan
- State Key Laboratory of Medical Molecular Biology, Department of Microbiology and Parasitology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, PR China
| | - Luqiao Huang
- Department of Colorectal Surgery, Xuzhou Central Hospital, Xuzhou, Jiangsu, PR China
| | - Zhengguo Zhang
- Department of Colorectal Surgery, Xuzhou Central Hospital, Xuzhou, Jiangsu, PR China
| |
Collapse
|
11
|
Cao L, Jing X, Liu L, Wang H, Zhu J, Zhou J, Peng H, Zhang J, Li F, Wang X, Zhao L. MZT2A serves as a prognostic biomarker and promotes the progression of kidney renal clear cell carcinoma. Heliyon 2024; 10:e35695. [PMID: 39170571 PMCID: PMC11336887 DOI: 10.1016/j.heliyon.2024.e35695] [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: 04/17/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
MZT2A is a novel core component in the γ-tubulin ring complex and aberrantly expressed in some types of tumors. However, MZT2A expression pattern across different cancers and its role in kidney renal clear cell carcinoma have not been sufficiently investigated. A thorough analysis of MZT2A expression landscape across 33 cancer types was conducted, utilizing 712 normal samples and 9807 tumor samples from TCGA (version 37.0), as well as 5112 normal samples from the GTEx databases. MZT2A's impact on KIRC cell viability and proliferation were evaluated through BrdU assays and monitored by cell imaging readers in MZT2A-expressing plasmid or siRNA-transfected cells. Additionally, the effects of MZT2A on cell apoptosis and cell cycle were detected using flow cytometry and Western blot. WGCNA analysis was employed to construct a co-expression gene network associated with MZT2A expression in KIRC, and Pearson correlation coefficient was utilized to examine the relationships between pairs of genes. MZT2A is overexpressed in 25 out of 33 types of cancer, including KIRC. In KIRC, high level of MZT2A was associated with higher clinical stage progression and poorer patients' survival. Downregulation of MZT2A decreased KIRC cell proliferation, retarded cell cycle and promoted apoptosis, while upregulation of MZT2A produced the opposite effects. WGCNA analysis and in vitro experiments revealed that MZT2A activated PI3K/AKT signaling pathway in KIRC. In all, MZT2A was overexpressed in most types of tumors. MZT2A served as an oncogene in KIRC and might be a potential target for guiding future treatments.
Collapse
Affiliation(s)
- Li Cao
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Xintao Jing
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Lijuan Liu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Hui Wang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Jiaqi Zhu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Jing Zhou
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Hang Peng
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
- Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, 710068, PR China
| | - Jinyuan Zhang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Fang Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Xiaofei Wang
- Biomedical Experimental Center of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Le Zhao
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| |
Collapse
|
12
|
Shu Y, Pang X, Li H, Deng C. A multidimensional analysis of ZW10 interacting kinetochore protein in human tumors. Am J Cancer Res 2024; 14:390-402. [PMID: 38323280 PMCID: PMC10839319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 01/25/2024] [Indexed: 02/08/2024] Open
Abstract
ZW10 interacting kinetochore protein (ZWINT), an essential part of the kinetochore complex, plays a crucial role in maintaining genome stability by correcting improper attachments between the kinetochore and microtubules. An initial analysis of The Cancer Genome Atlas and Gene Expression Omnibus databases revealed that ZWINT is significantly expressed across a diverse range of tumor types. We subsequently investigated the influence of ZWINT on clinical outcomes and potential signaling pathways. A multidimensional analysis of ZWINT revealed significant statistical associations between ZWINT expression and clinical outcomes, as well as the E2F1 oncogenic signature. Experimental validation confirmed the increased expression of ZWINT in both pancreatic cancer cell lines and pancreatic adenocarcinoma tissues. Furthermore, our findings indicate that ZWINT promotes the proliferation of PANC-1 cells through cell cycle regulation. This comprehensive analysis of ZWINT suggests a strong correlation between its expression and various types of tumors, especially pancreatic adenocarcinoma (PAAD), indicating its potential oncogenic role. These findings enhance our understanding of the function of ZWINT in carcinogenesis.
Collapse
Affiliation(s)
- Yufeng Shu
- Department of Gastroenterology, Third Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Xiaoyang Pang
- Department of Spinal Surgery, Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Huan Li
- Department of Gastroenterology, Third Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Chao Deng
- Department of Orthopedics, Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| |
Collapse
|
13
|
Desgraupes S, Etienne L, Arhel NJ. RANBP2 evolution and human disease. FEBS Lett 2023; 597:2519-2533. [PMID: 37795679 DOI: 10.1002/1873-3468.14749] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
Ran-binding protein 2 (RANBP2)/Nup358 is a nucleoporin and a key component of the nuclear pore complex. Through its multiple functions (e.g., SUMOylation, regulation of nucleocytoplasmic transport) and subcellular localizations (e.g., at the nuclear envelope, kinetochores, annulate lamellae), it is involved in many cellular processes. RANBP2 dysregulation or mutation leads to the development of human pathologies, such as acute necrotizing encephalopathy 1, cancer, neurodegenerative diseases, and it is also involved in viral infections. The chromosomal region containing the RANBP2 gene is highly dynamic, with high structural variation and recombination events that led to the appearance of a gene family called RANBP2 and GCC2 Protein Domains (RGPD), with multiple gene loss/duplication events during ape evolution. Although RGPD homoplasy and maintenance during evolution suggest they might confer an advantage to their hosts, their functions are still unknown and understudied. In this review, we discuss the appearance and importance of RANBP2 in metazoans and its function-related pathologies, caused by an alteration of its expression levels (through promotor activity, post-transcriptional, or post-translational modifications), its localization, or genetic mutations.
Collapse
Affiliation(s)
- Sophie Desgraupes
- Institut de Recherche en Infectiologie de Montpellier (IRIM), University of Montpellier, France
| | - Lucie Etienne
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, UCBL1, CNRS UMR 5308, ENS de Lyon, Université de Lyon, France
| | - Nathalie J Arhel
- Institut de Recherche en Infectiologie de Montpellier (IRIM), University of Montpellier, France
| |
Collapse
|
14
|
Kalbfuss N, Berger A, Gönczy P. Mapping of centriolar proteins onto the post-embryonic lineage of C. elegans. Dev Biol 2023; 502:68-76. [PMID: 37414202 DOI: 10.1016/j.ydbio.2023.07.001] [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: 04/25/2023] [Revised: 06/22/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Centrioles, together with the surrounding peri-centriolar material (PCM), constitute the centrosome, a major microtubule-organizing center of animal cells. Despite being critical in many cells for signaling, motility and division, centrioles can be eliminated in some systems, including in the vast majority of differentiating cells during embryogenesis in Caenorhabditis elegans. Whether the cells retaining centrioles in the resulting L1 larvae do so because they lack an activity that eliminates centrioles in the other cells is not known. Moreover, the extent to which centrioles and PCM remain present in later stages of worm development, when all cells but those of the germ line are terminally differentiated, is not known. Here, by fusing cells that lack centrioles with cells that retain them, we established that L1 larvae do not possess a diffusible elimination activity sufficient to remove centrioles. Moreover, analyzing PCM core proteins in L1 larval cells that retain centrioles, we found that some such proteins, but not all, are present as well. Furthermore, we uncovered that foci of centriolar proteins remain present in specific terminally differentiated cells of adult hermaphrodites and males, in particular in the somatic gonad. Correlating the time at which cells were born with the fate of their centrioles revealed that it is not cell age, but instead cell fate, that determines whether and when centrioles are eliminated. Overall, our work maps the localization of centriolar and PCM core proteins in the post-embryonic C. elegans lineage, thereby providing an essential blueprint for uncovering mechanisms modulating their presence and function.
Collapse
Affiliation(s)
- Nils Kalbfuss
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Antonin Berger
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, CH-1015, Switzerland.
| |
Collapse
|
15
|
Wu X, Li Z, Wang ZQ, Xu X. The neurological and non-neurological roles of the primary microcephaly-associated protein ASPM. Front Neurosci 2023; 17:1242448. [PMID: 37599996 PMCID: PMC10436222 DOI: 10.3389/fnins.2023.1242448] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Primary microcephaly (MCPH), is a neurological disorder characterized by small brain size that results in numerous developmental problems, including intellectual disability, motor and speech delays, and seizures. Hitherto, over 30 MCPH causing genes (MCPHs) have been identified. Among these MCPHs, MCPH5, which encodes abnormal spindle-like microcephaly-associated protein (ASPM), is the most frequently mutated gene. ASPM regulates mitotic events, cell proliferation, replication stress response, DNA repair, and tumorigenesis. Moreover, using a data mining approach, we have confirmed that high levels of expression of ASPM correlate with poor prognosis in several types of tumors. Here, we summarize the neurological and non-neurological functions of ASPM and provide insight into its implications for the diagnosis and treatment of MCPH and cancer.
Collapse
Affiliation(s)
- Xingxuan Wu
- Guangdong Key Laboratory for Genome Stability and Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, China
- Shenzhen University-Friedrich Schiller Universität Jena Joint PhD Program in Biomedical Sciences, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
- Laboratory of Genome Stability, Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Zheng Li
- Guangdong Key Laboratory for Genome Stability and Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Zhao-Qi Wang
- Shenzhen University-Friedrich Schiller Universität Jena Joint PhD Program in Biomedical Sciences, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
- Laboratory of Genome Stability, Leibniz Institute on Aging-Fritz Lipmann Institute, Jena, Germany
| | - Xingzhi Xu
- Guangdong Key Laboratory for Genome Stability and Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, China
- Shenzhen University-Friedrich Schiller Universität Jena Joint PhD Program in Biomedical Sciences, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
16
|
Gallisà-Suñé N, Sànchez-Fernàndez-de-Landa P, Zimmermann F, Serna M, Regué L, Paz J, Llorca O, Lüders J, Roig J. BICD2 phosphorylation regulates dynein function and centrosome separation in G2 and M. Nat Commun 2023; 14:2434. [PMID: 37105961 PMCID: PMC10140047 DOI: 10.1038/s41467-023-38116-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
The activity of dynein is regulated by a number of adaptors that mediate its interaction with dynactin, effectively activating the motor complex while also connecting it to different cargos. The regulation of adaptors is consequently central to dynein physiology but remains largely unexplored. We now describe that one of the best-known dynein adaptors, BICD2, is effectively activated through phosphorylation. In G2, phosphorylation of BICD2 by CDK1 promotes its interaction with PLK1. In turn, PLK1 phosphorylation of a single residue in the N-terminus of BICD2 results in a structural change that facilitates the interaction with dynein and dynactin, allowing the formation of active motor complexes. Moreover, modified BICD2 preferentially interacts with the nucleoporin RanBP2 once RanBP2 has been phosphorylated by CDK1. BICD2 phosphorylation is central for dynein recruitment to the nuclear envelope, centrosome tethering to the nucleus and centrosome separation in the G2 and M phases of the cell cycle. This work reveals adaptor activation through phosphorylation as crucial for the spatiotemporal regulation of dynein activity.
Collapse
Affiliation(s)
- Núria Gallisà-Suñé
- Department of Cells and Tissues, Molecular Biology Institute of Barcelona (IBMB-CSIC), Baldiri i Reixac 10-12, 08028, Barcelona, Spain
| | - Paula Sànchez-Fernàndez-de-Landa
- Department of Cells and Tissues, Molecular Biology Institute of Barcelona (IBMB-CSIC), Baldiri i Reixac 10-12, 08028, Barcelona, Spain
- Aging and Metabolism Programme, IRB Barcelona, Barcelona, Spain
| | - Fabian Zimmermann
- Mechanisms of Disease Programme, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028, Barcelona, Spain
| | - Marina Serna
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, E-28029, Madrid, Spain
| | - Laura Regué
- Department of Cells and Tissues, Molecular Biology Institute of Barcelona (IBMB-CSIC), Baldiri i Reixac 10-12, 08028, Barcelona, Spain
| | - Joel Paz
- Mechanisms of Disease Programme, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028, Barcelona, Spain
| | - Oscar Llorca
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, E-28029, Madrid, Spain
| | - Jens Lüders
- Mechanisms of Disease Programme, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028, Barcelona, Spain
| | - Joan Roig
- Department of Cells and Tissues, Molecular Biology Institute of Barcelona (IBMB-CSIC), Baldiri i Reixac 10-12, 08028, Barcelona, Spain.
| |
Collapse
|
17
|
Mackels L, Liu X, Bonne G, Servais L. TOR1AIP1-Associated Nuclear Envelopathies. Int J Mol Sci 2023; 24:ijms24086911. [PMID: 37108075 PMCID: PMC10138496 DOI: 10.3390/ijms24086911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Human TOR1AIP1 encodes LAP1, a nuclear envelope protein expressed in most human tissues, which has been linked to various biological processes and human diseases. The clinical spectrum of diseases related to mutations in TOR1AIP1 is broad, including muscular dystrophy, congenital myasthenic syndrome, cardiomyopathy, and multisystemic disease with or without progeroid features. Although rare, these recessively inherited disorders often lead to early death or considerable functional impairment. Developing a better understanding of the roles of LAP1 and mutant TOR1AIP1-associated phenotypes is paramount to allow therapeutic development. To facilitate further studies, this review provides an overview of the known interactions of LAP1 and summarizes the evidence for the function of this protein in human health. We then review the mutations in the TOR1AIP1 gene and the clinical and pathological characteristics of subjects with these mutations. Lastly, we discuss challenges to be addressed in the future.
Collapse
Affiliation(s)
- Laurane Mackels
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK
- Adult Neurology Department, Citadelle Hospital, 4000 Liège, Belgium
| | - Xincheng Liu
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK
| | - Gisèle Bonne
- Sorbonne University, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Laurent Servais
- Neuromuscular Center, Division of Paediatrics, University Hospital of Liège, University of Liège, 4000 Liège, Belgium
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
18
|
Zhan X, Liu Y, Jannu AJ, Huang S, Ye B, Wei W, Pandya PH, Ye X, Pollok KE, Renbarger JL, Huang K, Zhang J. Identify potential driver genes for PAX-FOXO1 fusion-negative rhabdomyosarcoma through frequent gene co-expression network mining. Front Oncol 2023; 13:1080989. [PMID: 36793601 PMCID: PMC9924292 DOI: 10.3389/fonc.2023.1080989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/12/2023] [Indexed: 02/03/2023] Open
Abstract
Background Rhabdomyosarcoma (RMS) is a soft tissue sarcoma usually originated from skeletal muscle. Currently, RMS classification based on PAX-FOXO1 fusion is widely adopted. However, compared to relatively clear understanding of the tumorigenesis in the fusion-positive RMS, little is known for that in fusion-negative RMS (FN-RMS). Methods We explored the molecular mechanisms and the driver genes of FN-RMS through frequent gene co-expression network mining (fGCN), differential copy number (CN) and differential expression analyses on multiple RMS transcriptomic datasets. Results We obtained 50 fGCN modules, among which five are differentially expressed between different fusion status. A closer look showed 23% of Module 2 genes are concentrated on several cytobands of chromosome 8. Upstream regulators such as MYC, YAP1, TWIST1 were identified for the fGCN modules. Using in a separate dataset we confirmed that, comparing to FP-RMS, 59 Module 2 genes show consistent CN amplification and mRNA overexpression, among which 28 are on the identified chr8 cytobands. Such CN amplification and nearby MYC (also resides on one of the above cytobands) and other upstream regulators (YAP1, TWIST1) may work together to drive FN-RMS tumorigenesis and progression. Up to 43.1% downstream targets of Yap1 and 45.8% of the targets of Myc are differentially expressed in FN-RMS vs. normal comparisons, which also confirmed the driving force of these regulators. Discussion We discovered that copy number amplification of specific cytobands on chr8 and the upstream regulators MYC, YAP1 and TWIST1 work together to affect the downstream gene co-expression and promote FN-RMS tumorigenesis and progression. Our findings provide new insights for FN-RMS tumorigenesis and offer promising targets for precision therapy. Experimental investigation about the functions of identified potential drivers in FN-RMS are in progress.
Collapse
Affiliation(s)
- Xiaohui Zhan
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Yusong Liu
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China
| | - Asha Jacob Jannu
- Department of Biostatistics and Health Data Science, Indiana University, School of Medicine, Indianapolis, IN, United States
| | | | - Bo Ye
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Wei Wei
- Department of Bioinformatics, School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Pankita H Pandya
- Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, IN, United States
| | - Xiufen Ye
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China
| | - Karen E Pollok
- Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, IN, United States
| | - Jamie L Renbarger
- Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, IN, United States
| | - Kun Huang
- Department of Biostatistics and Health Data Science, Indiana University, School of Medicine, Indianapolis, IN, United States
| | - Jie Zhang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
19
|
Liu Y, Ilinski A, Gerstenfeld LC, Bragdon B. Prx1 cell subpopulations identified in various tissues with diverse quiescence and activation ability following fracture and BMP2 stimulation. Front Physiol 2023; 14:1106474. [PMID: 36793419 PMCID: PMC9922707 DOI: 10.3389/fphys.2023.1106474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/18/2023] [Indexed: 01/31/2023] Open
Abstract
The expression of Prx1 has been used as a marker to define the skeletal stem cells (SSCs) populations found within the bone marrow and periosteum that contribute to bone regeneration. However, Prx1 expressing SSCs (Prx1-SSCs) are not restricted to the bone compartments, but are also located within the muscle and able to contribute to ectopic bone formation. Little is known however, about the mechanism(s) regulating Prx1-SSCs that reside in muscle and how they participate in bone regeneration. This study compared both the intrinsic and extrinsic factors of the periosteum and muscle derived Prx1-SSCs and analyzed their regulatory mechanisms of activation, proliferation, and skeletal differentiation. There was considerable transcriptomic heterogeneity in the Prx1-SSCs found in muscle or the periosteum however in vitro cells from both tissues showed tri-lineage (adipose, cartilage and bone) differentiation. At homeostasis, periosteal-derived Prx1 cells were proliferative and low levels of BMP2 were able to promote their differentiation, while the muscle-derived Prx1 cells were quiescent and refractory to comparable levels of BMP2 that promoted periosteal cell differentiation. The transplantation of Prx1-SCC from muscle and periosteum into either the same site from which they were isolated, or their reciprocal sites showed that periosteal cell transplanted onto the surface of bone tissues differentiated into bone and cartilage cells but was incapable of similar differentiation when transplanted into muscle. Prx1-SSCs from the muscle showed no ability to differentiate at either site of transplantation. Both fracture and ten times the BMP2 dose was needed to promote muscle-derived cells to rapidly enter the cell cycle as well as undergo skeletal cell differentiation. This study elucidates the diversity of the Prx1-SSC population showing that cells within different tissue sites are intrinsically different. While muscle tissue must have factors that promote Prx1-SSC to remain quiescent, either bone injury or high levels of BMP2 can activate these cells to both proliferate and undergo skeletal cell differentiation. Finally, these studies raise the possibility that muscle SSCs are potential target for skeletal repair and bone diseases.
Collapse
Affiliation(s)
| | | | | | - Beth Bragdon
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
20
|
Ali A, Vineethakumari C, Lacasa C, Lüders J. Microtubule nucleation and γTuRC centrosome localization in interphase cells require ch-TOG. Nat Commun 2023; 14:289. [PMID: 36702836 PMCID: PMC9879976 DOI: 10.1038/s41467-023-35955-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
Organization of microtubule arrays requires spatio-temporal regulation of the microtubule nucleator γ-tubulin ring complex (γTuRC) at microtubule organizing centers (MTOCs). MTOC-localized adapter proteins are thought to recruit and activate γTuRC, but the molecular underpinnings remain obscure. Here we show that at interphase centrosomes, rather than adapters, the microtubule polymerase ch-TOG (also named chTOG or CKAP5) ultimately controls γTuRC recruitment and activation. ch-TOG co-assembles with γTuRC to stimulate nucleation around centrioles. In the absence of ch-TOG, γTuRC fails to localize to these sites, but not the centriole lumen. However, whereas some ch-TOG is stably bound at subdistal appendages, it only transiently associates with PCM. ch-TOG's dynamic behavior requires its tubulin-binding TOG domains and a C-terminal region involved in localization. In addition, ch-TOG also promotes nucleation from the Golgi. Thus, at interphase centrosomes stimulation of nucleation and γTuRC attachment are mechanistically coupled through transient recruitment of ch-TOG, and ch-TOG's nucleation-promoting activity is not restricted to centrosomes.
Collapse
Affiliation(s)
- Aamir Ali
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Chithran Vineethakumari
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Cristina Lacasa
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Jens Lüders
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain.
| |
Collapse
|
21
|
Berman AY, Wieczorek M, Aher A, Olinares PDB, Chait BT, Kapoor TM. A nucleotide binding-independent role for γ-tubulin in microtubule capping and cell division. J Cell Biol 2023; 222:213828. [PMID: 36695784 PMCID: PMC9930161 DOI: 10.1083/jcb.202204102] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/20/2022] [Accepted: 12/22/2022] [Indexed: 01/26/2023] Open
Abstract
The γ-tubulin ring complex (γ-TuRC) has essential roles in centrosomal and non-centrosomal microtubule organization during vertebrate mitosis. While there have been important advances in understanding γ-TuRC-dependent microtubule nucleation, γ-TuRC capping of microtubule minus-ends remains poorly characterized. Here, we utilized biochemical reconstitutions and cellular assays to characterize the human γ-TuRC's capping activity. Single filament assays showed that the γ-TuRC remained associated with a nucleated microtubule for tens of minutes. In contrast, caps at dynamic microtubule minus-ends displayed lifetimes of ∼1 min. Reconstituted γ-TuRCs with nucleotide-binding deficient γ-tubulin (γ-tubulinΔGTP) formed ring-shaped complexes that did not nucleate microtubules but capped microtubule minus-ends with lifetimes similar to those measured for wild-type complexes. In dividing cells, microtubule regrowth assays revealed that while knockdown of γ-tubulin suppressed non-centrosomal microtubule formation, add-back of γ-tubulinΔGTP could substantially restore this process. Our results suggest that γ-TuRC capping is a nucleotide-binding-independent activity that plays a role in non-centrosomal microtubule organization during cell division.
Collapse
Affiliation(s)
- Adi Y. Berman
- https://ror.org/0420db125Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA,Tri-Institutional PhD Program in Chemical Biology, The Rockefeller University, New York, NY, USA
| | - Michal Wieczorek
- https://ror.org/0420db125Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA
| | - Amol Aher
- https://ror.org/0420db125Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA
| | - Paul Dominic B. Olinares
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY, USA
| | - Brian T. Chait
- Tri-Institutional PhD Program in Chemical Biology, The Rockefeller University, New York, NY, USA,Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY, USA
| | - Tarun M. Kapoor
- https://ror.org/0420db125Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY, USA,Tri-Institutional PhD Program in Chemical Biology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
22
|
Nachmias D, Melnikov N, Zorea A, Sharon M, Yemini R, De-Picchoto Y, Tsirkas I, Aharoni A, Frohn B, Schwille P, Zarivach R, Mizrahi I, Elia N. Asgard ESCRT-III and VPS4 reveal conserved chromatin binding properties of the ESCRT machinery. THE ISME JOURNAL 2023; 17:117-129. [PMID: 36221007 PMCID: PMC9751279 DOI: 10.1038/s41396-022-01328-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
The archaeal Asgard superphylum currently stands as the most promising prokaryotic candidate, from which eukaryotic cells emerged. This unique superphylum encodes for eukaryotic signature proteins (ESP) that could shed light on the origin of eukaryotes, but the properties and function of these proteins is largely unresolved. Here, we set to understand the function of an Asgard archaeal protein family, namely the ESCRT machinery, that is conserved across all domains of life and executes basic cellular eukaryotic functions, including membrane constriction during cell division. We find that ESCRT proteins encoded in Loki archaea, express in mammalian and yeast cells, and that the Loki ESCRT-III protein, CHMP4-7, resides in the eukaryotic nucleus in both organisms. Moreover, Loki ESCRT-III proteins associated with chromatin, recruited their AAA-ATPase VPS4 counterpart to organize in discrete foci in the mammalian nucleus, and directly bind DNA. The human ESCRT-III protein, CHMP1B, exhibited similar nuclear properties and recruited both human and Asgard VPS4s to nuclear foci, indicating interspecies interactions. Mutation analysis revealed a role for the N terminal region of ESCRT-III in mediating these phenotypes in both human and Asgard ESCRTs. These findings suggest that ESCRT proteins hold chromatin binding properties that were highly preserved through the billion years of evolution separating Asgard archaea and humans. The conserved chromatin binding properties of the ESCRT membrane remodeling machinery, reported here, may have important implications for the origin of eukaryogenesis.
Collapse
Affiliation(s)
- Dikla Nachmias
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Nataly Melnikov
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Alvah Zorea
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Maya Sharon
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Reut Yemini
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Yasmin De-Picchoto
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Ioannis Tsirkas
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Amir Aharoni
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Bela Frohn
- Department of Cellular and Molecular Biophysics, Max-Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Petra Schwille
- Department of Cellular and Molecular Biophysics, Max-Planck Institute of Biochemistry, Martinsried, 82152, Germany
| | - Raz Zarivach
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- The Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Itzhak Mizrahi
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel
| | - Natalie Elia
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel.
- National Institute for Biotechnology in the Negev (NIBN), Ben-Gurion University of the Negev, Beer Sheva, 84105, Israel.
| |
Collapse
|
23
|
Tang X, Wei W, Snowball JM, Nakayasu ES, Bell SM, Ansong C, Lin X, Whitsett JA. EMC3 regulates mesenchymal cell survival via control of the mitotic spindle assembly. iScience 2022; 26:105667. [PMID: 36624844 PMCID: PMC9823123 DOI: 10.1016/j.isci.2022.105667] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 08/15/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Eukaryotic cells transit through the cell cycle to produce two daughter cells. Dysregulation of the cell cycle leads to cell death or tumorigenesis. Herein, we found a subunit of the ER membrane complex, EMC3, as a key regulator of cell cycle. Conditional deletion of Emc3 in mouse embryonic mesoderm led to reduced size and patterning defects of multiple organs. Emc3 deficiency impaired cell proliferation, causing spindle assembly defects, chromosome mis-segregation, cell cycle arrest at G2/M, and apoptosis. Upon entry into mitosis, mesenchymal cells upregulate EMC3 protein levels and localize EMC3 to the mitotic centrosomes. Further analysis indicated that EMC3 works together with VCP to tightly regulate the levels and activity of Aurora A, an essential factor for centrosome function and mitotic spindle assembly: while overexpression of EMC3 or VCP degraded Aurora A, their loss led to increased Aurora A stability but reduced Aurora A phosphorylation in mitosis.
Collapse
Affiliation(s)
- Xiaofang Tang
- Perinatal Institute, Divisions of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, MLC 7029, Cincinnati, OH 45229, USA,Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, 2nd Nanjiang Rd, Nansha District, Guangzhou 511458, China
| | - Wei Wei
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, No. 2005 Songhu Rd, Shanghai 200438, China
| | - John M. Snowball
- Perinatal Institute, Divisions of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, MLC 7029, Cincinnati, OH 45229, USA
| | - Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99354, USA
| | - Sheila M. Bell
- Perinatal Institute, Divisions of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, MLC 7029, Cincinnati, OH 45229, USA
| | - Charles Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, 902 Battelle Blvd, Richland, WA 99354, USA
| | - Xinhua Lin
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, 2nd Nanjiang Rd, Nansha District, Guangzhou 511458, China,State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, No. 2005 Songhu Rd, Shanghai 200438, China,Corresponding author
| | - Jeffrey A. Whitsett
- Perinatal Institute, Divisions of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, MLC 7029, Cincinnati, OH 45229, USA,Corresponding author
| |
Collapse
|
24
|
Wu T, Dong J, Fu J, Kuang Y, Chen B, Gu H, Luo Y, Gu R, Zhang M, Li W, Dong X, Sun X, Sang Q, Wang L. The mechanism of acentrosomal spindle assembly in human oocytes. Science 2022; 378:eabq7361. [DOI: 10.1126/science.abq7361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Meiotic spindle assembly ensures proper chromosome segregation in oocytes. However, the mechanisms behind spindle assembly in human oocytes remain largely unknown. We used three-dimensional high-resolution imaging of more than 2000 human oocytes to identify a structure that we named the human oocyte microtubule organizing center (huoMTOC). The proteins TACC3, CCP110, CKAP5, and DISC1 were found to be essential components of the huoMTOC. The huoMTOC arises beneath the oocyte cortex and migrates adjacent to the nuclear envelope before nuclear envelope breakdown (NEBD). After NEBD, the huoMTOC fragments and relocates on the kinetochores to initiate microtubule nucleation and spindle assembly. Disrupting the huoMTOC led to spindle assembly defects and oocyte maturation arrest. These results reveal a physiological mechanism of huoMTOC-regulated spindle assembly in human oocytes.
Collapse
Affiliation(s)
- Tianyu Wu
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Jie Dong
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Jing Fu
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Fudan University, Shanghai 200032, China
| | - Hao Gu
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Yuxi Luo
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Ruihuan Gu
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Meiling Zhang
- Center for Reproductive Medicine and Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Wen Li
- Center for Reproductive Medicine and Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaoxi Sun
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Qing Sang
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Lei Wang
- Institute of Pediatrics, Children’s Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| |
Collapse
|
25
|
Suresh P, Galstyan V, Phillips R, Dumont S. Modeling and mechanical perturbations reveal how spatially regulated anchorage gives rise to spatially distinct mechanics across the mammalian spindle. eLife 2022; 11:e79558. [PMID: 36346735 PMCID: PMC9642996 DOI: 10.7554/elife.79558] [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: 04/26/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022] Open
Abstract
During cell division, the spindle generates force to move chromosomes. In mammals, microtubule bundles called kinetochore-fibers (k-fibers) attach to and segregate chromosomes. To do so, k-fibers must be robustly anchored to the dynamic spindle. We previously developed microneedle manipulation to mechanically challenge k-fiber anchorage, and observed spatially distinct response features revealing the presence of heterogeneous anchorage (Suresh et al., 2020). How anchorage is precisely spatially regulated, and what forces are necessary and sufficient to recapitulate the k-fiber's response to force remain unclear. Here, we develop a coarse-grained k-fiber model and combine with manipulation experiments to infer underlying anchorage using shape analysis. By systematically testing different anchorage schemes, we find that forces solely at k-fiber ends are sufficient to recapitulate unmanipulated k-fiber shapes, but not manipulated ones for which lateral anchorage over a 3 μm length scale near chromosomes is also essential. Such anchorage robustly preserves k-fiber orientation near chromosomes while allowing pivoting around poles. Anchorage over a shorter length scale cannot robustly restrict pivoting near chromosomes, while anchorage throughout the spindle obstructs pivoting at poles. Together, this work reveals how spatially regulated anchorage gives rise to spatially distinct mechanics in the mammalian spindle, which we propose are key for function.
Collapse
Affiliation(s)
- Pooja Suresh
- Biophysics Graduate Program, University of California, San FranciscoSan FranciscoUnited States
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
| | - Vahe Galstyan
- Biochemistry and Molecular Biophysics Option, California Institute of TechnologyPasadenaUnited States
- A. Alikhanyan National Laboratory (Yerevan Physics Institute)YerevanArmenia
| | - Rob Phillips
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
- Department of Physics, California Institute of TechnologyPasadenaUnited States
- Chan Zuckerberg Biohub, San FranciscoSan FranciscoUnited States
| | - Sophie Dumont
- Biophysics Graduate Program, University of California, San FranciscoSan FranciscoUnited States
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Chan Zuckerberg Biohub, San FranciscoSan FranciscoUnited States
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
26
|
Shankar S, Hsu ZT, Ezquerra A, Li CC, Huang TL, Coyaud E, Viais R, Grauffel C, Raught B, Lim C, Lüders J, Tsai SY, Hsia KC. Α γ-tubulin complex-dependent pathway suppresses ciliogenesis by promoting cilia disassembly. Cell Rep 2022; 41:111642. [DOI: 10.1016/j.celrep.2022.111642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/30/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022] Open
|
27
|
Gabel CA, Li Z, DeMarco AG, Zhang Z, Yang J, Hall MC, Barford D, Chang L. Molecular architecture of the augmin complex. Nat Commun 2022; 13:5449. [PMID: 36114186 PMCID: PMC9481612 DOI: 10.1038/s41467-022-33227-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/05/2022] [Indexed: 12/21/2022] Open
Abstract
Accurate segregation of chromosomes during mitosis depends on the correct assembly of the mitotic spindle, a bipolar structure composed mainly of microtubules. The augmin complex, or homologous to augmin subunits (HAUS) complex, is an eight-subunit protein complex required for building robust mitotic spindles in metazoa. Augmin increases microtubule density within the spindle by recruiting the γ-tubulin ring complex (γ-TuRC) to pre-existing microtubules and nucleating branching microtubules. Here, we elucidate the molecular architecture of augmin by single particle cryo-electron microscopy (cryo-EM), computational methods, and crosslinking mass spectrometry (CLMS). Augmin's highly flexible structure contains a V-shaped head and a filamentous tail, with the head existing in either extended or contracted conformational states. Our work highlights how cryo-EM, complemented by computational advances and CLMS, can elucidate the structure of a challenging protein complex and provides insights into the function of augmin in mediating microtubule branching nucleation.
Collapse
Affiliation(s)
- Clinton A Gabel
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Zhuang Li
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Andrew G DeMarco
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Ziguo Zhang
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Jing Yang
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Mark C Hall
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - David Barford
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Leifu Chang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA.
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
28
|
Li J, Xu J, Yang T, Chen J, Li F, Shen B, Fan C. Genome-wide methylation analyses of human sperm unravel novel differentially methylated regions in asthenozoospermia. Epigenomics 2022; 14:951-964. [PMID: 36004499 DOI: 10.2217/epi-2022-0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims & objectives: To investigate DNA methylation patterns in asthenozoospermic and normozoospermic sperm and to explore the potential roles of differential methylations in the etiology of the disease. Materials & methods: The authors performed whole-genome bisulfite sequencing analysis between normozoospermic controls and asthenozoospermic individuals. Results: The authors identified 238 significant differentially methylated regions. These differentially methylated regions were annotated to 114 protein-coding genes, with many genes showing associations with spermatogenesis, sperm motility etc. Conclusion: There are plenty of genomic regions exhibiting altered DNA methylation in asthenozoospermia, a number of which are located within or adjacent to sperm-related genes, suggesting novel methylation markers of asthenozoospermia and potential epigenetic regulation mechanisms through DNA methylation in the disease.
Collapse
Affiliation(s)
- Jingjing Li
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, China
| | - Jinyan Xu
- Human Sperm Bank, Key Laboratory of Birth Defects & Related Diseases of Women & Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, 610041, China
| | - Tingting Yang
- Human Sperm Bank, Key Laboratory of Birth Defects & Related Diseases of Women & Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, 610041, China
| | - Jianhai Chen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, China
| | - Fuping Li
- Human Sperm Bank, Key Laboratory of Birth Defects & Related Diseases of Women & Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, 610041, China
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610212, China
| | - Chuanzhu Fan
- Department of Biological Sciences, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
29
|
Ali M, LaCanna R, Lian Z, Huang J, Tan Y, Shao W, Yu X, Tian Y. Transcriptional responses to injury of regenerative lung alveolar epithelium. iScience 2022; 25:104843. [PMID: 35996586 PMCID: PMC9391595 DOI: 10.1016/j.isci.2022.104843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/01/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022] Open
Abstract
The significance of alveolar epithelial type 2 (AT2) cell proliferation for lung alveolar epithelial homeostasis and regeneration after injury has been widely accepted. However, the heterogeneity of AT2 cell population for cell proliferation capacity remains disputed. By single-cell RNA sequencing and genetic lineage labeling using the Ki67 knock-in mouse model, we map all proliferative AT2 cells in homeostatic and regenerating murine lungs after injury induced by Streptococcus pneumoniae infection. The proliferative AT2 cell population displays a unique transcriptional program, which is regulated by activating transcription factor 3 (ATF3) and thyroid hormone receptor alpha (THRA) transcription factors. Overexpression of these two transcription factors in AT2 cells promoted AT2 cell proliferation and improved lung function after injury. These results indicate that increased expression of ATF3 and THRA at the onset of lung epithelial regeneration is required to permit rapid AT2 cell proliferation and hence progression through the recovery of lung epithelium.
Collapse
Affiliation(s)
- Mir Ali
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Ryan LaCanna
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Zhaorui Lian
- Coriell Institute for Medical Research, Camden, NJ 08103, USA
| | - Jian Huang
- Coriell Institute for Medical Research, Camden, NJ 08103, USA
| | - Yinfei Tan
- Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - Wenna Shao
- Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiang Yu
- Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying Tian
- Department of Cardiovascular Sciences, Center for Translational Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| |
Collapse
|
30
|
Willekers S, Tessadori F, van der Vaart B, Henning HH, Stucchi R, Altelaar M, Roelen BAJ, Akhmanova A, Bakkers J. The centriolar satellite protein Cfap53 facilitates formation of the zygotic microtubule organizing center in the zebrafish embryo. Development 2022; 149:dev198762. [PMID: 35980365 PMCID: PMC9481976 DOI: 10.1242/dev.198762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 07/20/2022] [Indexed: 12/02/2023]
Abstract
In embryos of most animal species, the zygotic centrosome is assembled by the centriole derived from the sperm cell and pericentriolar proteins present in the oocyte. This zygotic centrosome acts as a microtubule organizing center (MTOC) to assemble the sperm aster and mitotic spindle. As MTOC formation has been studied mainly in adult cells, very little is known about the formation of the zygotic MTOC. Here, we show that zebrafish (Danio rerio) embryos lacking either maternal or paternal Cfap53, a centriolar satellite protein, arrest during the first cell cycle. Although Cfap53 is dispensable for sperm aster function, it aids proper formation of the mitotic spindle. During cell division, Cfap53 colocalizes with γ-tubulin and with other centrosomal and centriolar satellite proteins at the MTOC. Furthermore, we find that γ-tubulin localization at the MTOC is impaired in the absence of Cfap53. Based on these results, we propose a model in which Cfap53 deposited in the oocyte and the sperm participates in the organization of the zygotic MTOC to allow mitotic spindle formation.
Collapse
Affiliation(s)
- Sven Willekers
- Hubrecht Institute-KNAW, Utrecht 3584 CT, The Netherlands
| | | | - Babet van der Vaart
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Heiko H. Henning
- Equine Sciences, Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CM, The Netherlands
| | - Riccardo Stucchi
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Bernard A. J. Roelen
- Embryology, Anatomy and Physiology, Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW, Utrecht 3584 CT, The Netherlands
- Department of Pediatric Cardiology, Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584 EA, The Netherlands
| |
Collapse
|
31
|
Zhang Y, Hong X, Hua S, Jiang K. Reconstitution and mechanistic dissection of the human microtubule branching machinery. J Cell Biol 2022; 221:e202109053. [PMID: 35604367 PMCID: PMC9129923 DOI: 10.1083/jcb.202109053] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 03/07/2022] [Accepted: 05/02/2022] [Indexed: 01/07/2023] Open
Abstract
Branching microtubule (MT) nucleation is mediated by the augmin complex and γ-tubulin ring complex (γ-TuRC). However, how these two complexes work together to promote this process remains elusive. Here, using purified components from native and recombinant sources, we demonstrate that human augmin and γ-TuRC are sufficient to reconstitute the minimal MT branching machinery, in which NEDD1 bridges between augmin holo complex and GCP3/MZT1 subcomplex of γ-TuRC. The single-molecule experiment suggests that oligomerization of augmin may activate the branching machinery. We provide direct biochemical evidence that CDK1- and PLK1-dependent phosphorylation are crucial for NEDD1 binding to augmin, for their synergistic MT-binding activities, and hence for branching MT nucleation. In addition, we unveil that NEDD1 possesses an unanticipated intrinsic affinity for MTs via its WD40 domain, which also plays a pivotal role in the branching process. In summary, our study provides a comprehensive understanding of the underlying mechanisms of branching MT nucleation in human cells.
Collapse
Affiliation(s)
- Yaqian Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Xing Hong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology and Key Laboratory of Oral Biomedicine Ministry of Education, School & 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 & 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 & Hospital of Stomatology, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
32
|
Adashev VE, Bazylev SS, Potashnikova DM, Godneeva BK, Shatskikh AS, Olenkina OM, Olenina LV, Kotov AA. Comparative transcriptional analysis uncovers molecular processes in early and mature somatic cyst cells of Drosophila testes. Eur J Cell Biol 2022; 101:151246. [PMID: 35667338 DOI: 10.1016/j.ejcb.2022.151246] [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: 05/31/2021] [Revised: 04/29/2022] [Accepted: 05/27/2022] [Indexed: 11/18/2022] Open
Abstract
The tight interaction between somatic and germline cells is conserved in animal spermatogenesis. The testes of Drosophila melanogaster are the model of choice to identify processes responsible for mature gamete production. However, processes of differentiation and soma-germline interactions occurring in somatic cyst cells are currently understudied. Here we focused on the comparison of transcriptome expression patterns of early and mature somatic cyst cells to find out the developmental changes taking place in them. We employed a FACS-based approach for the isolation of early and mature somatic cyst cells from fly testes, subsequent preparation of RNA-Seq libraries, and analysis of gene differential expression in the sorted cells. We found increased expression of genes involved in cell cycle-related processes in early cyst cells, which is necessary for the proliferation and self-renewal of a crucial population of early cyst cells, cyst stem cells. Genes proposedly required for lamellipodium-like projection organization for proper cyst formation were also detected among the upregulated ones in early cyst cells. Gene Ontology and interactome analyses of upregulated genes in mature cyst cells revealed a striking over-representation of gene categories responsible for metabolic and catabolic cellular processes, as well as genes supporting the energetic state of the cells provided by oxidative phosphorylation that is carried out in mitochondria. Our comparative analyses of differentially expressed genes revealed major peculiarities in early and mature cyst cells and provide novel insight into their regulation, which is important for male fertility.
Collapse
Affiliation(s)
- Vladimir E Adashev
- Institute of Molecular Genetics of National Research Center "Kurchatov Institute", 2 Kurchatov Sq., Moscow 123182, Russia.
| | - Sergei S Bazylev
- Institute of Molecular Genetics of National Research Center "Kurchatov Institute", 2 Kurchatov Sq., Moscow 123182, Russia.
| | - Daria M Potashnikova
- Lomonosov Moscow State University, School of Biology, Department of Cell Biology and Histology, Moscow 119234, Russia.
| | - Baira K Godneeva
- Institute of Molecular Genetics of National Research Center "Kurchatov Institute", 2 Kurchatov Sq., Moscow 123182, Russia.
| | - Aleksei S Shatskikh
- Institute of Molecular Genetics of National Research Center "Kurchatov Institute", 2 Kurchatov Sq., Moscow 123182, Russia.
| | - Oxana M Olenkina
- Institute of Molecular Genetics of National Research Center "Kurchatov Institute", 2 Kurchatov Sq., Moscow 123182, Russia.
| | - Ludmila V Olenina
- Institute of Molecular Genetics of National Research Center "Kurchatov Institute", 2 Kurchatov Sq., Moscow 123182, Russia.
| | - Alexei A Kotov
- Institute of Molecular Genetics of National Research Center "Kurchatov Institute", 2 Kurchatov Sq., Moscow 123182, Russia.
| |
Collapse
|
33
|
Ki67 for evaluating the prognosis of gastrointestinal stromal tumors: A systematic review and meta‑analysis. Oncol Lett 2022; 23:189. [PMID: 35527778 PMCID: PMC9073573 DOI: 10.3892/ol.2022.13309] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Overexpression of Ki67 is observed in tumor cells, and it has been suggested to be a marker for cancer prognosis. However, the relationship between Ki67 expression and the risk of recurrence of gastrointestinal stromal tumors (GISTs) remains poorly defined. In the present study, a meta-analysis was used to examine the associations between Ki67 levels and GIST recurrence. Studies reporting GIST and Ki67 were found by searching Cochrane Library, PubMed and Embase until October 14, 2021. The Newcastle-Ottawa Scale (NOS) was used to verify the quality of the evidence. Totally, 1682 patient cases were included. The odds ratio (OR) estimates and 95% confidence interval (CI) for each publication were determined by a fixed-effects (Mantel-Haenszel) model. A total of 20 studies that fulfilled the inclusion criteria were finally included in the analysis. The average score of quality evaluation was 6.4 points according to NOS. It was found that Ki67 levels were significantly higher in the NIH L group compared with the NIH VL group (OR: 0.51; 95% CI: 0.26-0.99; P=0.04; P heterogeneity=0.44). There was also greater Ki67 overexpression in the NIH I group compared with the NIH L group (OR: 0.45, 95% CI: 0.31-0.65; P<0.0001; P heterogeneity=0.32), while Ki67 levels were greater in the NIH H group than in the NIH I group (OR: 0.20; 95% CI: 0.15-0.28; P<0.00001; P heterogeneity=0.56). In conclusion, Ki67 overexpression may be a useful marker of the risk of recurrent GIST transformation.
Collapse
|
34
|
Gan P, Wang Z, Morales MG, Zhang Y, Bassel-Duby R, Liu N, Olson EN. RBPMS is an RNA-binding protein that mediates cardiomyocyte binucleation and cardiovascular development. Dev Cell 2022; 57:959-973.e7. [PMID: 35472321 PMCID: PMC9116735 DOI: 10.1016/j.devcel.2022.03.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/04/2022] [Accepted: 03/25/2022] [Indexed: 12/13/2022]
Abstract
Noncompaction cardiomyopathy is a common congenital cardiac disorder associated with abnormal ventricular cardiomyocyte trabeculation and impaired pump function. The genetic basis and underlying mechanisms of this disorder remain elusive. We show that the genetic deletion of RNA-binding protein with multiple splicing (Rbpms), an uncharacterized RNA-binding factor, causes perinatal lethality in mice due to congenital cardiovascular defects. The loss of Rbpms causes premature onset of cardiomyocyte binucleation and cell cycle arrest during development. Human iPSC-derived cardiomyocytes with RBPMS gene deletion have a similar blockade to cytokinesis. Sequencing analysis revealed that RBPMS plays a role in RNA splicing and influences RNAs involved in cytoskeletal signaling pathways. We found that RBPMS mediates the isoform switching of the heart-enriched LIM domain protein Pdlim5. The loss of Rbpms leads to an abnormal accumulation of Pdlim5-short isoforms, disrupting cardiomyocyte cytokinesis. Our findings connect premature cardiomyocyte binucleation to noncompaction cardiomyopathy and highlight the role of RBPMS in this process.
Collapse
Affiliation(s)
- Peiheng Gan
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhaoning Wang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Maria Gabriela Morales
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yu Zhang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ning Liu
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
35
|
Cho NH, Cheveralls KC, Brunner AD, Kim K, Michaelis AC, Raghavan P, Kobayashi H, Savy L, Li JY, Canaj H, Kim JY, Stewart EM, Gnann C, McCarthy F, Cabrera JP, Brunetti RM, Chhun BB, Dingle G, Hein MY, Huang B, Mehta SB, Weissman JS, Gómez-Sjöberg R, Itzhak DN, Royer LA, Mann M, Leonetti MD. OpenCell: Endogenous tagging for the cartography of human cellular organization. Science 2022; 375:eabi6983. [PMID: 35271311 PMCID: PMC9119736 DOI: 10.1126/science.abi6983] [Citation(s) in RCA: 277] [Impact Index Per Article: 92.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Elucidating the wiring diagram of the human cell is a central goal of the postgenomic era. We combined genome engineering, confocal live-cell imaging, mass spectrometry, and data science to systematically map the localization and interactions of human proteins. Our approach provides a data-driven description of the molecular and spatial networks that organize the proteome. Unsupervised clustering of these networks delineates functional communities that facilitate biological discovery. We found that remarkably precise functional information can be derived from protein localization patterns, which often contain enough information to identify molecular interactions, and that RNA binding proteins form a specific subgroup defined by unique interaction and localization properties. Paired with a fully interactive website (opencell.czbiohub.org), our work constitutes a resource for the quantitative cartography of human cellular organization.
Collapse
Affiliation(s)
| | | | - Andreas-David Brunner
- Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Kibeom Kim
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - André C. Michaelis
- Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | | | - Laura Savy
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jason Y. Li
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Hera Canaj
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | | | - Christian Gnann
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Stockholm, Sweden
| | | | | | - Rachel M. Brunetti
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | | | - Greg Dingle
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | | | - Bo Huang
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | | | - Jonathan S. Weissman
- Whitehead Institute, Koch Institute, Howard Hughes Medical Institute, and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
| | | | | | | | - Matthias Mann
- Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
- NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
36
|
Gräf R, Grafe M, Meyer I, Mitic K, Pitzen V. The Dictyostelium Centrosome. Cells 2021; 10:cells10102657. [PMID: 34685637 PMCID: PMC8534566 DOI: 10.3390/cells10102657] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 12/13/2022] Open
Abstract
The centrosome of Dictyostelium amoebae contains no centrioles and consists of a cylindrical layered core structure surrounded by a corona harboring microtubule-nucleating γ-tubulin complexes. It is the major centrosomal model beyond animals and yeasts. Proteomics, protein interaction studies by BioID and superresolution microscopy methods led to considerable progress in our understanding of the composition, structure and function of this centrosome type. We discuss all currently known components of the Dictyostelium centrosome in comparison to other centrosomes of animals and yeasts.
Collapse
|
37
|
Sekulovski S, Devant P, Panizza S, Gogakos T, Pitiriciu A, Heitmeier K, Ramsay EP, Barth M, Schmidt C, Tuschl T, Baas F, Weitzer S, Martinez J, Trowitzsch S. Assembly defects of human tRNA splicing endonuclease contribute to impaired pre-tRNA processing in pontocerebellar hypoplasia. Nat Commun 2021; 12:5610. [PMID: 34584079 PMCID: PMC8479045 DOI: 10.1038/s41467-021-25870-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
Introns of human transfer RNA precursors (pre-tRNAs) are excised by the tRNA splicing endonuclease TSEN in complex with the RNA kinase CLP1. Mutations in TSEN/CLP1 occur in patients with pontocerebellar hypoplasia (PCH), however, their role in the disease is unclear. Here, we show that intron excision is catalyzed by tetrameric TSEN assembled from inactive heterodimers independently of CLP1. Splice site recognition involves the mature domain and the anticodon-intron base pair of pre-tRNAs. The 2.1-Å resolution X-ray crystal structure of a TSEN15-34 heterodimer and differential scanning fluorimetry analyses show that PCH mutations cause thermal destabilization. While endonuclease activity in recombinant mutant TSEN is unaltered, we observe assembly defects and reduced pre-tRNA cleavage activity resulting in an imbalanced pre-tRNA pool in PCH patient-derived fibroblasts. Our work defines the molecular principles of intron excision in humans and provides evidence that modulation of TSEN stability may contribute to PCH phenotypes.
Collapse
Affiliation(s)
- Samoil Sekulovski
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Pascal Devant
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt/Main, Germany
- Ph.D. Program in Virology, Harvard Medical School, Boston, MA, USA
- Harvard Medical School and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Silvia Panizza
- Max Perutz Labs, Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Tasos Gogakos
- Laboratory for RNA Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Anda Pitiriciu
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Katharina Heitmeier
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt/Main, Germany
| | | | - Marie Barth
- Interdisciplinary research center HALOmem, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Carla Schmidt
- Interdisciplinary research center HALOmem, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Thomas Tuschl
- Laboratory for RNA Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Frank Baas
- Department of Clinical Genetics, Leiden University, Leiden, Netherlands
| | - Stefan Weitzer
- Max Perutz Labs, Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Javier Martinez
- Max Perutz Labs, Medical University of Vienna, Vienna Biocenter (VBC), Vienna, Austria.
| | - Simon Trowitzsch
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Frankfurt/Main, Germany.
| |
Collapse
|
38
|
Kretzschmar K, Boonekamp KE, Bleijs M, Asra P, Koomen M, Chuva de Sousa Lopes SM, Giovannone B, Clevers H. Troy/Tnfrsf19 marks epidermal cells that govern interfollicular epidermal renewal and cornification. Stem Cell Reports 2021; 16:2379-2394. [PMID: 34358453 PMCID: PMC8452520 DOI: 10.1016/j.stemcr.2021.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 01/01/2023] Open
Abstract
The skin epidermis is a highly compartmentalized tissue consisting of a cornifying epithelium called the interfollicular epidermis (IFE) and associated hair follicles (HFs). Several stem cell populations have been described that mark specific compartments in the skin but none of them is specific to the IFE. Here, we identify Troy as a marker of IFE and HF infundibulum basal layer cells in developing and adult human and mouse epidermis. Genetic lineage-tracing experiments demonstrate that Troy-expressing basal cells contribute to long-term renewal of all layers of the cornifying epithelium. Single-cell transcriptomics and organoid assays of Troy-expressing cells, as well as their progeny, confirmed stem cell identity as well as the ability to generate differentiating daughter cells. In conclusion, we define Troy as a marker of epidermal basal cells that govern interfollicular epidermal renewal and cornification.
Collapse
Affiliation(s)
- Kai Kretzschmar
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, the Netherlands; Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, University Hospital Würzburg, 97080 Würzburg, Germany.
| | - Kim E Boonekamp
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, the Netherlands; German Cancer Research Centre (DKFZ), 69120 Heidelberg, Germany
| | - Margit Bleijs
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, the Netherlands; Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, the Netherlands
| | - Priyanca Asra
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, the Netherlands
| | - Mandy Koomen
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, the Netherlands
| | | | | | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, the Netherlands; Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, the Netherlands.
| |
Collapse
|
39
|
Wieczorek M, Ti SC, Urnavicius L, Molloy KR, Aher A, Chait BT, Kapoor TM. Biochemical reconstitutions reveal principles of human γ-TuRC assembly and function. J Cell Biol 2021; 220:211719. [PMID: 33496729 PMCID: PMC7844428 DOI: 10.1083/jcb.202009146] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/10/2020] [Accepted: 01/04/2021] [Indexed: 12/22/2022] Open
Abstract
The formation of cellular microtubule networks is regulated by the γ-tubulin ring complex (γ-TuRC). This ∼2.3 MD assembly of >31 proteins includes γ-tubulin and GCP2-6, as well as MZT1 and an actin-like protein in a “lumenal bridge” (LB). The challenge of reconstituting the γ-TuRC has limited dissections of its assembly and function. Here, we report a biochemical reconstitution of the human γ-TuRC (γ-TuRC-GFP) as a ∼35 S complex that nucleates microtubules in vitro. In addition, we generate a subcomplex, γ-TuRCΔLB-GFP, which lacks MZT1 and actin. We show that γ-TuRCΔLB-GFP nucleates microtubules in a guanine nucleotide–dependent manner and with similar efficiency as the holocomplex. Electron microscopy reveals that γ-TuRC-GFP resembles the native γ-TuRC architecture, while γ-TuRCΔLB-GFP adopts a partial cone shape presenting only 8–10 γ-tubulin subunits and lacks a well-ordered lumenal bridge. Our results show that the γ-TuRC can be reconstituted using a limited set of proteins and suggest that the LB facilitates the self-assembly of regulatory interfaces around a microtubule-nucleating “core” in the holocomplex.
Collapse
Affiliation(s)
- Michal Wieczorek
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY
| | - Shih-Chieh Ti
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY
| | - Linas Urnavicius
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY.,Laboratory of Cell Biology, The Rockefeller University, New York, NY
| | - Kelly R Molloy
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY
| | - Amol Aher
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University, New York, NY
| | - Tarun M Kapoor
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, New York, NY
| |
Collapse
|
40
|
Abstract
As one of four filament types, microtubules are a core component of the cytoskeleton and are essential for cell function. Yet how microtubules are nucleated from their building blocks, the αβ-tubulin heterodimer, has remained a fundamental open question since the discovery of tubulin 50 years ago. Recent structural studies have shed light on how γ-tubulin and the γ-tubulin complex proteins (GCPs) GCP2 to GCP6 form the γ-tubulin ring complex (γ-TuRC). In parallel, functional and single-molecule studies have informed on how the γ-TuRC nucleates microtubules in real time, how this process is regulated in the cell and how it compares to other modes of nucleation. Another recent surprise has been the identification of a second essential nucleation factor, which turns out to be the well-characterized microtubule polymerase XMAP215 (also known as CKAP5, a homolog of chTOG, Stu2 and Alp14). This discovery helps to explain why the observed nucleation activity of the γ-TuRC in vitro is relatively low. Taken together, research in recent years has afforded important insight into how microtubules are made in the cell and provides a basis for an exciting era in the cytoskeleton field.
Collapse
Affiliation(s)
- Akanksha Thawani
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sabine Petry
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
41
|
Interactome Analysis of KIN (Kin17) Shows New Functions of This Protein. Curr Issues Mol Biol 2021; 43:767-781. [PMID: 34449532 PMCID: PMC8929021 DOI: 10.3390/cimb43020056] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/16/2021] [Accepted: 06/24/2021] [Indexed: 11/16/2022] Open
Abstract
KIN (Kin17) protein is overexpressed in a number of cancerous cell lines, and is therefore considered a possible cancer biomarker. It is a well-conserved protein across eukaryotes and is ubiquitously expressed in all cell types studied, suggesting an important role in the maintenance of basic cellular function which is yet to be well determined. Early studies on KIN suggested that this nuclear protein plays a role in cellular mechanisms such as DNA replication and/or repair; however, its association with chromatin depends on its methylation state. In order to provide a better understanding of the cellular role of this protein, we investigated its interactome by proximity-dependent biotin identification coupled to mass spectrometry (BioID-MS), used for identification of protein-protein interactions. Our analyses detected interaction with a novel set of proteins and reinforced previous observations linking KIN to factors involved in RNA processing, notably pre-mRNA splicing and ribosome biogenesis. However, little evidence supports that this protein is directly coupled to DNA replication and/or repair processes, as previously suggested. Furthermore, a novel interaction was observed with PRMT7 (protein arginine methyltransferase 7) and we demonstrated that KIN is modified by this enzyme. This interactome analysis indicates that KIN is associated with several cell metabolism functions, and shows for the first time an association with ribosome biogenesis, suggesting that KIN is likely a moonlight protein.
Collapse
|
42
|
Jiang X, Ho DBT, Mahe K, Mia J, Sepulveda G, Antkowiak M, Jiang L, Yamada S, Jao LE. Condensation of pericentrin proteins in human cells illuminates phase separation in centrosome assembly. J Cell Sci 2021; 134:jcs258897. [PMID: 34308971 PMCID: PMC8349556 DOI: 10.1242/jcs.258897] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/08/2021] [Indexed: 11/24/2022] Open
Abstract
At the onset of mitosis, centrosomes expand the pericentriolar material (PCM) to maximize their microtubule-organizing activity. This step, termed centrosome maturation, ensures proper spindle organization and faithful chromosome segregation. However, as the centrosome expands, how PCM proteins are recruited and held together without membrane enclosure remains elusive. We found that endogenously expressed pericentrin (PCNT), a conserved PCM scaffold protein, condenses into dynamic granules during late G2/early mitosis before incorporating into mitotic centrosomes. Furthermore, the N-terminal portion of PCNT, enriched with conserved coiled-coils (CCs) and low-complexity regions (LCRs), phase separates into dynamic condensates that selectively recruit PCM proteins and nucleate microtubules in cells. We propose that CCs and LCRs, two prevalent sequence features in the centrosomal proteome, are preserved under evolutionary pressure in part to mediate liquid-liquid phase separation, a process that bestows upon the centrosome distinct properties critical for its assembly and functions.
Collapse
Affiliation(s)
- Xueer Jiang
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Dac Bang Tam Ho
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Karan Mahe
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Jennielee Mia
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Guadalupe Sepulveda
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Mark Antkowiak
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Linhao Jiang
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| | - Soichiro Yamada
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Li-En Jao
- Department of Cell Biology and Human Anatomy, University of California, Davis, School of Medicine, Davis, CA 95616, USA
| |
Collapse
|
43
|
Wang Y, Chen Z, Jia C, Bai X, Jiang Y, Zou Z. Analysis of the mTOR Interactome using SILAC technology revealed NICE-4 as a novel regulator of mTORC1 activity. Life Sci 2021; 281:119745. [PMID: 34171383 DOI: 10.1016/j.lfs.2021.119745] [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: 01/06/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 10/21/2022]
Abstract
The evolutionarily conserved mechanistic target of rapamycin (mTOR) forms two functionally distinct complexes, -the mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2)-which differ in their subunit composition. Although the function of mTORC1 has been studied extensively, the interaction between mTORC1 and the ubiquitin-proteasome system (UPS) remains unclear. To facilitate a thorough understanding of the mechanismby which UPS regulates mTORC1 activity, steady isotope labeling with amino acids in cell culture (SILAC) technology was used to screen for potential mTORC1-interacting UPS members. Fourteen previously unknown proteins bound to mTOR in HEK293 cells with a SILAC ratio (heavy/light, H/L) above 2, five of which are components of the UPS. Subsequent immunoprecipitation analysis confirmed that ubiquitin-relevant protein 2-like (UBAP2L, also known as NICE-4) binds to both mTOR and Raptor, but not Rictor, suggesting that NICE-4 specifically interacts with mTORC1, but not mTORC2. Interestingly, NICE-4 is essential for basic mTORC1 activity in both HeLa cancer cells and HEK293 cells. In addition, NICE-4 depletion markedly suppressed proliferation of both HeLa and HEK293 cells as well as survival of HeLa cells. Collectively, these results revealed the identity of novel mTOR-interacting UPS proteins and established NICE-4 as a critical UPS member that maintains mTORC1 activity.
Collapse
Affiliation(s)
- Yun Wang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China.
| | - Zhenguo Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China.
| | - Chunhong Jia
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China.
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA 15213, USA.
| | - Zhipeng Zou
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China.
| |
Collapse
|
44
|
Böhler A, Vermeulen BJA, Würtz M, Zupa E, Pfeffer S, Schiebel E. The gamma-tubulin ring complex: Deciphering the molecular organization and assembly mechanism of a major vertebrate microtubule nucleator. Bioessays 2021; 43:e2100114. [PMID: 34160844 DOI: 10.1002/bies.202100114] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 11/10/2022]
Abstract
Microtubules are protein cylinders with functions in cell motility, signal sensing, cell organization, intracellular transport, and chromosome segregation. One of the key properties of microtubules is their dynamic architecture, allowing them to grow and shrink in length by adding or removing copies of their basic subunit, the heterodimer αβ-tubulin. In higher eukaryotes, de novo assembly of microtubules from αβ-tubulin is initiated by a 2 MDa multi-subunit complex, the gamma-tubulin ring complex (γ-TuRC). For many years, the structure of the γ-TuRC and the function of its subunits remained enigmatic, although structural data from the much simpler yeast counterpart, the γ-tubulin small complex (γ-TuSC), were available. Two recent breakthroughs in the field, high-resolution structural analysis and recombinant reconstitution of the complex, have revolutionized our knowledge about the architecture and function of the γ-TuRC and will form the basis for addressing outstanding questions about biogenesis and regulation of this essential microtubule organizer.
Collapse
Affiliation(s)
- Anna Böhler
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Bram J A Vermeulen
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Martin Würtz
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Erik Zupa
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Stefan Pfeffer
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| | - Elmar Schiebel
- Zentrum für Molekulare Biologie der Universität Heidelberg, Heidelberg, Germany
| |
Collapse
|
45
|
Elting MW. Cytoskeletal biophysics: Passive crosslinker adapts to keep microtubule bundles on track. Curr Biol 2021; 31:R793-R796. [PMID: 34157264 DOI: 10.1016/j.cub.2021.04.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Assembly of the mitotic spindle requires dynamic adaptation and coordination among an array of motors and crosslinkers. A new study demonstrates in vitro how the mitotic crosslinker PRC1 can tune its behavior to regulate the speed of microtubule sliding.
Collapse
Affiliation(s)
- Mary Williard Elting
- Department of Physics, Cluster for Quantitative and Computational Developmental Biology, North Carolina State University, Raleigh, NC 27607, USA.
| |
Collapse
|
46
|
Wang H, Jiang X, Cheng Y, Ren H, Hu Y, Zhang Y, Su H, Zou Z, Wang Q, Liu Z, Zhang J, Qiu X. MZT2A promotes NSCLC viability and invasion by increasing Akt phosphorylation via the MOZART2 domain. Cancer Sci 2021; 112:2210-2222. [PMID: 33754417 PMCID: PMC8177791 DOI: 10.1111/cas.14900] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 12/17/2022] Open
Abstract
Mitotic spindle organizing protein 2A (MZT2A) is localized at the centrosome and regulates microtubule nucleation activity in cells. This study assessed the role of MZT2A in non-small-cell lung cancer (NSCLC). Differential MZT2A expression was bioinformatically assessed using TCGA database, the GEPIA database, and Kaplan-Meier survival data to determine the association between MZT2A expression and NSCLC prognosis. Furthermore, NSCLC tissue specimens were evaluated by immunohistochemistry. MZT2A was overexpressed or knocked down in NSCLC cells using cDNA and siRNA, respectively. The cells were subjected to various assays and treated with the selective Akt inhibitor LY294002 or co-transfected with galectin-3-binding protein (LGALS3BP) siRNA. MZT2A mRNA and protein levels were upregulated in NSCLC lesions and MTZ2A expression was associated with poor NSCLC prognosis. MZT2A protein was also highly expressed in NSCLC cells compared with the expression in normal bronchial cells. MZT2A expression promoted NSCLC cell viability and invasion, whereas MTZ2A siRNA had the opposite effect on NSCLC cells in vitro. At the protein level, MZT2A induced Akt phosphorylation, promoting NSCLC proliferation and invasion (but the selective Akt inhibitor blocked these effects) through upregulation of LGALS3BP via the MTZ2A MOZART2 domain, whereas LGALS3BP siRNA suppressed MTZ2A activity in NSCLC cells. The limited in vivo experiments confirmed the in vitro data. In conclusion, MZT2A exhibits oncogenic activity by activating LGALS3BP and Akt in NSCLC. Future studies will assess MTZ2A as a biomarker to predict NSCLC prognosis or as a target in the control of NSCLC progression.
Collapse
Affiliation(s)
- Huanxi Wang
- Department of PathologyChina Medical UniversityShenyangChina
| | - Xizi Jiang
- Department of PathologyChina Medical UniversityShenyangChina
| | - Yu Cheng
- Department of PathologyChina Medical UniversityShenyangChina
- Departemt of PathologyCancer Research LaboratoryChengde Medical CollegeChengdeChina
| | - Hongjiu Ren
- Department of PathologyChina Medical UniversityShenyangChina
| | - Yujiao Hu
- Department of PathologyChina Medical UniversityShenyangChina
| | - Yao Zhang
- Department of PathologyChina Medical UniversityShenyangChina
| | - Hongbo Su
- Department of PathologyChina Medical UniversityShenyangChina
| | - Zifang Zou
- Department of Thoracic SurgeryThe First Affiliated Hospital of China Medical UniversityShenyangChina
| | - Qiongzi Wang
- Department of PathologyChina Medical UniversityShenyangChina
| | - Zongang Liu
- Department of Thoracic SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Jiameng Zhang
- Department of PathologyChina Medical UniversityShenyangChina
| | - Xueshan Qiu
- Department of PathologyChina Medical UniversityShenyangChina
- Department of PathologyThe First Affiliated Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
47
|
Tovey CA, Tsuji C, Egerton A, Bernard F, Guichet A, de la Roche M, Conduit PT. Autoinhibition of Cnn binding to γ-TuRCs prevents ectopic microtubule nucleation and cell division defects. J Cell Biol 2021; 220:212197. [PMID: 34042945 PMCID: PMC8164090 DOI: 10.1083/jcb.202010020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 03/25/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022] Open
Abstract
γ-Tubulin ring complexes (γ-TuRCs) nucleate microtubules. They are recruited to centrosomes in dividing cells via binding to N-terminal CM1 domains within γ-TuRC–tethering proteins, including Drosophila Centrosomin (Cnn). Binding promotes microtubule nucleation and is restricted to centrosomes in dividing cells, but the mechanism regulating binding remains unknown. Here, we identify an extreme N-terminal CM1 autoinhibition (CAI) domain found specifically within the centrosomal isoform of Cnn (Cnn-C) that inhibits γ-TuRC binding. Robust binding occurs after removal of the CAI domain or with the addition of phosphomimetic mutations, suggesting that phosphorylation helps relieve inhibition. We show that regulation of Cnn binding to γ-TuRCs is isoform specific and that misregulation of binding can result in ectopic cytosolic microtubules and major defects during cell division. We also find that human CDK5RAP2 is autoinhibited from binding γ-TuRCs, suggesting conservation across species. Overall, our results shed light on how and why CM1 domain binding to γ-TuRCs is regulated.
Collapse
Affiliation(s)
- Corinne A Tovey
- Department of Zoology, University of Cambridge, Cambridge, UK.,Université de Paris, Centre National de la Recherche Scientifique, Institut Jacques Monod, Paris, France
| | - Chisato Tsuji
- Department of Zoology, University of Cambridge, Cambridge, UK
| | - Alice Egerton
- Department of Zoology, University of Cambridge, Cambridge, UK
| | - Fred Bernard
- Université de Paris, Centre National de la Recherche Scientifique, Institut Jacques Monod, Paris, France
| | - Antoine Guichet
- Université de Paris, Centre National de la Recherche Scientifique, Institut Jacques Monod, Paris, France
| | - Marc de la Roche
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Paul T Conduit
- Department of Zoology, University of Cambridge, Cambridge, UK.,Université de Paris, Centre National de la Recherche Scientifique, Institut Jacques Monod, Paris, France
| |
Collapse
|
48
|
Kolora SRR, Gysi DM, Schaffer S, Grimm-Seyfarth A, Szabolcs M, Faria R, Henle K, Stadler PF, Schlegel M, Nowick K. Accelerated Evolution of Tissue-Specific Genes Mediates Divergence Amidst Gene Flow in European Green Lizards. Genome Biol Evol 2021; 13:6275683. [PMID: 33988711 PMCID: PMC8382678 DOI: 10.1093/gbe/evab109] [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] [Accepted: 05/13/2021] [Indexed: 11/12/2022] Open
Abstract
The European green lizards of the Lacerta viridis complex consist of two closely related species, L. viridis and Lacerta bilineata that split less than 7 million years ago in the presence of gene flow. Recently, a third lineage, referred to as the “Adriatic” was described within the L. viridis complex distributed from Slovenia to Greece. However, whether gene flow between the Adriatic lineage and L. viridis or L. bilineata has occurred and the evolutionary processes involved in their diversification are currently unknown. We hypothesized that divergence occurred in the presence of gene flow between multiple lineages and involved tissue-specific gene evolution. In this study, we sequenced the whole genome of an individual of the Adriatic lineage and tested for the presence of gene flow amongst L. viridis, L. bilineata, and Adriatic. Additionally, we sequenced transcriptomes from multiple tissues to understand tissue-specific effects. The species tree supports that the Adriatic lineage is a sister taxon to L. bilineata. We detected gene flow between the Adriatic lineage and L. viridis suggesting that the evolutionary history of the L. viridis complex is likely shaped by gene flow. Interestingly, we observed topological differences between the autosomal and Z-chromosome phylogenies with a few fast evolving genes on the Z-chromosome. Genes highly expressed in the ovaries and strongly co-expressed in the brain experienced accelerated evolution presumably contributing to establishing reproductive isolation in the L. viridis complex.
Collapse
Affiliation(s)
- Sree Rohit Raj Kolora
- German Centre for Integrative Biodiversity Research (iDiv) Halle Jena Leipzig, Leipzig, Germany.,Bioinformatics Group, Department of Computer Science, and Interdisciplinary Center for Bioinformatics, Universität Leipzig, Leipzig, Germany.,Molecular Evolution & Animal Systematics, University of Leipzig, Leipzig, Germany.,Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Deisy Morselli Gysi
- Bioinformatics Group, Department of Computer Science, and Interdisciplinary Center for Bioinformatics, Universität Leipzig, Leipzig, Germany.,Swarm Intelligence and Complex Systems Group, Faculty of Mathematics and Computer Science, University of Leipzig, Leipzig, Germany.,Center for Complex Networks Research, Northeastern University, Boston, MA, USA
| | - Stefan Schaffer
- German Centre for Integrative Biodiversity Research (iDiv) Halle Jena Leipzig, Leipzig, Germany.,Molecular Evolution & Animal Systematics, University of Leipzig, Leipzig, Germany
| | - Annegret Grimm-Seyfarth
- Department of Conservation Biology, UFZ-Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Márton Szabolcs
- Department of Tisza River Research, Danube Research Institute, Centre for Ecological Research, Hungarian Academy of Sciences, Debrecen, Hungary
| | - Rui Faria
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO, Laboratório Associado, Universidade do Porto, Vairão, Portugal
| | - Klaus Henle
- German Centre for Integrative Biodiversity Research (iDiv) Halle Jena Leipzig, Leipzig, Germany.,Department of Conservation Biology, UFZ-Helmholtz Centre for Environmental Research, Leipzig, Germany
| | - Peter F Stadler
- German Centre for Integrative Biodiversity Research (iDiv) Halle Jena Leipzig, Leipzig, Germany.,Bioinformatics Group, Department of Computer Science, and Interdisciplinary Center for Bioinformatics, Universität Leipzig, Leipzig, Germany.,Competence Center for Scalable Data Services and Solutions Dresden/Leipzig, Universität Leipzig, Leipzig, Germany.,Max-Planck-Institute for Mathematics in the Sciences, Leipzig, Germany.,Department of Theoretical Chemistry, University of Vienna, Wien, Austria.,Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia.,Santa Fe Institute, New Mexico, USA
| | - Martin Schlegel
- German Centre for Integrative Biodiversity Research (iDiv) Halle Jena Leipzig, Leipzig, Germany.,Molecular Evolution & Animal Systematics, University of Leipzig, Leipzig, Germany
| | - Katja Nowick
- Institute for Biology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
49
|
Wieczorek M, Huang TL, Urnavicius L, Hsia KC, Kapoor TM. MZT Proteins Form Multi-Faceted Structural Modules in the γ-Tubulin Ring Complex. Cell Rep 2021; 31:107791. [PMID: 32610146 PMCID: PMC7416306 DOI: 10.1016/j.celrep.2020.107791] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/16/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
Microtubule organization depends on the γ-Tubulin ring complex (γ-TuRC), a ~2.3-MDa nucleation factor comprising an asymmetric assembly of γ-Tubulin and GCP2-GCP6. However, it is currently unclear how the γ-TuRC-associated microproteins MZT1 and MZT2 contribute to the structure and regulation of the holocomplex. Here, we report cryo-EM structures of MZT1 and MZT2 in the context of the native human γ-TuRC. MZT1 forms two subcomplexes with the N-terminal α-helical domains of GCP3 or GCP6 (GCP-NHDs) within the γ-TuRC “lumenal bridge.” We determine the X-ray structure of recombinant MZT1/GCP6-NHD and find it is similar to that within the native γ-TuRC. We identify two additional MZT/GCP-NHD-like subcomplexes, one of which is located on the outer face of the γ-TuRC and comprises MZT2 and GCP2-NHD in complex with a centrosomin motif 1 (CM1)-containing peptide. Our data reveal how MZT1 and MZT2 establish multi-faceted, structurally mimetic “modules” that can expand structural and regulatory interfaces in the γ-TuRC. Wieczorek et al. show how the microproteins MZT1 and MZT2 expand binding interfaces across the γ-TuRC—the cell’s microtubule nucleating machinery—by forming similarly shaped, “modular” subcomplexes with the α-helical N-terminal domains of different γ-Tubulin complex proteins (GCPs).
Collapse
Affiliation(s)
- Michal Wieczorek
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Tzu-Lun Huang
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica, and National Defense Medical Center, Taipei, Taiwan; Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Linas Urnavicius
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA; Laboratory of Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Kuo-Chiang Hsia
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica, and National Defense Medical Center, Taipei, Taiwan; Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemistry and Molecular Biology, College of Life Sciences, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Tarun M Kapoor
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
50
|
Tyc KM, El Yakoubi W, Bag A, Landis J, Zhan Y, Treff NR, Scott RT, Tao X, Schindler K, Xing J. Exome sequencing links CEP120 mutation to maternally derived aneuploid conception risk. Hum Reprod 2021; 35:2134-2148. [PMID: 32772081 DOI: 10.1093/humrep/deaa148] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/14/2020] [Indexed: 12/22/2022] Open
Abstract
STUDY QUESTION What are the genetic factors that increase the risk of aneuploid egg production? SUMMARY ANSWER A non-synonymous variant rs2303720 within centrosomal protein 120 (CEP120) disrupts female meiosis in vitro in mouse. WHAT IS KNOWN ALREADY The production of aneuploid eggs, with an advanced maternal age as an established contributing factor, is the major cause of IVF failure, early miscarriage and developmental anomalies. The identity of maternal genetic variants contributing to egg aneuploidy irrespective of age is missing. STUDY DESIGN, SIZE, DURATION Patients undergoing fertility treatment (n = 166) were deidentified and selected for whole-exome sequencing. PARTICIPANTS/MATERIALS, SETTING, METHODS Patients self-identified their ethnic groups and their ages ranged from 22 to 49 years old. The study was performed using genomes from White, non-Hispanic patients divided into controls (97) and cases (69) according to the number of aneuploid blastocysts derived during each IVF procedure. Following a gene prioritization strategy, a mouse oocyte system was used to validate the functional significance of the discovered associated genetic variants. MAIN RESULTS AND THE ROLE OF CHANCE Patients producing a high proportion of aneuploid blastocysts (considered aneuploid if they missed any of the 40 chromatids or had extra copies) were found to carry a higher mutational burden in genes functioning in cytoskeleton and microtubule pathways. Validation of the functional significance of a non-synonymous variant rs2303720 within Cep120 on mouse oocyte meiotic maturation revealed that ectopic expression of CEP120:p.Arg947His caused decreased spindle microtubule nucleation efficiency and increased incidence of aneuploidy. LIMITATIONS, REASONS FOR CAUTION Functional validation was performed using the mouse oocyte system. Because spindle building pathways differ between mouse and human oocytes, the defects we observed upon ectopic expression of the Cep120 variant may alter mouse oocyte meiosis differently than human oocyte meiosis. Further studies using knock-in 'humanized' mouse models and in human oocytes will be needed to translate our findings to human system. Possible functional differences of the variant between ethnic groups also need to be investigated. WIDER IMPLICATIONS OF THE FINDINGS Variants in centrosomal genes appear to be important contributors to the risk of maternal aneuploidy. Functional validation of these variants will eventually allow prescreening to select patients that have better chances to benefit from preimplantation genetic testing. STUDY FUNDING/COMPETING INTEREST(S) This study was funded through R01-HD091331 to K.S. and J.X. and EMD Serono Grant for Fertility Innovation to N.R.T. N.R.T. is a shareholder and an employee of Genomic Prediction. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Katarzyna M Tyc
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.,Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Warif El Yakoubi
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.,Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Aishee Bag
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jessica Landis
- Foundation for Embryonic Competence, Basking Ridge, NJ 07920, USA
| | - Yiping Zhan
- Foundation for Embryonic Competence, Basking Ridge, NJ 07920, USA
| | - Nathan R Treff
- Reproductive Medicine Associates of New Jersey, Basking Ridge, NJ 07920, USA
| | - Richard T Scott
- Reproductive Medicine Associates of New Jersey, Basking Ridge, NJ 07920, USA
| | - Xin Tao
- Foundation for Embryonic Competence, Basking Ridge, NJ 07920, USA
| | - Karen Schindler
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.,Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jinchuan Xing
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.,Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|