1
|
Mu Z, Zheng P, Liu S, Kang Y, Xie H. Plk4 regulates centriole duplication in the embryonic development of zebrafish. Dev Biol 2025; 517:148-156. [PMID: 39304174 DOI: 10.1016/j.ydbio.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/23/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
PLK4 plays a crucial role in centriole duplication, which is essential for maintaining cellular processes such as cell division, cytoskeletal stability, and cilia formation. However, the mechanisms of PLK4 remain incompletely understood, especially in the embryonic development of vertebrate species. In this study, we observed that Plk4 dysfunction led to abnormal embryonic development in zebrafish, characterized by symptoms such as dark and wrinkled skin, microphthalmia, and body axis curvature. In plk4 mutants, defects in centriole duplication led to abnormal cell division, apoptosis, and ciliogenesis defects. Moreover, overexpression of plk4 in zebrafish embryos caused excessive centrosome amplification, disrupting embryonic gastrulation through abnormal cell division and ultimately resulting in embryonic lethality. Furthermore, we identified the "cryptic" polo box (CPB) domain, consisting of two PBs (PB1 and PB2), as the critical centrosome localization domain of Plk4. Surprisingly, overexpression of these two PB domains alone was sufficient to induce embryonic lethality. Additionally, we discovered a truncated form of CPB that localizes to the centrosome without causing defects in embryonic development. Our results demonstrate that Plk4 tightly controls centriole duplication, which is essential for early embryonic development in zebrafish.
Collapse
Affiliation(s)
- Zhiyu Mu
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Pengfei Zheng
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Shuangyu Liu
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Yunsi Kang
- Key Laboratory of Evolution and Marine Biodiversity of the Ministry of Education, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Haibo Xie
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China; Key Laboratory of Evolution and Marine Biodiversity of the Ministry of Education, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
2
|
Huang F, Zhang C. Visualizing Cartwheel Disassembly Process During Mitosis in Fixed and Live Cells by Fluorescence Microscope. Methods Mol Biol 2025; 2874:77-85. [PMID: 39614048 DOI: 10.1007/978-1-0716-4236-8_7] [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: 12/01/2024]
Abstract
Centrosome is an evolutionarily conserved organelle that comprises two barrel-shaped centrioles surrounded by pericentriolar material (PCM). It functions as the major microtubule-organizing center (MTOC) to regulate cell polarity, motility, intracellular material transport during interphase, and bipolar spindle assembly during mitosis. Cartwheel assembly is considered the first step in the initiation of procentriole biogenesis at early S phase. In human cells, cartwheel is a transient scaffolding structure that is disassembled during procentriole maturation at mitotic exit. This chapter describes methods for synchronizing mammalian cells into mitosis by small molecule inhibitor treatment and visualizing cartwheel disassembly process during mitosis in fixed and live cells by fluorescence microscope.
Collapse
Affiliation(s)
- Fan Huang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Chuanmao Zhang
- The Academy for Cell and Life Health, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China.
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
3
|
Khoury Damaa M, Serizay J, Balagué R, Boudjema AR, Faucourt M, Delgehyr N, Goh KJ, Lu H, Tan EK, James CT, Faucon C, Mitri R, Bracht DC, Bingle CD, Dunn NR, Arnold SJ, Zaragosi LE, Barbry P, Koszul R, Omran H, Gil-Gómez G, Escudier E, Legendre M, Roy S, Spassky N, Meunier A. Cyclin O controls entry into the cell-cycle variant required for multiciliated cell differentiation. Cell Rep 2024:115117. [PMID: 39740663 DOI: 10.1016/j.celrep.2024.115117] [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: 08/28/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Multiciliated cells (MCCs) ensure fluid circulation in various organs. Their differentiation is marked by the amplification of cilia-nucleating centrioles, driven by a genuine cell-cycle variant, which is characterized by wave-like expression of canonical and non-canonical cyclins such as Cyclin O (CCNO). Patients with CCNO mutations exhibit a subtype of primary ciliary dyskinesia called reduced generation of motile cilia (RGMC). Here, we show that Ccno is activated at the crossroads of the onset of MCC differentiation, the entry into the MCC cell-cycle variant, and the activation of the centriole biogenesis program. Its absence blocks the G1/S-like transition of the cell-cycle variant, interrupts the centriologenesis transcription program, and compromises the production of centrioles and cilia in mouse brain and human respiratory MCCs. Altogether, our study identifies CCNO as a core regulator of entry into the MCC cell-cycle variant and the interruption of this variant as one etiology of RGMC.
Collapse
Affiliation(s)
- Michella Khoury Damaa
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Jacques Serizay
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France; Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, France
| | - Rémi Balagué
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Amélie-Rose Boudjema
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Nathalie Delgehyr
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Kim Jee Goh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
| | - Hao Lu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Ee Kim Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
| | - Cameron T James
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, 61 Biopolis Drive, Singapore 138673, Singapore; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2RX, UK; Singapore-MIT Association for Research and Technology, Critical Analytics for Manufacturing Personalised-Medicine (SMART CAMP), 1 CREATE Way, Singapore 138602, Singapore
| | - Catherine Faucon
- Centre Hospitalier Intercommunal de Créteil, Laboratoire de Microscopie Électronique, Service d'Anatomopathologie, 94010 Créteil, France
| | - Rana Mitri
- Centre Hospitalier Intercommunal de Créteil, Laboratoire de Microscopie Électronique, Service d'Anatomopathologie, 94010 Créteil, France
| | - Diana Carolin Bracht
- Department of General Pediatrics, University Hospital Muenster, 48149 Muenster, Germany
| | - Colin D Bingle
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2RX, UK
| | - Norris Ray Dunn
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore; Skin Research Institute of Singapore, 11 Mandalay Road #17-01 Clinical Sciences Building, Singapore 308232, Singapore
| | - Sebastian J Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstrasse 25, 79104 Freiburg, Germany; Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Schänzlestrasse18, 79104 Freiburg, Germany
| | - Laure-Emmanuelle Zaragosi
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
| | - Pascal Barbry
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France; 3IA Côte d'Azur, 06560 Sophia Antipolis, France
| | - Romain Koszul
- Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, France
| | - Heymut Omran
- Department of General Pediatrics, University Hospital Muenster, 48149 Muenster, Germany
| | - Gabriel Gil-Gómez
- Hospital del Mar Research Institute, Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Estelle Escudier
- Sorbonne Université, Inserm, Childhood genetic diseases UMR_S933, Hôpital Armand-Trousseau, 75012 Paris, France; AP-HP, Sorbonne Université, Hôpital Armand-Trousseau, 75012 Paris, France
| | - Marie Legendre
- Sorbonne Université, Inserm, Childhood genetic diseases UMR_S933, Hôpital Armand-Trousseau, 75012 Paris, France; AP-HP, Sorbonne Université, Hôpital Armand-Trousseau, 75012 Paris, France
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, 61 Biopolis Drive, Singapore 138673, Singapore; Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119288, Singapore
| | - Nathalie Spassky
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France
| | - Alice Meunier
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, Ecole Normale Supérieure, PSL Research University, Paris, France.
| |
Collapse
|
4
|
Sparr C, Meitinger F. Prolonged mitosis: A key indicator for detecting stressed and damaged cells. Curr Opin Cell Biol 2024; 92:102449. [PMID: 39721293 DOI: 10.1016/j.ceb.2024.102449] [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: 10/01/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024]
Abstract
During mitosis, chromosomes condense, align to form a metaphase plate and segregate to the two daughter cells. Mitosis is one of the most complex recurring transformations in the life of a cell and requires a high degree of reliability to ensure the error-free transmission of genetic information to the next cell generation. An abnormally prolonged mitosis indicates potential defects that compromise genomic integrity. The mitotic stopwatch pathway detects even moderately prolonged mitoses by integrating memories of mitotic durations, ultimately leading to p53-mediated cell cycle arrest or death. This mechanism competes with mitogen signaling to stop the proliferation of damaged and potentially dangerous cells at a pre-oncogenic stage. Mitosis is a highly vulnerable phase, which is affected by multiple types of cellular damages and diverse stresses. We discuss the hypothesis that the duration of mitosis serves as an indicator of cell health.
Collapse
Affiliation(s)
- Carmen Sparr
- Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| | - Franz Meitinger
- Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan.
| |
Collapse
|
5
|
Serizay J, Khoury Damaa M, Boudjema AR, Balagué R, Faucourt M, Delgehyr N, Noûs C, Zaragosi LE, Barbry P, Spassky N, Koszul R, Meunier A. Cyclin switch tailors a cell cycle variant to orchestrate multiciliogenesis. Cell Rep 2024:115103. [PMID: 39740664 DOI: 10.1016/j.celrep.2024.115103] [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: 08/28/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 01/02/2025] Open
Abstract
Meiosis, endoreplication, and asynthetic fissions are variations of the canonical cell cycle where either replication or mitotic divisions are muted. Here, we identify a cell cycle variantconserved across organs and mammals, where both replication and mitosis are muted, and that orchestrates the differentiation of post-mitotic progenitors into multiciliated cells (MCCs). MCC progenitors reactivate most of the cell cycle transcriptional program but replace the temporal expression of cyclins E2 and A2 with non-canonical cyclins O and A1. In addition, the primary APC/C inhibitor Emi1 is silenced. Re-expressing cyclins E2 and A2 and/or Emi1 can induce partial replication or mitosis. This shows that a cell can co-opt the cell cycle genetic program and regulate only certain elements to qualitatively and quantitatively divert CDK activity toward differentiation rather than division. We propose this cell cycle variant to exploit the existence of a cytoplasmic-or centriolar-CDK threshold lower than the S-phase threshold.
Collapse
Affiliation(s)
- Jacques Serizay
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France; Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, France.
| | - Michella Khoury Damaa
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Amélie-Rose Boudjema
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Rémi Balagué
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Nathalie Delgehyr
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Camille Noûs
- Cogitamus Laboratory, PSL University, Paris, France
| | - Laure-Emmanuelle Zaragosi
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
| | - Pascal Barbry
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Sophia Antipolis, France
| | - Nathalie Spassky
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France
| | - Romain Koszul
- Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, France
| | - Alice Meunier
- Institut de Biologie de l'ENS (IBENS), CNRS, INSERM, École Normale Supérieure, PSL Research University, Paris, France.
| |
Collapse
|
6
|
Tollervey F, Rios MU, Zagoriy E, Woodruff JB, Mahamid J. Molecular architectures of centrosomes in C. elegans embryos visualized by cryo-electron tomography. Dev Cell 2024:S1534-5807(24)00724-X. [PMID: 39721584 DOI: 10.1016/j.devcel.2024.12.002] [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: 04/03/2024] [Revised: 09/29/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
Centrosomes organize microtubules that are essential for mitotic divisions in animal cells. They consist of centrioles surrounded by pericentriolar material (PCM). Questions related to mechanisms of centriole assembly, PCM organization, and spindle microtubule formation remain unanswered, partly due to limited availability of molecular-resolution structural data inside cells. Here, we use cryo-electron tomography to visualize centrosomes across the cell cycle in cells isolated from C. elegans embryos. We describe a pseudo-timeline of centriole assembly and identify distinct structural features in both mother and daughter centrioles. We find that centrioles and PCM microtubules differ in protofilament number (13 versus 11), which could be explained by atypical γ-tubulin ring complexes with 11-fold symmetry identified at the minus ends of short PCM microtubule segments. We further characterize a porous and disordered network that forms the interconnected PCM. Thus, our work builds a three-dimensional structural atlas that helps explain how centrosomes assemble, grow, and achieve function.
Collapse
Affiliation(s)
- Fergus Tollervey
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany; Collaboration for Joint PhD Degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Manolo U Rios
- Department of Cell Biology and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Evgenia Zagoriy
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Jeffrey B Woodruff
- Department of Cell Biology and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Julia Mahamid
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany; Cell Biology and Biophysics Unit, EMBL, 69117 Heidelberg, Germany.
| |
Collapse
|
7
|
Ohtsuka S, Kato H, Ishikawa R, Watanabe H, Miyazaki R, Katsuragi SY, Yoshimura K, Yamada H, Sakai Y, Inoue Y, Takanashi Y, Sekihara K, Funai K, Sugimura H, Shinmura K. STIL Overexpression Is Associated with Chromosomal Numerical Abnormalities in Non-Small-Cell Lung Carcinoma Through Centrosome Amplification. Curr Oncol 2024; 31:7936-7949. [PMID: 39727708 DOI: 10.3390/curroncol31120585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
STIL is a regulatory protein essential for centriole biogenesis, and its dysregulation has been implicated in various diseases, including malignancies. However, its role in non-small-cell lung carcinoma (NSCLC) remains unclear. In this study, we examined STIL expression and its potential association with chromosomal numerical abnormalities (CNAs) in NSCLC using The Cancer Genome Atlas (TCGA) dataset, immunohistochemical analysis, and in vitro experiments with NSCLC cell lines designed to overexpress STIL. TCGA data revealed upregulated STIL mRNA expression in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), the two major subtypes of NSCLC. Immunohistochemical analysis of cases from our hospital (LUAD, n = 268; LUSC, n = 98) revealed STIL protein overexpression. To elucidate the functional role of STIL, an inducible STIL-overexpressing H1299 NSCLC cell line was generated. Overexpression of STIL in these cells promoted centrosome amplification, leading to chromosomal instability. Finally, analysis of arm-level chromosomal copy number alterations from the TCGA dataset revealed that elevated STIL mRNA expression was associated with CNAs in both LUAD and LUSC. These findings suggest that STIL overexpression is associated with CNAs in NSCLC, likely through centrosome amplification, which is linked to chromosomal instability and might represent a potential therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Shunsuke Ohtsuka
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Hisami Kato
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Rei Ishikawa
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Hirofumi Watanabe
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Ryosuke Miyazaki
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Shin-Ya Katsuragi
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Katsuhiro Yoshimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Hidetaka Yamada
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Yasuhiro Sakai
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Yusuke Inoue
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Yusuke Takanashi
- Department of Surgery 1, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Keigo Sekihara
- Department of Surgery 1, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Kazuhito Funai
- Department of Surgery 1, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Haruhiko Sugimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
- Kyoundo Hospital, Sasaki Foundation, Tokyo 101-0062, Japan
| | - Kazuya Shinmura
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| |
Collapse
|
8
|
Tilwani S, Gandhi K, Dalal SN. 14-3-3ε conditional knockout mice exhibit defects in the development of the epidermis. FEBS Lett 2024; 598:3005-3020. [PMID: 39511902 DOI: 10.1002/1873-3468.15051] [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: 02/27/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024]
Abstract
The epidermis is a stratified epithelium that functions as the first line of defense against pathogenic invasion and acts as a barrier preventing water loss. In this study, we aimed to decipher the role of 14-3-3ε in the development of the epidermis. We report that loss of 14-3-3ε in the epidermis of juvenile and adult mice reduces cell division in the basal layer and increases the percentage of cells with multiple centrosomes, leading to a reduction in the thickness of the basal and stratified layers. We also demonstrate a decrease in the expression of differentiation markers, although no gross morphological defects in the skin or adverse effects on the survival of the mice were observed. These results suggest that loss of 14-3-3ε in the epidermis may lead to defects in proliferation and differentiation.
Collapse
Affiliation(s)
- Sarika Tilwani
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Karan Gandhi
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Sorab N Dalal
- Cell and Tumor Biology, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
9
|
Nguyen TK, Baker S, Rodriguez JM, Arceri L, Wingert RA. Using Zebrafish to Study Multiciliated Cell Development and Disease States. Cells 2024; 13:1749. [PMID: 39513856 PMCID: PMC11545745 DOI: 10.3390/cells13211749] [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: 10/04/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Multiciliated cells (MCCs) serve many important functions, including fluid propulsion and chemo- and mechanosensing. Diseases ranging from rare conditions to the recent COVID-19 global health pandemic have been linked to MCC defects. In recent years, the zebrafish has emerged as a model to investigate the biology of MCCs. Here, we review the major events in MCC formation including centriole biogenesis and basal body docking. Then, we discuss studies on the role of MCCs in diseases of the brain, respiratory, kidney and reproductive systems, as well as recent findings about the link between MCCs and SARS-CoV-2. Next, we explore why the zebrafish is a useful model to study MCCs and provide a comprehensive overview of previous studies of genetic components essential for MCC development and motility across three major tissues in the zebrafish: the pronephros, brain ependymal cells and nasal placode. Taken together, here we provide a cohesive summary of MCC research using the zebrafish and its future potential for expanding our understanding of MCC-related disease states.
Collapse
Affiliation(s)
- Thanh Khoa Nguyen
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (S.B.); (J.-M.R.); (L.A.)
| | | | | | | | - Rebecca A. Wingert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (S.B.); (J.-M.R.); (L.A.)
| |
Collapse
|
10
|
Schapfl MA, LoMastro GM, Braun VZ, Hirai M, Levine MS, Kiermaier E, Labi V, Holland AJ, Villunger A. Centrioles are frequently amplified in early B cell development but dispensable for humoral immunity. Nat Commun 2024; 15:8890. [PMID: 39406735 PMCID: PMC11480410 DOI: 10.1038/s41467-024-53222-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Centrioles define centrosome structure and function. Deregulation of centriole numbers can cause developmental defects and cancer. The p53 tumor suppressor limits the growth of cells lacking or harboring additional centrosomes and can be engaged by the "mitotic surveillance" or the "PIDDosome pathway", respectively. Here, we show that early B cell progenitors frequently present extra centrioles, ensuing their high proliferative activity and related DNA damage. Extra centrioles are efficiently cleared during B cell maturation. In contrast, centriole loss upon Polo-like kinase 4 (Plk4) deletion causes apoptosis and arrests B cell development. This defect can be rescued by co-deletion of Usp28, a critical component of the mitotic surveillance pathway, that restores cell survival and maturation. Centriole-deficient mature B cells are proliferation competent and mount a humoral immune response. Our findings imply that progenitor B cells are intolerant to centriole loss but permissive to centriole amplification, a feature potentially facilitating their malignant transformation.
Collapse
Affiliation(s)
- Marina A Schapfl
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Gina M LoMastro
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vincent Z Braun
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Maretoshi Hirai
- Department of Pharmacology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Michelle S Levine
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eva Kiermaier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Verena Labi
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
- The Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
11
|
Yeow ZY, Sarju S, Breugel MV, Holland AJ. Mesoscale regulation of MTOCs by the E3 ligase TRIM37. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.09.617407. [PMID: 39416078 PMCID: PMC11482927 DOI: 10.1101/2024.10.09.617407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Centrosomes ensure accurate chromosome segregation during cell division. Although the regulation of centrosome number is well-established, less is known about the suppression of non-centrosomal MTOCs (ncMTOCs). The E3 ligase TRIM37, implicated in Mulibrey nanism and 17q23-amplified cancers, has emerged as a key regulator of both centrosomes and ncMTOCs. Yet, the mechanism by which TRIM37 achieves enzymatic activation to target these mesoscale structures had remained unknown. Here, we elucidate TRIM37's activation process, beginning with TRAF domain-directed substrate recognition, progressing through B-box domain-mediated oligomerization, and culminating in RING domain dimerization. Using optogenetics, we demonstrate that TRIM37's E3 activity is directly coupled to the assembly state of its substrates, activating only when centrosomal proteins cluster into higher-order assemblies resembling MTOCs. This regulatory framework provides a mechanistic basis for understanding TRIM37-driven pathologies and, by echoing TRIM5's restriction of the HIV capsid, unveils a conserved activation blueprint among TRIM proteins for controlling mesoscale assembly turnover.
Collapse
Affiliation(s)
- Zhong Y Yeow
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sonia Sarju
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mark V Breugel
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, E1 2AT, UK
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
12
|
Özkeçeci CF, Arslan M, Başaran EG, Ergen YM, Bozdoğan Ö, Balamtekin N. Non-syndromic perspective on a unique progressive familial intrahepatic cholestasis variant: ZFYVE19 mutation. Turk J Pediatr 2024; 66:505-510. [PMID: 39387424 DOI: 10.24953/turkjpediatr.2024.4655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/05/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND ZFYVE19 mutation has been recently identified as one of the non-syndromic causes of cholestasis. It is associated with elevated gamma-glutamyl transferase levels and is likely a cause of neonatal-onset and intrahepatic cholestasis. CASE Here, we report a rare case of ZFYVE19 defect, confirmed by whole exome sequencing (WES). Our patient, who is currently 4 years old, presented to us at the age of 2 years with elevated levels of serum transaminases and bilirubin. WES revealed a homozygous ZFYVE19 mutation despite preserved synthetic liver function. This gene has recently been identified in the literature as a cause of non-classical progressive familial intrahepatic cholestasis (OMIM # 619849). Treatment with an appropriate dose of ursodeoxycholic acid resulted in the regression of elevated liver enzymes and itching. The patient's body mass index progressively increased throughout the treatment period. No medication side effects were observed at any point. Currently, the patient remains asymptomatic during follow-up. CONCLUSION We have identified the ZFYVE19 mutation as a variant that is not accompanied by any other symptoms. However, we have limited knowledge about the progression of the disease and are closely monitoring the patient for potential liver-related issues. Using WES in cases of undiagnosed liver enzyme elevations or cholestasis can help identify new genes and improve our understanding of the underlying pathophysiology.
Collapse
Affiliation(s)
- Coşkun Fırat Özkeçeci
- Division of Pediatric Gastroenterology, Department of Pediatrics, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Melike Arslan
- Division of Pediatric Gastroenterology, Department of Pediatrics, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Edibe Gözde Başaran
- Division of Pediatric Gastroenterology, Department of Pediatrics, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Yasin Maruf Ergen
- Division of Pediatric Gastroenterology, Department of Pediatrics, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Önder Bozdoğan
- Department of Pathology, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Necati Balamtekin
- Division of Pediatric Gastroenterology, Department of Pediatrics, Gülhane Training and Research Hospital, Ankara, Türkiye
| |
Collapse
|
13
|
Zenge C, Ordureau A. Ubiquitin system mutations in neurological diseases. Trends Biochem Sci 2024; 49:875-887. [PMID: 38972780 PMCID: PMC11455613 DOI: 10.1016/j.tibs.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/31/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
Neuronal ubiquitin balance impacts the fate of countless cellular proteins, and its disruption is associated with various neurological disorders. The ubiquitin system is critical for proper neuronal cell state transitions and the clearance of misfolded or aggregated proteins that threaten cellular integrity. This article reviews the state of and recent advancements in our understanding of the disruptions to components of the ubiquitin system, in particular E3 ligases and deubiquitylases, in neurodevelopmental and neurodegenerative diseases. Specific focus is on enzymes with recent progress in their characterization, including identifying enzyme-substrate pairs, the use of stem cell and animal models, and the development of therapeutics for ubiquitin-related diseases.
Collapse
Affiliation(s)
- Colin Zenge
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
14
|
Kiermaier E, Stötzel I, Schapfl MA, Villunger A. Amplified centrosomes-more than just a threat. EMBO Rep 2024; 25:4153-4167. [PMID: 39285247 PMCID: PMC11467336 DOI: 10.1038/s44319-024-00260-0] [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: 04/19/2024] [Revised: 07/05/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Centrosomes are major organizing components of the tubulin-based cytoskeleton. In recent years, we have gained extensive knowledge about their structure, biogenesis, and function from single cells, cell-cell interactions to tissue homeostasis, including their role in human diseases. Centrosome abnormalities are linked to, among others primary microcephaly, birth defects, ciliopathies, and tumorigenesis. Centrosome amplification, a state where two or more centrosomes are present in the G1 phase of the cell cycle, correlates in cancer with karyotype alterations, clinical aggressiveness, and lymph node metastasis. However, amplified centrosomes also appear in healthy tissues and, independent of their established role, in multi-ciliation. One example is the liver where hepatocytes carry amplified centrosomes owing to whole-genome duplication events during organogenesis. More recently, amplified centrosomes have been found in neuronal progenitors and several cell types of hematopoietic origin in which they enhance cellular effector functions. These findings suggest that extra centrosomes do not necessarily pose a risk for genome integrity and are harnessed for physiological processes. Here, we compare established and emerging 'non-canonical functions' of amplified centrosomes in cancerous and somatic cells and discuss their role in cellular physiology.
Collapse
Affiliation(s)
- Eva Kiermaier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany.
| | - Isabel Stötzel
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Marina A Schapfl
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
- The Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Lazarettgasse 14, 1090, Vienna, Austria.
| |
Collapse
|
15
|
Dwivedi D, Meraldi P. Balancing Plk1 activity levels: The secret of synchrony between the cell and the centrosome cycle. Bioessays 2024; 46:e2400048. [PMID: 39128131 DOI: 10.1002/bies.202400048] [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: 02/29/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
The accuracy of cell division requires precise regulation of the cellular machinery governing DNA/genome duplication, ensuring its equal distribution among the daughter cells. The control of the centrosome cycle is crucial for the formation of a bipolar spindle, ensuring error-free segregation of the genome. The cell and centrosome cycles operate in close synchrony along similar principles. Both require a single duplication round in every cell cycle, and both are controlled by the activity of key protein kinases. Nevertheless, our comprehension of the precise cellular mechanisms and critical regulators synchronizing these two cycles remains poorly defined. Here, we present our hypothesis that the spatiotemporal regulation of a dynamic equilibrium of mitotic kinases activities forms a molecular clock that governs the synchronous progression of both the cell and the centrosome cycles.
Collapse
Affiliation(s)
- Devashish Dwivedi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Translational Research Centre in Onco-haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Translational Research Centre in Onco-haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
16
|
Stracker TH. Regulation of p53 by the mitotic surveillance/stopwatch pathway: implications in neurodevelopment and cancer. Front Cell Dev Biol 2024; 12:1451274. [PMID: 39398482 PMCID: PMC11466822 DOI: 10.3389/fcell.2024.1451274] [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: 06/18/2024] [Accepted: 09/06/2024] [Indexed: 10/15/2024] Open
Abstract
The transcription factor p53 (encoded by TP53) plays diverse roles in human development and disease. While best known for its role in tumor suppression, p53 signaling also influences mammalian development by triggering cell fate decisions in response to a wide variety of stresses. After over 4 decades of study, a new pathway that triggers p53 activation in response to mitotic delays was recently identified. Termed the mitotic surveillance or mitotic stopwatch pathway, the USP28 and 53BP1 proteins activate p53 in response to delayed mitotic progression to control cell fate and promote genomic stability. In this Minireview, I discuss its identification, potential roles in neurodevelopmental disorders and cancer, as well as explore outstanding questions about its function, regulation and potential use as a biomarker for anti-mitotic therapies.
Collapse
Affiliation(s)
- Travis H. Stracker
- Center for Cancer Research, Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
17
|
Krüger S, Pfaff N, Gräf R, Meyer I. Dynamic Mitotic Localization of the Centrosomal Kinases CDK1, Plk, AurK, and Nek2 in Dictyostelium amoebae. Cells 2024; 13:1513. [PMID: 39329697 PMCID: PMC11430746 DOI: 10.3390/cells13181513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024] Open
Abstract
The centrosome of the amoebozoan model Dictyostelium discoideum provides the best-established model for an acentriolar centrosome outside the Opisthokonta. Dictyostelium exhibits an unusual centrosome cycle, in which duplication is initiated only at the G2/M transition and occurs entirely during the M phase. Little is known about the role of conserved centrosomal kinases in this process. Therefore, we have generated knock-in strains for Aurora (AurK), CDK1, cyclin B, Nek2, and Plk, replacing the endogenous genes with constructs expressing the respective green fluorescent Neon fusion proteins, driven by the endogenous promoters, and studied their behavior in living cells. Our results show that CDK1 and cyclin B arrive at the centrosome first, already during G2, followed by Plk, Nek2, and AurK. Furthermore, CDK1/cyclin B and AurK were dynamically localized at kinetochores, and AurK in addition at nucleoli. The putative roles of all four kinases in centrosome duplication, mitosis, cytokinesis, and nucleolar dynamics are discussed.
Collapse
Affiliation(s)
| | | | | | - Irene Meyer
- Department of Cell Biology, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam-Golm, Germany; (S.K.); (R.G.)
| |
Collapse
|
18
|
Thomas A, Meraldi P. Centrosome age breaks spindle size symmetry even in cells thought to divide symmetrically. J Cell Biol 2024; 223:e202311153. [PMID: 39012627 PMCID: PMC11252449 DOI: 10.1083/jcb.202311153] [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: 11/27/2023] [Revised: 03/14/2024] [Accepted: 05/03/2024] [Indexed: 07/17/2024] Open
Abstract
Centrosomes are the main microtubule-organizing centers in animal cells. Due to the semiconservative nature of centrosome duplication, the two centrosomes differ in age. In asymmetric stem cell divisions, centrosome age can induce an asymmetry in half-spindle lengths. However, whether centrosome age affects the symmetry of the two half-spindles in tissue culture cells thought to divide symmetrically is unknown. Here, we show that in human epithelial and fibroblastic cell lines centrosome age imposes a mild spindle asymmetry that leads to asymmetric cell daughter sizes. At the mechanistic level, we show that this asymmetry depends on a cenexin-bound pool of the mitotic kinase Plk1, which favors the preferential accumulation on old centrosomes of the microtubule nucleation-organizing proteins pericentrin, γ-tubulin, and Cdk5Rap2, and microtubule regulators TPX2 and ch-TOG. Consistently, we find that old centrosomes have a higher microtubule nucleation capacity. We postulate that centrosome age breaks spindle size symmetry via microtubule nucleation even in cells thought to divide symmetrically.
Collapse
Affiliation(s)
- Alexandre Thomas
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Translational Research Centre in Onco-hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Translational Research Centre in Onco-hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
19
|
Skinner MW, Simington CJ, López-Jiménez P, Baran KA, Xu J, Dayani Y, Pryzhkova MV, Page J, Gómez R, Holland AJ, Jordan PW. Spermatocytes have the capacity to segregate chromosomes despite centriole duplication failure. EMBO Rep 2024; 25:3373-3405. [PMID: 38943004 PMCID: PMC11316026 DOI: 10.1038/s44319-024-00187-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/30/2024] Open
Abstract
Centrosomes are the canonical microtubule organizing centers (MTOCs) of most mammalian cells, including spermatocytes. Centrosomes comprise a centriole pair within a structurally ordered and dynamic pericentriolar matrix (PCM). Unlike in mitosis, where centrioles duplicate once per cycle, centrioles undergo two rounds of duplication during spermatogenesis. The first duplication is during early meiotic prophase I, and the second is during interkinesis. Using mouse mutants and chemical inhibition, we have blocked centriole duplication during spermatogenesis and determined that non-centrosomal MTOCs (ncMTOCs) can mediate chromosome segregation. This mechanism is different from the acentriolar MTOCs that form bipolar spindles in oocytes, which require PCM components, including gamma-tubulin and CEP192. From an in-depth analysis, we identified six microtubule-associated proteins, TPX2, KIF11, NuMA, and CAMSAP1-3, that localized to the non-centrosomal MTOC. These factors contribute to a mechanism that ensures bipolar MTOC formation and chromosome segregation during spermatogenesis when centriole duplication fails. However, despite the successful completion of meiosis and round spermatid formation, centriole inheritance and PLK4 function are required for normal spermiogenesis and flagella assembly, which are critical to ensure fertility.
Collapse
Affiliation(s)
- Marnie W Skinner
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Carter J Simington
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Pablo López-Jiménez
- Department of Biology, Autonomous University of Madrid, Madrid, Spain
- MRC Laboratory of Medical Sciences, London, W12 0NN, UK
| | - Kerstin A Baran
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jingwen Xu
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yaron Dayani
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Marina V Pryzhkova
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Jesús Page
- Department of Biology, Autonomous University of Madrid, Madrid, Spain
| | - Rocío Gómez
- Department of Biology, Autonomous University of Madrid, Madrid, Spain
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Philip W Jordan
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
20
|
Martinez A, Stemm-Wolf AJ, Sheridan RM, Taliaferro MJ, Pearson CG. The Unkempt RNA binding protein reveals a local translation program in centriole overduplication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605660. [PMID: 39131325 PMCID: PMC11312568 DOI: 10.1101/2024.07.29.605660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Excess centrosomes cause defects in mitosis, cell-signaling, and cell migration, and therefore their assembly is tightly regulated. Plk4 controls centriole duplication at the heart of centrosome assembly, and elevation of Plk4 promotes centrosome amplification (CA), a founding event of tumorigenesis. Here, we investigate the transcriptional consequences of elevated Plk4 and find Unkempt, a gene encoding an RNA binding protein with roles in translational regulation, to be one of only two upregulated mRNAs. Unk protein localizes to centrosomes and Cep131-positive centriolar satellites and is required for Plk4-induced centriole overduplication in an RNA-binding dependent manner. Translation is enriched at centrosomes and centriolar satellites with Unk and Cep131 promoting this localized translation. A transient centrosomal downregulation of translation occurs early in Plk4-induced CA. CNOT9, an Unk interactor and component of the translational inhibitory CCR4-NOT complex, localizes to centrosomes at this time. In summary, centriolar satellites and Unk promote local translation as part of a translational program that ensures centriole duplication.
Collapse
Affiliation(s)
- Abraham Martinez
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Alexander J. Stemm-Wolf
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Ryan M. Sheridan
- RNA Bioscience Initiative (RBI), University of Colorado, Anschutz Medical Campus, Aurora CO 80045
| | - Matthew J. Taliaferro
- RNA Bioscience Initiative (RBI), University of Colorado, Anschutz Medical Campus, Aurora CO 80045
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Chad G. Pearson
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
21
|
Yang J, Zhang YN, Wang RX, Hao CZ, Qiu Y, Chi H, Luan WS, Tang H, Zhang XJ, Sun X, Sheps JA, Ling V, Cao M, Wang JS. ZFYVE19 deficiency: a ciliopathy involving failure of cell division, with cell death. J Med Genet 2024; 61:750-758. [PMID: 38816193 PMCID: PMC11287636 DOI: 10.1136/jmg-2023-109779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/04/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND AND AIMS Variants in ZFYVE19 underlie a disorder characterised by progressive portal fibrosis, portal hypertension and eventual liver decompensation. We aim to create an animal model to elucidate the pathogenic mechanism. METHODS Zfyve19 knockout (Zfyve19-/- ) mice were generated and exposed to different liver toxins. Their livers were characterised at the tissue, cellular and molecular levels. Findings were compared with those in wild-type mice and in ZFYVE19-deficient patients. ZFYVE19 knockout and knockdown retinal pigment epithelial-1 cells and mouse embryonic fibroblasts were generated to study cell division and cell death. RESULTS The Zfyve19-/- mice were normal overall, particularly with respect to hepatobiliary features. However, when challenged with α-naphthyl isothiocyanate, Zfyve19-/- mice developed changes resembling those in ZFYVE19-deficient patients, including elevated serum liver injury markers, increased numbers of bile duct profiles with abnormal cholangiocyte polarity and biliary fibrosis. Failure of cell division, centriole and cilia abnormalities, and increased cell death were observed in knockdown/knockout cells. Increased cell death and altered mRNA expression of cell death-related signalling pathways was demonstrated in livers from Zfyve19-/- mice and patients. Transforming growth factor-β (TGF-β) and Janus kinase-Signal Transducer and Activator of Transcription 3 (JAK-STAT3) signalling pathways were upregulated in vivo, as were chemokines such as C-X-C motif ligands 1, 10 and 12. CONCLUSIONS Our findings demonstrated that ZFYVE19 deficiency is a ciliopathy with novel histological features. Failure of cell division with ciliary abnormalities and cell death activates macrophages and may thus lead to biliary fibrosis via TGF-β pathway in the disease.
Collapse
Affiliation(s)
- Jing Yang
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
- The Center for Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
- Department of Pediatric Gastroenterology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ya-Nan Zhang
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
- The Center for Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Ren-Xue Wang
- BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Chen-Zhi Hao
- The Center for Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Yiling Qiu
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
- The Center for Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Hao Chi
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
| | - Wei-Sha Luan
- Department of Pediatrics, Jinshan Hospital of Fudan University, Shanghai, China
| | - HongYi Tang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiu-Juan Zhang
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - XuXu Sun
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Victor Ling
- BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Muqing Cao
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-She Wang
- The Center for Liver Diseases, Children's Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Birth Defect, Shanghai, China
| |
Collapse
|
22
|
Meyer-Gerards C, Bazzi H. Developmental and tissue-specific roles of mammalian centrosomes. FEBS J 2024. [PMID: 38935637 DOI: 10.1111/febs.17212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/08/2024] [Accepted: 06/14/2024] [Indexed: 06/29/2024]
Abstract
Centrosomes are dominant microtubule organizing centers in animal cells with a pair of centrioles at their core. They template cilia during interphase and help organize the mitotic spindle for a more efficient cell division. Here, we review the roles of centrosomes in the early developing mouse and during organ formation. Mammalian cells respond to centrosome loss-of-function by activating the mitotic surveillance pathway, a timing mechanism that, when a defined mitotic duration is exceeded, leads to p53-dependent cell death in the descendants. Mouse embryos without centrioles are highly susceptible to this pathway and undergo embryonic arrest at mid-gestation. The complete loss of the centriolar core results in earlier and more severe phenotypes than that of other centrosomal proteins. Finally, different developing tissues possess varying thresholds and mount graded responses to the loss of centrioles that go beyond the germ layer of origin.
Collapse
Affiliation(s)
- Charlotte Meyer-Gerards
- Department of Cell Biology of the Skin, Medical Faculty, University of Cologne, Germany
- Department of Dermatology and Venereology, Medical Faculty, University of Cologne, Germany
- The Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Medical Faculty, University of Cologne, Germany
- Graduate School for Biological Sciences, University of Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Germany
| | - Hisham Bazzi
- Department of Cell Biology of the Skin, Medical Faculty, University of Cologne, Germany
- Department of Dermatology and Venereology, Medical Faculty, University of Cologne, Germany
- The Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Medical Faculty, University of Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Germany
| |
Collapse
|
23
|
Yeh HW, Chen PP, Yeh TC, Lin SL, Chen YT, Lin WP, Chen T, Pang JM, Lin KT, Wang LHC, Lin YC, Shih O, Jeng US, Hsia KC, Cheng HC. Cep57 regulates human centrosomes through multivalent interactions. Proc Natl Acad Sci U S A 2024; 121:e2305260121. [PMID: 38857398 PMCID: PMC11194501 DOI: 10.1073/pnas.2305260121] [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: 04/04/2023] [Accepted: 04/15/2024] [Indexed: 06/12/2024] Open
Abstract
Human Cep57 is a coiled-coil scaffold at the pericentriolar matrix (PCM), controlling centriole duplication and centrosome maturation for faithful cell division. Genetic truncation mutations of Cep57 are associated with the mosaic-variegated aneuploidy (MVA) syndrome. During interphase, Cep57 forms a complex with Cep63 and Cep152, serving as regulators for centrosome maturation. However, the molecular interplay of Cep57 with these essential scaffolding proteins remains unclear. Here, we demonstrate that Cep57 undergoes liquid-liquid phase separation (LLPS) driven by three critical domains (NTD, CTD, and polybasic LMN). In vitro Cep57 condensates catalyze microtubule nucleation via the LMN motif-mediated tubulin concentration. In cells, the LMN motif is required for centrosomal microtubule aster formation. Moreover, Cep63 restricts Cep57 assembly, expansion, and microtubule polymerization activity. Overexpression of competitive constructs for multivalent interactions, including an MVA mutation, leads to excessive centrosome duplication. In Cep57-depleted cells, self-assembly mutants failed to rescue centriole disengagement and PCM disorganization. Thus, Cep57's multivalent interactions are pivotal for maintaining the accurate structural and functional integrity of human centrosomes.
Collapse
Affiliation(s)
- Hung-Wei Yeh
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Po-Pang Chen
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Tzu-Chen Yeh
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Shiou-Lan Lin
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Yue-Ting Chen
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Wan-Ping Lin
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Ting Chen
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Jia Meng Pang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Kai-Ti Lin
- Institute of Biotechnology, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Lily Hui-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Orion Shih
- National Synchrotron Radiation Research Center, Hsinchu30076, Taiwan
| | - U-Ser Jeng
- National Synchrotron Radiation Research Center, Hsinchu30076, Taiwan
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu30013, Taiwan
| | - Kuo-Chiang Hsia
- Institute of Molecular Biology, Academia Sinica, Taipei11529, Taiwan
| | - Hui-Chun Cheng
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu30013, Taiwan
| |
Collapse
|
24
|
Sladky VC, Strong MA, Tapias-Gomez D, Jewett CE, Drown CG, Scott PM, Holland AJ. The AID2 system offers a potent tool for rapid, reversible, or sustained degradation of essential proteins in live mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597287. [PMID: 38895390 PMCID: PMC11185741 DOI: 10.1101/2024.06.04.597287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Studying essential genes required for dynamic processes in live mice is challenging as genetic perturbations are irreversible and limited by slow protein depletion kinetics. The first-generation auxin-inducible-degron (AID) system is a powerful tool for analyzing inducible protein loss in cultured cells. However, auxin administration is toxic to mice, preventing its long-term use in animals. Here, we use an optimized second-generation AID system to achieve the conditional and reversible loss of the essential centrosomal protein CEP192 in live mice. We show that the auxin derivative 5-Ph-IAA is well tolerated over two weeks and drives near-complete CEP192-mAID degradation in less than one hour in vivo. Prolonged CEP192 loss led to cell division failure and cell death in proliferative tissues. Thus, the second-generation AID system is well suited for rapid and/or sustained protein depletion in live mice, offering a valuable new tool for interrogating protein function in vivo.
Collapse
Affiliation(s)
- Valentina C Sladky
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Margaret A Strong
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Daniel Tapias-Gomez
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Cayla E Jewett
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Chelsea G Drown
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Phillip M Scott
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 21205, MD, Baltimore, USA
| |
Collapse
|
25
|
Hawkins LM, Wang C, Chaput D, Batra M, Marsilia C, Awshah D, Suvorova ES. The Crk4-Cyc4 complex regulates G 2/M transition in Toxoplasma gondii. EMBO J 2024; 43:2094-2126. [PMID: 38600241 PMCID: PMC11148040 DOI: 10.1038/s44318-024-00095-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/12/2024] Open
Abstract
A versatile division of apicomplexan parasites and a dearth of conserved regulators have hindered the progress of apicomplexan cell cycle studies. While most apicomplexans divide in a multinuclear fashion, Toxoplasma gondii tachyzoites divide in the traditional binary mode. We previously identified five Toxoplasma CDK-related kinases (Crk). Here, we investigated TgCrk4 and its cyclin partner TgCyc4. We demonstrated that TgCrk4 regulates conventional G2 phase processes, such as repression of chromosome rereplication and centrosome reduplication, and acts upstream of the spindle assembly checkpoint. The spatial TgCyc4 dynamics supported the TgCrk4-TgCyc4 complex role in the coordination of chromosome and centrosome cycles. We also identified a dominant TgCrk4-TgCyc4 complex interactor, TgiRD1 protein, related to DNA replication licensing factor CDT1 but played no role in licensing DNA replication in the G1 phase. Our results showed that TgiRD1 also plays a role in controlling chromosome and centrosome reduplication. Global phosphoproteome analyses identified TgCrk4 substrates, including TgORC4, TgCdc20, TgGCP2, and TgPP2ACA. Importantly, the phylogenetic and structural studies suggest the Crk4-Cyc4 complex is limited to a minor group of the binary dividing apicomplexans.
Collapse
Affiliation(s)
- Lauren M Hawkins
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Chengqi Wang
- College of Public Health, University of South Florida, Tampa, FL, 33612, USA
| | - Dale Chaput
- Proteomics Core, College of Arts and Sciences, University of South Florida, Tampa, FL, 33612, USA
| | - Mrinalini Batra
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Clem Marsilia
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Danya Awshah
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Elena S Suvorova
- Division of Infectious Diseases, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.
| |
Collapse
|
26
|
Grossmann J, Kratz AS, Kordonsky A, Prag G, Hoffmann I. CRL4 DCAF1 ubiquitin ligase regulates PLK4 protein levels to prevent premature centriole duplication. Life Sci Alliance 2024; 7:e202402668. [PMID: 38490717 PMCID: PMC10942865 DOI: 10.26508/lsa.202402668] [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: 02/21/2024] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024] Open
Abstract
Centrioles play important roles in the assembly of centrosomes and cilia. Centriole duplication occurs once per cell cycle and is dependent on polo-like kinase 4 (PLK4). To prevent centriole amplification, which is a hallmark of cancer, PLK4 protein levels need to be tightly regulated. Here, we show that the Cullin4A/B-DDB1-DCAF1, CRL4DCAF1, E3 ligase targets PLK4 for degradation in human cells. DCAF1 binds and ubiquitylates PLK4 in the G2 phase to prevent premature centriole duplication in mitosis. In contrast to the regulation of PLK4 by SCFβ-TrCP, the interaction between PLK4 and DCAF1 is independent of PLK4 kinase activity and mediated by polo-boxes 1 and 2 of PLK4, suggesting that DCAF1 promotes PLK4 ubiquitylation independently of β-TrCP. Thus, the SCFSlimb/β-TrCP pathway, targeting PLK4 for ubiquitylation based on its phosphorylation state and CRL4DCAF1, which ubiquitylates PLK4 by binding to the conserved PB1-PB2 domain, appear to be complementary ways to control PLK4 abundance to prevent centriole overduplication.
Collapse
Affiliation(s)
- Josina Grossmann
- Cell Cycle Control and Carcinogenesis, German Cancer Research Center, DKFZ, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Anne-Sophie Kratz
- Cell Cycle Control and Carcinogenesis, German Cancer Research Center, DKFZ, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Alina Kordonsky
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Gali Prag
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ingrid Hoffmann
- Cell Cycle Control and Carcinogenesis, German Cancer Research Center, DKFZ, Heidelberg, Germany
| |
Collapse
|
27
|
Laporte MH, Gambarotto D, Bertiaux É, Bournonville L, Louvel V, Nunes JM, Borgers S, Hamel V, Guichard P. Time-series reconstruction of the molecular architecture of human centriole assembly. Cell 2024; 187:2158-2174.e19. [PMID: 38604175 PMCID: PMC11060037 DOI: 10.1016/j.cell.2024.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/21/2023] [Accepted: 03/19/2024] [Indexed: 04/13/2024]
Abstract
Centriole biogenesis, as in most organelle assemblies, involves the sequential recruitment of sub-structural elements that will support its function. To uncover this process, we correlated the spatial location of 24 centriolar proteins with structural features using expansion microscopy. A time-series reconstruction of protein distributions throughout human procentriole assembly unveiled the molecular architecture of the centriole biogenesis steps. We found that the process initiates with the formation of a naked cartwheel devoid of microtubules. Next, the bloom phase progresses with microtubule blade assembly, concomitantly with radial separation and rapid cartwheel growth. In the subsequent elongation phase, the tubulin backbone grows linearly with the recruitment of the A-C linker, followed by proteins of the inner scaffold (IS). By following six structural modules, we modeled 4D assembly of the human centriole. Collectively, this work provides a framework to investigate the spatial and temporal assembly of large macromolecules.
Collapse
Affiliation(s)
- Marine H Laporte
- University of Geneva, Department of Molecular and Cellular Biology, Faculty of Sciences, Geneva, Switzerland
| | - Davide Gambarotto
- University of Geneva, Department of Molecular and Cellular Biology, Faculty of Sciences, Geneva, Switzerland
| | - Éloïse Bertiaux
- University of Geneva, Department of Molecular and Cellular Biology, Faculty of Sciences, Geneva, Switzerland
| | - Lorène Bournonville
- University of Geneva, Department of Molecular and Cellular Biology, Faculty of Sciences, Geneva, Switzerland
| | - Vincent Louvel
- University of Geneva, Department of Molecular and Cellular Biology, Faculty of Sciences, Geneva, Switzerland
| | - José M Nunes
- University of Geneva, Department of Genetic and evolution, Faculty of Sciences, Geneva, Switzerland
| | - Susanne Borgers
- University of Geneva, Department of Molecular and Cellular Biology, Faculty of Sciences, Geneva, Switzerland
| | - Virginie Hamel
- University of Geneva, Department of Molecular and Cellular Biology, Faculty of Sciences, Geneva, Switzerland.
| | - Paul Guichard
- University of Geneva, Department of Molecular and Cellular Biology, Faculty of Sciences, Geneva, Switzerland.
| |
Collapse
|
28
|
Tollervey F, Rios MU, Zagoriy E, Woodruff JB, Mahamid J. Native molecular architectures of centrosomes in C. elegans embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587742. [PMID: 38617234 PMCID: PMC11014625 DOI: 10.1101/2024.04.03.587742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Centrosomes organize microtubules that are essential for mitotic divisions in animal cells. They consist of centrioles surrounded by Pericentriolar Material (PCM). Questions related to mechanisms of centriole assembly, PCM organization, and microtubule formation remain unanswered, in part due to limited availability of molecular-resolution structural analyses in situ. Here, we use cryo-electron tomography to visualize centrosomes across the cell cycle in cells isolated from C. elegans embryos. We describe a pseudo-timeline of centriole assembly and identify distinct structural features including a cartwheel in daughter centrioles, and incomplete microtubule doublets surrounded by a star-shaped density in mother centrioles. We find that centriole and PCM microtubules differ in protofilament number (13 versus 11) indicating distinct nucleation mechanisms. This difference could be explained by atypical γ-tubulin ring complexes with 11-fold symmetry identified at the minus ends of short PCM microtubules. We further characterize a porous and disordered network that forms the interconnected PCM. Thus, our work builds a three-dimensional structural atlas that helps explain how centrosomes assemble, grow, and achieve function.
Collapse
Affiliation(s)
- Fergus Tollervey
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
- Collaboration for Joint PhD Degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Manolo U. Rios
- Department of Cell Biology and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Evgenia Zagoriy
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Jeffrey B. Woodruff
- Department of Cell Biology and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Julia Mahamid
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
- Cell Biology and Biophysics Unit, EMBL, 69117 Heidelberg, Germany
| |
Collapse
|
29
|
Ganga AK, Sweeney LK, Ramos AR, Bishop CS, Hamel V, Guichard P, Breslow DK. A disease-associated PPP2R3C-MAP3K1 phospho-regulatory module controls centrosome function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.587836. [PMID: 38617270 PMCID: PMC11014585 DOI: 10.1101/2024.04.02.587836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Centrosomes have critical roles in microtubule organization and in cell signaling.1-8 However, the mechanisms that regulate centrosome function are not fully defined, and thus how defects in centrosomal regulation contribute to disease is incompletely understood. From functional genomic analyses, we find here that PPP2R3C, a PP2A phosphatase subunit, is a distal centriole protein and functional partner of centriolar proteins CEP350 and FOP. We further show that a key function of PPP2R3C is to counteract the kinase activity of MAP3K1. In support of this model, MAP3K1 knockout suppresses growth defects caused by PPP2R3C inactivation, and MAP3K1 and PPP2R3C have opposing effects on basal and microtubule stress-induced JNK signaling. Illustrating the importance of balanced MAP3K1 and PPP2R3C activities, acute overexpression of MAP3K1 severely inhibits centrosome function and triggers rapid centriole disintegration. Additionally, inactivating PPP2R3C mutations and activating MAP3K1 mutations both cause congenital syndromes characterized by gonadal dysgenesis.9-15 As a syndromic PPP2R3C variant is defective in centriolar localization and binding to centriolar protein FOP, we propose that imbalanced activity of this centrosomal kinase-phosphatase pair is the shared cause of these disorders. Thus, our findings reveal a new centrosomal phospho-regulatory module, shed light on disorders of gonadal development, and illustrate the power of systems genetics to identify previously unrecognized gene functions.
Collapse
Affiliation(s)
- Anil Kumar Ganga
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Lauren K. Sweeney
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Armando Rubio Ramos
- Department of Molecular and Cellular Biology, University of Geneva, Faculty of Sciences, Geneva, Switzerland
| | - Cassandra S. Bishop
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Virginie Hamel
- Department of Molecular and Cellular Biology, University of Geneva, Faculty of Sciences, Geneva, Switzerland
| | - Paul Guichard
- Department of Molecular and Cellular Biology, University of Geneva, Faculty of Sciences, Geneva, Switzerland
| | - David K. Breslow
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| |
Collapse
|
30
|
Hosea R, Hillary S, Naqvi S, Wu S, Kasim V. The two sides of chromosomal instability: drivers and brakes in cancer. Signal Transduct Target Ther 2024; 9:75. [PMID: 38553459 PMCID: PMC10980778 DOI: 10.1038/s41392-024-01767-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/18/2024] [Accepted: 02/06/2024] [Indexed: 04/02/2024] Open
Abstract
Chromosomal instability (CIN) is a hallmark of cancer and is associated with tumor cell malignancy. CIN triggers a chain reaction in cells leading to chromosomal abnormalities, including deviations from the normal chromosome number or structural changes in chromosomes. CIN arises from errors in DNA replication and chromosome segregation during cell division, leading to the formation of cells with abnormal number and/or structure of chromosomes. Errors in DNA replication result from abnormal replication licensing as well as replication stress, such as double-strand breaks and stalled replication forks; meanwhile, errors in chromosome segregation stem from defects in chromosome segregation machinery, including centrosome amplification, erroneous microtubule-kinetochore attachments, spindle assembly checkpoint, or defective sister chromatids cohesion. In normal cells, CIN is deleterious and is associated with DNA damage, proteotoxic stress, metabolic alteration, cell cycle arrest, and senescence. Paradoxically, despite these negative consequences, CIN is one of the hallmarks of cancer found in over 90% of solid tumors and in blood cancers. Furthermore, CIN could endow tumors with enhanced adaptation capabilities due to increased intratumor heterogeneity, thereby facilitating adaptive resistance to therapies; however, excessive CIN could induce tumor cells death, leading to the "just-right" model for CIN in tumors. Elucidating the complex nature of CIN is crucial for understanding the dynamics of tumorigenesis and for developing effective anti-tumor treatments. This review provides an overview of causes and consequences of CIN, as well as the paradox of CIN, a phenomenon that continues to perplex researchers. Finally, this review explores the potential of CIN-based anti-tumor therapy.
Collapse
Affiliation(s)
- Rendy Hosea
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Sharon Hillary
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Sumera Naqvi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China.
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China.
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
31
|
Athwal H, Kochiyanil A, Bhat V, Allan AL, Parsyan A. Centrosomes and associated proteins in pathogenesis and treatment of breast cancer. Front Oncol 2024; 14:1370565. [PMID: 38606093 PMCID: PMC11007099 DOI: 10.3389/fonc.2024.1370565] [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: 01/14/2024] [Accepted: 03/04/2024] [Indexed: 04/13/2024] Open
Abstract
Breast cancer is the most prevalent malignancy among women worldwide. Despite significant advances in treatment, it remains one of the leading causes of female mortality. The inability to effectively treat advanced and/or treatment-resistant breast cancer demonstrates the need to develop novel treatment strategies and targeted therapies. Centrosomes and their associated proteins have been shown to play key roles in the pathogenesis of breast cancer and thus represent promising targets for drug and biomarker development. Centrosomes are fundamental cellular structures in the mammalian cell that are responsible for error-free execution of cell division. Centrosome amplification and aberrant expression of its associated proteins such as Polo-like kinases (PLKs), Aurora kinases (AURKs) and Cyclin-dependent kinases (CDKs) have been observed in various cancers, including breast cancer. These aberrations in breast cancer are thought to cause improper chromosomal segregation during mitosis, leading to chromosomal instability and uncontrolled cell division, allowing cancer cells to acquire new genetic changes that result in evasion of cell death and the promotion of tumor formation. Various chemical compounds developed against PLKs and AURKs have shown meaningful antitumorigenic effects in breast cancer cells in vitro and in vivo. The mechanism of action of these inhibitors is likely related to exacerbation of numerical genomic instability, such as aneuploidy or polyploidy. Furthermore, growing evidence demonstrates enhanced antitumorigenic effects when inhibitors specific to centrosome-associated proteins are used in combination with either radiation or chemotherapy drugs in breast cancer. This review focuses on the current knowledge regarding the roles of centrosome and centrosome-associated proteins in breast cancer pathogenesis and their utility as novel targets for breast cancer treatment.
Collapse
Affiliation(s)
- Harjot Athwal
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Arpitha Kochiyanil
- Faculty of Science, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Vasudeva Bhat
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
| | - Alison L. Allan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Armen Parsyan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- London Regional Cancer Program, London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Division of General Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Surgery, St. Joseph’s Health Care London and London Health Sciences Centre, London, ON, Canada
| |
Collapse
|
32
|
Englisch AS, Hofbrucker-MacKenzie SA, Izadi-Seitz M, Kessels MM, Qualmann B. Ankrd26 is a retinoic acid-responsive plasma membrane-binding and -shaping protein critical for proper cell differentiation. Cell Rep 2024; 43:113939. [PMID: 38493476 DOI: 10.1016/j.celrep.2024.113939] [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: 07/26/2023] [Revised: 01/17/2024] [Accepted: 02/23/2024] [Indexed: 03/19/2024] Open
Abstract
Morphogens are important triggers for differentiation processes. Yet, downstream effectors that organize cell shape changes in response to morphogenic cues, such as retinoic acid, largely remain elusive. Additionally, derailed plasma membrane-derived signaling often is associated with cancer. We identify Ankrd26 as a critical player in cellular differentiation and as plasma membrane-localized protein able to self-associate and form clusters at the plasma membrane in response to retinoic acid. We show that Ankrd26 uses an N-terminal amphipathic structure for membrane binding and bending. Importantly, in an acute myeloid leukemia-associated Ankrd26 mutant, this critical structure was absent, and Ankrd26's membrane association and shaping abilities were impaired. In line with this, the mutation rendered Ankrd26 inactive in both gain-of-function and loss-of-function/rescue studies addressing retinoic acid/brain-derived neurotrophic factor (BDNF)-induced neuroblastoma differentiation. Our results highlight the importance and molecular details of Ankrd26-mediated organizational platforms for cellular differentiation at the plasma membrane and how impairment of these platforms leads to cancer-associated pathomechanisms involving these Ankrd26 properties.
Collapse
Affiliation(s)
- Anna Sofie Englisch
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Nonnenplan 2-4, 07743 Jena, Germany
| | - Sarah Ann Hofbrucker-MacKenzie
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Nonnenplan 2-4, 07743 Jena, Germany
| | - Maryam Izadi-Seitz
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Nonnenplan 2-4, 07743 Jena, Germany
| | - Michael Manfred Kessels
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Nonnenplan 2-4, 07743 Jena, Germany.
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Nonnenplan 2-4, 07743 Jena, Germany.
| |
Collapse
|
33
|
Takeda Y, Chinen T, Honda S, Takatori S, Okuda S, Yamamoto S, Fukuyama M, Takeuchi K, Tomita T, Hata S, Kitagawa D. Molecular basis promoting centriole triplet microtubule assembly. Nat Commun 2024; 15:2216. [PMID: 38519454 PMCID: PMC10960023 DOI: 10.1038/s41467-024-46454-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 02/28/2024] [Indexed: 03/25/2024] Open
Abstract
The triplet microtubule, a core structure of centrioles crucial for the organization of centrosomes, cilia, and flagella, consists of unclosed incomplete microtubules. The mechanisms of its assembly represent a fundamental open question in biology. Here, we discover that the ciliopathy protein HYLS1 and the β-tubulin isotype TUBB promote centriole triplet microtubule assembly. HYLS1 or a C-terminal tail truncated version of TUBB generates tubulin-based superstructures composed of centriole-like incomplete microtubule chains when overexpressed in human cells. AlphaFold-based structural models and mutagenesis analyses further suggest that the ciliopathy-related residue D211 of HYLS1 physically traps the wobbling C-terminal tail of TUBB, thereby suppressing its inhibitory role in the initiation of the incomplete microtubule assembly. Overall, our findings provide molecular insights into the biogenesis of atypical microtubule architectures conserved for over a billion years.
Collapse
Affiliation(s)
- Yutaka Takeda
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Takumi Chinen
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan.
| | - Shunnosuke Honda
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Sho Takatori
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Shotaro Okuda
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Shohei Yamamoto
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Masamitsu Fukuyama
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Koh Takeuchi
- Laboratory of Physical Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan
| | - Shoji Hata
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan.
| | - Daiju Kitagawa
- Laboratory of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
34
|
Curinha A, Huang Z, Anglen T, Strong MA, Gliech CR, Jewett CE, Friskes A, Holland AJ. Centriole structural integrity defects are a crucial feature of Hydrolethalus Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.06.583733. [PMID: 38496445 PMCID: PMC10942441 DOI: 10.1101/2024.03.06.583733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Hydrolethalus Syndrome (HLS) is a lethal, autosomal recessive ciliopathy caused by the mutation of the conserved centriole protein HYLS1. However, how HYLS1 facilitates the centriole-based templating of cilia is poorly understood. Here, we show that mice harboring the HYLS1 disease mutation die shortly after birth and exhibit developmental defects that recapitulate several manifestations of the human disease. These phenotypes arise from tissue-specific defects in cilia assembly and function caused by a loss of centriole integrity. We show that HYLS1 is recruited to the centriole by CEP120 and functions to recruit centriole inner scaffold proteins that stabilize the centriolar microtubule wall. The HLS mutation disrupts the interaction of HYLS1 with CEP120 leading to HYLS1 displacement and degeneration of the centriole distal end. We propose that tissue-specific defects in centriole integrity caused by the HYLS1 mutation prevent ciliogenesis and drive HLS phenotypes.
Collapse
Affiliation(s)
- Ana Curinha
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhaoyu Huang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Taylor Anglen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Margaret A Strong
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Colin R Gliech
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cayla E Jewett
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anoek Friskes
- Division of Cell Biology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
35
|
Park JE, Kim TS, Zeng Y, Mikolaj M, Il Ahn J, Alam MS, Monnie CM, Shi V, Zhou M, Chun TW, Maldarelli F, Narayan K, Ahn J, Ashwell JD, Strebel K, Lee KS. Centrosome amplification and aneuploidy driven by the HIV-1-induced Vpr•VprBP•Plk4 complex in CD4 + T cells. Nat Commun 2024; 15:2017. [PMID: 38443376 PMCID: PMC10914751 DOI: 10.1038/s41467-024-46306-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 02/14/2024] [Indexed: 03/07/2024] Open
Abstract
HIV-1 infection elevates the risk of developing various cancers, including T-cell lymphoma. Whether HIV-1-encoded proteins directly contribute to oncogenesis remains unknown. We observe that approximately 1-5% of CD4+ T cells from the blood of people living with HIV-1 exhibit over-duplicated centrioles, suggesting that centrosome amplification underlies the development of HIV-1-associated cancers by driving aneuploidy. Through affinity purification, biochemical, and cellular analyses, we discover that Vpr, an accessory protein of HIV-1, hijacks the centriole duplication machinery and induces centrosome amplification and aneuploidy. Mechanistically, Vpr forms a cooperative ternary complex with an E3 ligase subunit, VprBP, and polo-like kinase 4 (Plk4). Unexpectedly, however, the complex enhances Plk4's functionality by promoting its relocalization to the procentriole assembly and induces centrosome amplification. Loss of either Vpr's C-terminal 17 residues or VprBP acidic region, the two elements required for binding to Plk4 cryptic polo-box, abrogates Vpr's capacity to induce these events. Furthermore, HIV-1 WT, but not its Vpr mutant, induces multiple centrosomes and aneuploidy in human primary CD4+ T cells. We propose that the Vpr•VprBP•Plk4 complex serves as a molecular link that connects HIV-1 infection to oncogenesis and that inhibiting the Vpr C-terminal motif may reduce the occurrence of HIV-1-associated cancers.
Collapse
Affiliation(s)
- Jung-Eun Park
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tae-Sung Kim
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yan Zeng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Melissa Mikolaj
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Jong Il Ahn
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Muhammad S Alam
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Christina M Monnie
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA
| | - Victoria Shi
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ming Zhou
- Protein Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Jinwoo Ahn
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA
| | - Jonathan D Ashwell
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Klaus Strebel
- Laboratory of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kyung S Lee
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
36
|
Simerly C, Robertson E, Harrison C, Ward S, George C, Deleon J, Hartnett C, Schatten G. Male meiotic spindle poles are stabilized by TACC3 and cKAP5/chTOG differently from female meiotic or somatic mitotic spindles in mice. Sci Rep 2024; 14:4808. [PMID: 38413710 PMCID: PMC10899211 DOI: 10.1038/s41598-024-55376-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/22/2024] [Indexed: 02/29/2024] Open
Abstract
Transforming acidic acid coiled-coil protein 3 (TACC3) and cytoskeleton associated protein 5 (cKAP5; or colonic hepatic tumor overexpressed gene, chTOG) are vital for spindle assembly and stabilization initiated through TACC3 Aurora-A kinase interaction. Here, TACC3 and cKAP5/chTOG localization with monospecific antibodies is investigated in eGFP-centrin-2- expressing mouse meiotic spermatocytes. Both proteins bind spermatocyte spindle poles but neither kinetochore nor interpolar microtubules, unlike in mitotic mouse fibroblasts or female meiotic oocyte spindles. Spermatocytes do not display a liquid-like spindle domain (LISD), although fusing them into maturing oocytes generates LISD-like TACC3 condensates around sperm chromatin but sparse microtubule assembly. Microtubule inhibitors do not reduce TACC3 and cKAP5/chTOG spindle pole binding. MLN 8237 Aurora-A kinase inhibitor removes TACC3, not cKAP5/chTOG, disrupting spindle organization, chromosome alignment, and impacting spindle pole γ-tubulin intensity. The LISD disruptor 1,6-hexanediol abolished TACC3 in spermatocytes, impacting spindle bipolarity and chromosome organization. Cold microtubule disassembly and rescue experiments in the presence of 1,6-hexanediol reinforce the concept that spermatocyte TACC3 spindle pole presence is not required for spindle pole microtubule assembly. Collectively, meiotic spermatocytes without a LISD localize TACC3 and cKAP5/chTOG exclusively at spindle poles to support meiotic spindle pole stabilization during male meiosis, different from either female meiosis or mitosis.
Collapse
Affiliation(s)
- Calvin Simerly
- Departments of Cell Biology, Ob-Gyn-Repro Sci, and Bioengineering, Pittsburgh Development Center of Magee-Womens Research Institute, University of Pittsburgh Medical Center, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Emily Robertson
- Departments of Cell Biology, Ob-Gyn-Repro Sci, and Bioengineering, Pittsburgh Development Center of Magee-Womens Research Institute, University of Pittsburgh Medical Center, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Caleb Harrison
- Departments of Cell Biology, Ob-Gyn-Repro Sci, and Bioengineering, Pittsburgh Development Center of Magee-Womens Research Institute, University of Pittsburgh Medical Center, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Sydney Ward
- Departments of Cell Biology, Ob-Gyn-Repro Sci, and Bioengineering, Pittsburgh Development Center of Magee-Womens Research Institute, University of Pittsburgh Medical Center, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Charlize George
- Departments of Cell Biology, Ob-Gyn-Repro Sci, and Bioengineering, Pittsburgh Development Center of Magee-Womens Research Institute, University of Pittsburgh Medical Center, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Jasmine Deleon
- Departments of Cell Biology, Ob-Gyn-Repro Sci, and Bioengineering, Pittsburgh Development Center of Magee-Womens Research Institute, University of Pittsburgh Medical Center, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Carrie Hartnett
- Departments of Cell Biology, Ob-Gyn-Repro Sci, and Bioengineering, Pittsburgh Development Center of Magee-Womens Research Institute, University of Pittsburgh Medical Center, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Gerald Schatten
- Departments of Cell Biology, Ob-Gyn-Repro Sci, and Bioengineering, Pittsburgh Development Center of Magee-Womens Research Institute, University of Pittsburgh Medical Center, 204 Craft Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
37
|
Wang T, Fan G, Xia Y, Zou Y, Liu Y, Wang J, Hu Y, Teng J, Huang N, Chen J. Dual roles of CCDC102A in governing centrosome duplication and cohesion. Cell Rep 2024; 43:113696. [PMID: 38280197 DOI: 10.1016/j.celrep.2024.113696] [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: 06/13/2023] [Revised: 12/03/2023] [Accepted: 01/05/2024] [Indexed: 01/29/2024] Open
Abstract
In animal cells, the dysregulation of centrosome duplication and cohesion maintenance leads to abnormal spindle assembly and chromosomal instability, contributing to developmental disorders and tumorigenesis. However, the molecular mechanisms involved in maintaining accurate centrosome number control and tethering are not fully understood. Here, we identified coiled-coil domain-containing 102A (CCDC102A) as a centrosomal protein exhibiting a barrel-like structure in the proximal regions of parent centrioles, where it prevents centrosome overduplication by restricting interactions between Cep192 and Cep152 on centrosomes, thereby ensuring bipolar spindle formation. Additionally, CCDC102A regulates the centrosome linker by recruiting and binding C-Nap1; it is removed from the centrosome after Nek2A-mediated phosphorylation at the onset of mitosis. Overall, our results indicate that CCDC102A participates in controlling centrosome number and maintaining centrosome cohesion, suggesting that a well-tuned system regulates centrosome structure and function throughout the cell cycle.
Collapse
Affiliation(s)
- Tianning Wang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing 100871, China; Breast Disease Diagnosis and Treatment Center/Department of Thyroid Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China; Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, China
| | - Guiliang Fan
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yuqing Xia
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yuhong Zou
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yunjie Liu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing 100871, China
| | - Jiaxin Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; Institute of Neuroscience, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yingchun Hu
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing 100871, China
| | - Junlin Teng
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing 100871, China.
| | - Ning Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China; Institute of Neuroscience, Translational Medicine Institute, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Jianguo Chen
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing 100871, China; Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.
| |
Collapse
|
38
|
Grzonka M, Bazzi H. Mouse SAS-6 is required for centriole formation in embryos and integrity in embryonic stem cells. eLife 2024; 13:e94694. [PMID: 38407237 PMCID: PMC10917421 DOI: 10.7554/elife.94694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 12/12/2023] [Indexed: 02/27/2024] Open
Abstract
SAS-6 (SASS6) is essential for centriole formation in human cells and other organisms but its functions in the mouse are unclear. Here, we report that Sass6-mutant mouse embryos lack centrioles, activate the mitotic surveillance cell death pathway, and arrest at mid-gestation. In contrast, SAS-6 is not required for centriole formation in mouse embryonic stem cells (mESCs), but is essential to maintain centriole architecture. Of note, centrioles appeared after just one day of culture of Sass6-mutant blastocysts, from which mESCs are derived. Conversely, the number of cells with centrosomes is drastically decreased upon the exit from a mESC pluripotent state. At the mechanistic level, the activity of the master kinase in centriole formation, PLK4, associated with increased centriolar and centrosomal protein levels, endow mESCs with the robustness in using a SAS-6-independent centriole-biogenesis pathway. Collectively, our data suggest a differential requirement for mouse SAS-6 in centriole formation or integrity depending on PLK4 activity and centrosome composition.
Collapse
Affiliation(s)
- Marta Grzonka
- Department of Cell Biology of the Skin and Department of Dermatology and Venereology, Medical Faculty, University of CologneCologneGermany
- The Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Medical Faculty, University of CologneCologneGermany
- Graduate School for Biological Sciences, University of CologneCologneGermany
| | - Hisham Bazzi
- Department of Cell Biology of the Skin and Department of Dermatology and Venereology, Medical Faculty, University of CologneCologneGermany
- The Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Medical Faculty, University of CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of CologneCologneGermany
| |
Collapse
|
39
|
Cheng T, Mariappan A, Langner E, Shim K, Gopalakrishnan J, Mahjoub MR. Inhibiting centrosome clustering reduces cystogenesis and improves kidney function in autosomal dominant polycystic kidney disease. JCI Insight 2024; 9:e172047. [PMID: 38385746 DOI: 10.1172/jci.insight.172047] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a monogenic disorder accounting for approximately 5% of patients with renal failure, yet therapeutics for the treatment of ADPKD remain limited. ADPKD tissues display abnormalities in the biogenesis of the centrosome, a defect that can cause genome instability, aberrant ciliary signaling, and secretion of pro-inflammatory factors. Cystic cells form excess centrosomes via a process termed centrosome amplification (CA), which causes abnormal multipolar spindle configurations, mitotic catastrophe, and reduced cell viability. However, cells with CA can suppress multipolarity via "centrosome clustering," a key mechanism by which cells circumvent apoptosis. Here, we demonstrate that inhibiting centrosome clustering can counteract the proliferation of renal cystic cells with high incidences of CA. Using ADPKD human cells and mouse models, we show that preventing centrosome clustering with 2 inhibitors, CCB02 and PJ34, blocks cyst initiation and growth in vitro and in vivo. Inhibiting centrosome clustering activates a p53-mediated surveillance mechanism leading to apoptosis, reduced cyst expansion, decreased interstitial fibrosis, and improved kidney function. Transcriptional analysis of kidneys from treated mice identified pro-inflammatory signaling pathways implicated in CA-mediated cystogenesis and fibrosis. Our results demonstrate that centrosome clustering is a cyst-selective target for the improvement of renal morphology and function in ADPKD.
Collapse
Affiliation(s)
- Tao Cheng
- Department of Medicine, Nephrology Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Aruljothi Mariappan
- Institute of Human Genetics, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Ewa Langner
- Department of Medicine, Nephrology Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kyuhwan Shim
- Department of Medicine, Nephrology Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jay Gopalakrishnan
- Institute of Human Genetics, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Jena, Jena, Germany
| | - Moe R Mahjoub
- Department of Medicine, Nephrology Division, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
40
|
Zhou Y, Yang J, Huang L, Liu C, Yu M, Chen R, Zhou Q. Nudt21-mediated alternative polyadenylation of MZT1 3'UTR contributes to pancreatic cancer progression. iScience 2024; 27:108822. [PMID: 38303721 PMCID: PMC10831950 DOI: 10.1016/j.isci.2024.108822] [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: 07/18/2023] [Revised: 11/26/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024] Open
Abstract
Alternative polyadenylation (APA) is an important post-transcriptional regulatory mechanism and is involved in many diseases, but its function and mechanism in regulating pancreatic cancer (PC) pathogenesis remain unclear. In this study, we found that the 3' UTR shortening of MZT1 was the most prominent APA event in PC liver metastases. The short-3'UTR isoform exerted a stronger effect in promoting cell proliferation and migration both in vitro and in vivo. NUDT21, a core cleavage factor involved in APA, promoted the usage of proximal polyadenylation sites (PASs) on MZT1 mRNA by binding to the UGUA element located upstream of the proximal PAS. High percentage of distal polyA site usage index of MZT1 was significantly associated with a better prognosis. These findings demonstrate a crucial mechanism that NUDT21-mediated APA of MZT1 could promote the progression of PC. Our findings provided a better understanding of the connection between PC progression and APA machinery.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jiabin Yang
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Leyi Huang
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Chao Liu
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Min Yu
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Rufu Chen
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Quanbo Zhou
- Department of Pancreatic Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Department of Pancreatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
41
|
Lei Q, Yu Q, Yang N, Xiao Z, Song C, Zhang R, Yang S, Liu Z, Deng H. Therapeutic potential of targeting polo-like kinase 4. Eur J Med Chem 2024; 265:116115. [PMID: 38199166 DOI: 10.1016/j.ejmech.2023.116115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
Polo-like kinase 4 (PLK4), a highly conserved serine/threonine kinase, masterfully regulates centriole duplication in a spatiotemporal manner to ensure the fidelity of centrosome duplication and proper mitosis. Abnormal expression of PLK4 contributes to genomic instability and associates with a poor prognosis in cancer. Inhibition of PLK4 is demonstrated to exhibit significant efficacy against various types of human cancers, further highlighting its potential as a promising therapeutic target for cancer treatment. As such, numerous small-molecule inhibitors with distinct chemical scaffolds targeting PLK4 have been extensively investigated for the treatment of different human cancers, with several undergoing clinical evaluation (e.g., CFI-400945). Here, we review the structure, distribution, and biological functions of PLK4, encapsulate its intricate regulatory mechanisms of expression, and highlighting its multifaceted roles in cancer development and metastasis. Moreover, the recent advancements of PLK4 inhibitors in patent or literature are summarized, and their therapeutic potential as monotherapies or combination therapies with other anticancer agents are also discussed.
Collapse
Affiliation(s)
- Qian Lei
- Department of Respiratory and Critical Care Medicine, West China Hospital and Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Quanwei Yu
- Department of Respiratory and Critical Care Medicine, West China Hospital and Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Na Yang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhaolin Xiao
- Department of Respiratory and Critical Care Medicine, West China Hospital and Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chao Song
- Department of Respiratory and Critical Care Medicine, West China Hospital and Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rui Zhang
- Department of Pharmacy, Guizhou Provincial People's Hospital, Guizhou, Guiyang, 550002, China
| | - Shuxin Yang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zhihao Liu
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Hui Deng
- Department of Respiratory and Critical Care Medicine, West China Hospital and Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
42
|
Liu Y, He M, Ke X, Chen Y, Zhu J, Tan Z, Chen J. Centrosome amplification-related signature correlated with immune microenvironment and treatment response predicts prognosis and improves diagnosis of hepatocellular carcinoma by integrating machine learning and single-cell analyses. Hepatol Int 2024; 18:108-130. [PMID: 37154991 DOI: 10.1007/s12072-023-10538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/08/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND Centrosome amplification is a well-recognized oncogenic driver of tumor initiation and progression across a variety of malignancies and has been linked with tumor aggressiveness, metastasis, and adverse prognosis. Nevertheless, the significance of centrosome amplification in HCC is not well understood. METHODS The TCGA dataset was downloaded for centrosome amplification-related signature construction using the LASSO-penalized Cox regression algorithm, while the ICGC dataset was obtained for signature validation. Single-cell RNA sequencing from GSE149614 was analyzed to profile gene expression and the liver tumor niche. RESULTS A total of 134 centrosome amplification-related prognostic genes in HCC were detected and 6 key prognostic genes (SSX2IP, SPAG4, SAC3D1, NPM1, CSNK1D, and CEP55) among them were screened out to construct a signature with both high sensitivity and specificity in diagnosis and prognosis of HCC patients. The signature, as an independent factor, was associated with frequent recurrences, high mortality rates, advanced clinicopathologic features, and high vascular invasions. Moreover, the signature was intimately associated with cell cycle-related pathways and TP53 mutation profile, suggesting its underlying role in accelerating cell cycle progression and leading to liver cancer development. Meanwhile, the signature was also closely correlated with immunosuppressive cell infiltration and immune checkpoint expression, making it a vital immunosuppressive factor in the tumor microenvironment. Upon single-cell RNA sequencing, SSX2IP and SAC3D1 were found to be specially expressed in liver cancer stem-like cells, where they promoted cell cycle progression and hypoxia. CONCLUSIONS This study provided a direct molecular link of centrosome amplification with clinical characteristics, tumor microenvironment, and clinical drug-response, highlighting the critical role of centrosome amplification in liver cancer development and therapy resistance, thereby providing valuable insights into prognostic prediction and therapeutic response of HCC.
Collapse
Affiliation(s)
- Yanli Liu
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy & Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology & Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
- Central Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Min He
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy & Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology & Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
- Central Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Xinrong Ke
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy & Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology & Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
- Central Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Yuting Chen
- State Key Laboratory of Respiratory Disease, The Second Clinical Medical School, Guangzhou Medical University, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Jie Zhu
- State Key Laboratory of Respiratory Disease, The Second Clinical Medical School, Guangzhou Medical University, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Ziqing Tan
- State Key Laboratory of Respiratory Disease, The Second Clinical Medical School, Guangzhou Medical University, Guangzhou, 510180, Guangdong, People's Republic of China
| | - Jingqi Chen
- Guangzhou Key Laboratory for Research and Development of Nano-Biomedical Technology for Diagnosis and Therapy & Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Oncology & Translational Medicine Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China.
| |
Collapse
|
43
|
Langner E, Cheng T, Kefaloyianni E, Gluck C, Wang B, Mahjoub MR. Cep120 is essential for kidney stromal progenitor cell growth and differentiation. EMBO Rep 2024; 25:428-454. [PMID: 38177914 PMCID: PMC10897188 DOI: 10.1038/s44319-023-00019-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 01/06/2024] Open
Abstract
Mutations in genes that disrupt centrosome structure or function can cause congenital kidney developmental defects and lead to fibrocystic pathologies. Yet, it is unclear how defective centrosome biogenesis impacts renal progenitor cell physiology. Here, we examined the consequences of impaired centrosome duplication on kidney stromal progenitor cell growth, differentiation, and fate. Conditional deletion of the ciliopathy gene Cep120, which is essential for centrosome duplication, in the stromal mesenchyme resulted in reduced abundance of interstitial lineages including pericytes, fibroblasts and mesangial cells. These phenotypes were caused by a combination of delayed mitosis, activation of the mitotic surveillance pathway leading to apoptosis, and changes in both Wnt and Hedgehog signaling that are key for differentiation of stromal cells. Cep120 ablation resulted in small hypoplastic kidneys with medullary atrophy and delayed nephron maturation. Finally, Cep120 and centrosome loss in the interstitium sensitized kidneys of adult mice, causing rapid fibrosis after renal injury via enhanced TGF-β/Smad3-Gli2 signaling. Our study defines the cellular and developmental defects caused by loss of Cep120 and aberrant centrosome biogenesis in the embryonic kidney stroma.
Collapse
Affiliation(s)
- Ewa Langner
- Department of Medicine (Nephrology Division), Washington University, St Louis, MO, USA
| | - Tao Cheng
- Department of Medicine (Nephrology Division), Washington University, St Louis, MO, USA
| | - Eirini Kefaloyianni
- Department of Medicine (Rheumatology Division), Washington University, St Louis, MO, USA
| | - Charles Gluck
- Department of Medicine (Nephrology Division), Washington University, St Louis, MO, USA
| | - Baolin Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, NY, USA
| | - Moe R Mahjoub
- Department of Medicine (Nephrology Division), Washington University, St Louis, MO, USA.
- Department of Cell Biology and Physiology, Washington University, St Louis, MO, USA.
| |
Collapse
|
44
|
Lyu Q, Li Q, Zhou J, Zhao H. Formation and function of multiciliated cells. J Cell Biol 2024; 223:e202307150. [PMID: 38032388 PMCID: PMC10689204 DOI: 10.1083/jcb.202307150] [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: 07/31/2023] [Revised: 10/29/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023] Open
Abstract
In vertebrates, multiciliated cells (MCCs) are terminally differentiated cells that line the airway tracts, brain ventricles, and reproductive ducts. Each MCC contains dozens to hundreds of motile cilia that beat in a synchronized manner to drive fluid flow across epithelia, the dysfunction of which is associated with a group of human diseases referred to as motile ciliopathies, such as primary cilia dyskinesia. Given the dynamic and complex process of multiciliogenesis, the biological events essential for forming multiple motile cilia are comparatively unelucidated. Thanks to advancements in genetic tools, omics technologies, and structural biology, significant progress has been achieved in the past decade in understanding the molecular mechanism underlying the regulation of multiple motile cilia formation. In this review, we discuss recent studies with ex vivo culture MCC and animal models, summarize current knowledge of multiciliogenesis, and particularly highlight recent advances and their implications.
Collapse
Affiliation(s)
- Qian Lyu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Qingchao Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Huijie Zhao
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
45
|
Turan FB, Ercan ME, Firat-Karalar EN. A Chemically Inducible Organelle Rerouting Assay to Probe Primary Cilium Assembly, Maintenance, and Disassembly in Cultured Cells. Methods Mol Biol 2024; 2725:55-78. [PMID: 37856017 DOI: 10.1007/978-1-0716-3507-0_3] [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: 10/20/2023]
Abstract
The primary cilium is a conserved, microtubule-based organelle that protrudes from the surface of most vertebrate cells as well as sensory cells of many organisms. It transduces extracellular chemical and mechanical cues to regulate diverse cellular processes during development and physiology. Loss-of-function studies via RNA interference and CRISPR/Cas9-mediated gene knockouts have been the main tool for elucidating the functions of proteins, protein complexes, and organelles implicated in cilium biology. However, these methods are limited in studying acute spatiotemporal functions of proteins as well as the connection between their cellular positioning and functions. A powerful approach based on inducible recruitment of plus or minus end-directed molecular motors to the protein of interest enables fast and precise control of protein activity in time and in space. In this chapter, we present a chemically inducible heterodimerization method for functional perturbation of centriolar satellites, an emerging membrane-less organelle involved in cilium biogenesis and function. The method we present is based on rerouting of centriolar satellites to the cell center or the periphery in mammalian epithelial cells. We also describe how this method can be applied to study the temporal functions of centriolar satellites during primary cilium assembly, maintenance, and disassembly.
Collapse
Affiliation(s)
- F Basak Turan
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - M Erdem Ercan
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey
| | - Elif Nur Firat-Karalar
- Department of Molecular Biology and Genetics, Koç University, Istanbul, Turkey.
- Koc University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
46
|
Boudjema AR, Al Jord A, Lemaître AI, Faucourt M, Delgehyr N, Spassky N, Meunier A. Live-Imaging Centriole Amplification in Mouse Brain Multiciliated Cells. Methods Mol Biol 2024; 2725:167-180. [PMID: 37856024 DOI: 10.1007/978-1-0716-3507-0_10] [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: 10/20/2023]
Abstract
Multiciliated cells (MCC) display on their apical surface hundreds of beating cilia that propel physiological fluids. They line brain ventricles where they propel the cerebrospinal liquid, airways where they clear mucus and pathogens and reproductive ducts where they concentrate the sperm in males or drive the egg along the oviducts in females. Motile cilia are nucleated from basal bodies which are modified centrioles. MCC therefore evade centriole archetypal duplication program to make several hundreds and nucleate an identical number of motile cilia. Defects in this centriole amplification process lead to severe human pathologies called "ciliary aplasia" or "acilia syndrome" and more recently renamed "reduced generation of motile cilia" (RGMC). Patients with this syndrome present frequent hydrocephaly, lung failure, and subfertility. In this manuscript, we describe the protocol we developed and optimized over the years to live image the centriole amplification dynamics. We explain why mouse brain MCC is a good model and provide the tips to enable successful spatially and temporally resolved monitoring of this massive organelle reorganization.
Collapse
Affiliation(s)
- Amélie-Rose Boudjema
- Institut de Biologie de l'École Normale Supérieure (IBENS), CNRS, UMR 8197, INSERM, U1024, Paris Sciences et Lettres (PSL), Research University, Paris, France
| | - Adel Al Jord
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS 7241 INSERM U1050, PSL Research University, Paris, France
| | - Anne-Iris Lemaître
- Heart Failure Unit, Cardiology Department, Centre Hospitalier Universitaire (CHU) Haut-Lévèque, Bordeaux, France
| | - Marion Faucourt
- Institut de Biologie de l'École Normale Supérieure (IBENS), CNRS, UMR 8197, INSERM, U1024, Paris Sciences et Lettres (PSL), Research University, Paris, France
| | - Nathalie Delgehyr
- Institut de Biologie de l'École Normale Supérieure (IBENS), CNRS, UMR 8197, INSERM, U1024, Paris Sciences et Lettres (PSL), Research University, Paris, France
| | - Nathalie Spassky
- Institut de Biologie de l'École Normale Supérieure (IBENS), CNRS, UMR 8197, INSERM, U1024, Paris Sciences et Lettres (PSL), Research University, Paris, France
| | - Alice Meunier
- Institut de Biologie de l'École Normale Supérieure (IBENS), CNRS, UMR 8197, INSERM, U1024, Paris Sciences et Lettres (PSL), Research University, Paris, France.
| |
Collapse
|
47
|
Biswas L, Schindler K. Predicting Infertility: How Genetic Variants in Oocyte Spindle Genes Affect Egg Quality. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2024; 238:1-22. [PMID: 39030352 DOI: 10.1007/978-3-031-55163-5_1] [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] [Indexed: 07/21/2024]
Abstract
Successful reproduction relies on the union of a single chromosomally normal egg and sperm. Chromosomally normal eggs develop from precursor cells, called oocytes, that have undergone accurate chromosome segregation. The process of chromosome segregation is governed by the oocyte spindle, a unique cytoskeletal machine that splits chromatin content of the meiotically dividing oocyte. The oocyte spindle develops and functions in an idiosyncratic process, which is vulnerable to genetic variation in spindle-associated proteins. Human genetic variants in several spindle-associated proteins are associated with poor clinical fertility outcomes, suggesting that heritable etiologies for oocyte dysfunction leading to infertility exist and that the spindle is a crux for female fertility. This chapter examines the mammalian oocyte spindle through the lens of human genetic variation, covering the genes TUBB8, TACC3, CEP120, AURKA, AURKC, AURKB, BUB1B, and CDC20. Specifically, it explores how patient-identified variants perturb spindle development and function, and it links these molecular changes in the oocyte to their cognate clinical consequences, such as oocyte maturation arrest, elevated egg aneuploidy, primary ovarian insufficiency, and recurrent pregnancy loss. This discussion demonstrates that small genetic errors in oocyte meiosis can result in remarkably far-ranging embryonic consequences, and thus reveals the importance of the oocyte's fine machinery in sustaining life.
Collapse
Affiliation(s)
- Leelabati Biswas
- Department of Genetics, Rutgers University, Piscataway, NJ, USA
- Human Genetics Institute of New Jersey, Piscataway, NJ, USA
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Karen Schindler
- Department of Genetics, Rutgers University, Piscataway, NJ, USA.
- Human Genetics Institute of New Jersey, Piscataway, NJ, USA.
| |
Collapse
|
48
|
Chen Z, Liu X, Kawakami M, Liu X, Baker A, Bhatawadekar A, Tyutyunyk-Massey L, Narayan K, Dmitrovsky E. CDK2 inhibition disorders centrosome stoichiometry and alters cellular outcomes in aneuploid cancer cells. Cancer Biol Ther 2023; 24:2279241. [PMID: 38031910 PMCID: PMC10766391 DOI: 10.1080/15384047.2023.2279241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Cyclin-dependent Kinase 2 (CDK2) inhibition prevents supernumerary centrosome clustering. This causes multipolarity, anaphase catastrophe and apoptotic death of aneuploid cancers. This study elucidated how CDK2 antagonism affected centrosome stoichiometry. Focused ion beam scanning electron microscopy (FIB-SEM) and immunofluorescent imaging were used. Studies interrogated multipolar mitosis after pharmacologic or genetic repression of CDK2. CDK2/9 antagonism with CYC065 (Fadraciclib)-treatment disordered centrosome stoichiometry in aneuploid cancer cells, preventing centrosome clustering. This caused ring-like chromosomes or multipolar cancer cells to form before onset of cell death. Intriguingly, CDK2 inhibition caused a statistically significant increase in single centrioles rather than intact centrosomes with two centrioles in cancer cells having chromosome rings or multipolarity. Statistically significant alterations in centrosome stoichiometry were undetected in other mitotic cancer cells. To confirm this pharmacodynamic effect, CDK2 but not CDK9 siRNA-mediated knockdown augmented cancer cells with chromosome ring or multipolarity formation. Notably, engineered gain of CDK2, but not CDK9 expression, reversed emergence of cancer cells with chromosome rings or multipolarity, despite CYC065-treatment. In marked contrast, CDK2 inhibition of primary human alveolar epithelial cells did not confer statistically significant increases of cells with ring-like chromosomes or multipolarity. Hence, CDK2 antagonism caused differential effects in malignant versus normal alveolar epithelial cells. Translational relevance was confirmed by CYC065-treatment of syngeneic lung cancers in mice. Mitotic figures in tumors exhibited chromosome rings or multipolarity. Thus, CDK2 inhibition preferentially disorders centrosome stoichiometry in cancer cells. Engaging this disruption is a strategy to explore against aneuploid cancers in future clinical trials.
Collapse
Affiliation(s)
- Zibo Chen
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Xi Liu
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Masanori Kawakami
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Xiuxia Liu
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Allison Baker
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda and Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Aayush Bhatawadekar
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda and Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Liliya Tyutyunyk-Massey
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda and Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Ethan Dmitrovsky
- Molecular Pharmacology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| |
Collapse
|
49
|
Pierron M, Woglar A, Busso C, Jha K, Mikeladze‐Dvali T, Croisier M, Gönczy P. Centriole elimination during Caenorhabditis elegans oogenesis initiates with loss of the central tube protein SAS-1. EMBO J 2023; 42:e115076. [PMID: 37987153 PMCID: PMC10711648 DOI: 10.15252/embj.2023115076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/22/2023] Open
Abstract
In most metazoans, centrioles are lost during oogenesis, ensuring that the zygote is endowed with the correct number of two centrioles, which are paternally contributed. How centriole architecture is dismantled during oogenesis is not understood. Here, we analyze with unprecedent detail the ultrastructural and molecular changes during oogenesis centriole elimination in Caenorhabditis elegans. Centriole elimination begins with loss of the so-called central tube and organelle widening, followed by microtubule disassembly. The resulting cluster of centriolar proteins then disappears gradually, usually moving in a microtubule- and dynein-dependent manner to the plasma membrane. Our analysis indicates that neither Polo-like kinases nor the PCM, which modulate oogenesis centriole elimination in Drosophila, do so in C. elegans. Furthermore, we demonstrate that the central tube protein SAS-1 normally departs initially from the organelle, which loses integrity earlier in sas-1 mutants. Overall, our work provides novel mechanistic insights regarding the fundamental process of oogenesis centriole elimination.
Collapse
Affiliation(s)
- Marie Pierron
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| | - Alexander Woglar
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| | - Coralie Busso
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| | - Keshav Jha
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| | | | - Marie Croisier
- BIO‐EM platform, School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life SciencesSwiss Federal Institute of Technology Lausanne (EPFL)LausanneSwitzerland
| |
Collapse
|
50
|
Sullenberger C, Kong D, Avazpour P, Luvsanjav D, Loncarek J. Centrosomal organization of Cep152 provides flexibility in Plk4 and procentriole positioning. J Cell Biol 2023; 222:e202301092. [PMID: 37707473 PMCID: PMC10501443 DOI: 10.1083/jcb.202301092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/25/2023] [Accepted: 08/17/2023] [Indexed: 09/15/2023] Open
Abstract
Centriole duplication is a high-fidelity process driven by Polo-like kinase 4 (Plk4) and a few conserved initiators. Dissecting how Plk4 and its receptors organize within centrosomes is critical to understand the centriole duplication process and biochemical and architectural differences between centrosomes of different species. Here, at nanoscale resolution, we dissect centrosomal localization of Plk4 in G1 and S phase in its catalytically active and inhibited state during centriole duplication and amplification. We build a precise distribution map of Plk4 and its receptor Cep152, as well as Cep44, Cep192, and Cep152-anchoring factors Cep57 and Cep63. We find that Cep57, Cep63, Cep44, and Cep192 localize in ninefold symmetry. However, during centriole maturation, Cep152, which we suggest is the major Plk4 receptor, develops a more complex pattern. We propose that the molecular arrangement of Cep152 creates flexibility for Plk4 and procentriole placement during centriole initiation. As a result, procentrioles form at variable positions in relation to the mother centriole microtubule triplets.
Collapse
Affiliation(s)
- Catherine Sullenberger
- Cancer Innovation Laboratory, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD, USA
| | - Dong Kong
- Cancer Innovation Laboratory, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD, USA
| | - Pegah Avazpour
- Cancer Innovation Laboratory, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD, USA
| | - Delgermaa Luvsanjav
- Cancer Innovation Laboratory, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD, USA
| | - Jadranka Loncarek
- Cancer Innovation Laboratory, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD, USA
| |
Collapse
|