1
|
Shin B, Kim M, Lee Y, Rhee K. M phase-specific generation of supernumerary centrioles in cancer cells. Mol Biol Cell 2025; 36:ar65. [PMID: 40266756 DOI: 10.1091/mbc.e24-08-0386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
Abstract
Many cancer cells maintain supernumerary centrioles, despite the potential risks associated with catastrophic outcomes during mitosis. In this study, we searched for cancer cell lines in which supernumerary centrioles are generated during the M phase and identified a few cell lines among the dozen examined. PLK4 activity is also required for M phase-specific generation of supernumerary centrioles. We observed that mitotic centrioles prematurely separate in many cancer cells when levels of pericentriolar material (PCM) proteins, such as PCNT and CEP215, are low. Furthermore, the presence of supernumerary centrioles was correlated with reduced mitotic PCM levels. Notably, overexpression of PCNT led to a reduction in supernumerary centrioles in MDA-MB-157 cells. These findings suggest that diminution of mitotic PCM may be a cause of M phase-specific generation of supernumerary centrioles in selected cancer cells.
Collapse
Affiliation(s)
- Byungho Shin
- Department of Biological Sciences, Seoul National University, Seoul, South Korea 08826
| | - Myungse Kim
- Department of Biological Sciences, Seoul National University, Seoul, South Korea 08826
| | - Yejoo Lee
- Department of Biological Sciences, Seoul National University, Seoul, South Korea 08826
| | - Kunsoo Rhee
- Department of Biological Sciences, Seoul National University, Seoul, South Korea 08826
| |
Collapse
|
2
|
Curinha A, Huang Z, Anglen T, Strong MA, Gliech CR, Jewett CE, Friskes A, Phan TP, Nicholas Z, Holland AJ. Centriole structural integrity defects are a crucial feature of hydrolethalus syndrome. J Cell Biol 2025; 224:e202403022. [PMID: 40009365 PMCID: PMC11864076 DOI: 10.1083/jcb.202403022] [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: 03/04/2024] [Revised: 11/16/2024] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Hydrolethalus syndrome (HLS) is a lethal, autosomal recessive ciliopathy caused by the mutation of the conserved centriole protein HYLS1. How HYLS1 controls centriole function 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 HLS. These phenotypes arise from a loss of centriole integrity that causes tissue-specific defects in cilia assembly and function. We show that HYLS1 is recruited to the centriole by CEP120 and stabilizes the localization of centriole inner scaffold proteins that ensure the integrity of the centriolar microtubule wall. The HLS disease mutation reduced the centriole localization of HYLS1 and caused degeneration of the centriole distal end. We propose that tissue-specific defects in centriole integrity caused by the HYLS1 mutation prevent ciliogenesis and contribute to 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 Rheumatology, 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, Netherlands
| | - Thao P. Phan
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Zachary Nicholas
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew J. Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Skinner MW, Nhan PB, Simington CJ, Jordan PW. Meiotic divisions and round spermatid formation do not require centriole duplication in mice. PLoS Genet 2025; 21:e1011698. [PMID: 40294089 PMCID: PMC12064039 DOI: 10.1371/journal.pgen.1011698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 05/09/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
Centrosomes, composed of centrioles and pericentriolar matrix proteins, are traditionally viewed as essential microtubule-organizing centers (MTOCs) that facilitate bipolar spindle formation and chromosome segregation during spermatogenesis. In this study, we investigated the role of centrioles in male germ cell development by using a murine conditional knockout (cKO) of Sas4, a critical component of centriole biogenesis. We found that while centriole duplication was impaired in Sas4 cKO spermatocytes, these cells were still capable of progressing through meiosis I and II. Chromosome segregation was able to proceed through the formation of a non-centrosomal MTOC, indicating that centrioles are not required for meiotic divisions. However, spermatids that inherited fewer than two centrioles exhibited severe defects in spermiogenesis, including improper manchette formation, constricted perinuclear rings, disrupted acrosome morphology, and failure to form flagella. Consequently, Sas4 cKO males were infertile due to the absence of functional spermatozoa. Our findings demonstrate that while centrioles are dispensable for meiosis in male germ cells, they are essential for spermiogenesis and sperm maturation. This work provides key insights into the role of centrosomes in male fertility and may have implications for understanding certain conditions of male infertility associated with centriole defects.
Collapse
Affiliation(s)
- Marnie W. Skinner
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Paula B. Nhan
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Carter J. Simington
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Philip W. Jordan
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- School of Biomedicine, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
4
|
Iyer SS, Chen F, Ogunmolu FE, Moradi S, Volkov VA, van Grinsven EJ, van Hoorn C, Wu J, Andrea N, Hua S, Jiang K, Vakonakis I, Potočnjak M, Herzog F, Gigant B, Gudimchuk N, Stecker KE, Dogterom M, Steinmetz MO, Akhmanova A. Centriolar cap proteins CP110 and CPAP control slow elongation of microtubule plus ends. J Cell Biol 2025; 224:e202406061. [PMID: 39847124 PMCID: PMC11756378 DOI: 10.1083/jcb.202406061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 01/30/2025] Open
Abstract
Centrioles are microtubule-based organelles required for the formation of centrosomes and cilia. Centriolar microtubules, unlike their cytosolic counterparts, are stable and grow very slowly, but the underlying mechanisms are poorly understood. Here, we reconstituted in vitro the interplay between the proteins that cap distal centriole ends and control their elongation: CP110, CEP97, and CPAP/SAS-4. We found that whereas CEP97 does not bind to microtubules directly, CP110 autonomously binds microtubule plus ends, blocks their growth, and inhibits depolymerization. Cryo-electron tomography revealed that CP110 associates with the luminal side of microtubule plus ends and suppresses protofilament flaring. CP110 directly interacts with CPAP, which acts as a microtubule polymerase that overcomes CP110-induced growth inhibition. Together, the two proteins impose extremely slow processive microtubule growth. Disruption of CP110-CPAP interaction in cells inhibits centriole elongation and increases incidence of centriole defects. Our findings reveal how two centriolar cap proteins with opposing activities regulate microtubule plus-end elongation and explain their antagonistic relationship during centriole formation.
Collapse
Affiliation(s)
- Saishree S. Iyer
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Fangrui Chen
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Funso E. Ogunmolu
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Shoeib Moradi
- Division of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institut, Villigen, Switzerland
| | - Vladimir A. Volkov
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, The Netherlands
| | - Emma J. van Grinsven
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Chris van Hoorn
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Jingchao Wu
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Nemo Andrea
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, The Netherlands
| | - Shasha Hua
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Kai Jiang
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | | | - Mia Potočnjak
- Ludwig-Maximilians-Universität München, Munich, Germany
| | - Franz Herzog
- Ludwig-Maximilians-Universität München, Munich, Germany
| | - Benoît Gigant
- CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, Gif-sur-Yvette, France
| | - Nikita Gudimchuk
- Department of Physics, and Center for Theoretical Problems of Physico-Chemical Pharmacology, Russian Academy of Sciences, Lomonosov Moscow State University, Moscow, Russia
| | - Kelly E. Stecker
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Marileen Dogterom
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, The Netherlands
| | - Michel O. Steinmetz
- Division of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institut, Villigen, Switzerland
- University of Basel, Biozentrum, Basel, Switzerland
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
5
|
Nagy A, Kovacs L, Rangone H, Fu J, Ladinsky M, Glover DM. Interactions of N- and C-terminal parts of Ana1 permitting centriole duplication but not elongation. Open Biol 2025; 15:240325. [PMID: 39904373 PMCID: PMC11793955 DOI: 10.1098/rsob.240325] [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: 10/30/2024] [Revised: 12/18/2024] [Accepted: 01/09/2025] [Indexed: 02/06/2025] Open
Abstract
The conserved process of centriole duplication requires the establishment of a Sas6-centred cartwheel initiated by Plk4's phosphorylation of Ana1/STIL. Subsequently, the centriole undergoes conversion to a centrosome requiring its radial expansion and elongation, mediated by a network requiring interactions between Cep135, Ana1/Cep295 and Asterless/Cep152. Here, we show that mutant alleles encoding overlapping N- and C-terminal parts of Ana1 are capable of intragenic complementation to rescue radial expansion. This permits the recruitment of Asl and thereby centriole duplication and mechanosensory cilia formation to restore the coordination defects of these mutants. This genetic combination also rescues centriole duplication in the male germ line but does not rescue the elongation of the triplet microtubule-containing centrioles of primary spermatocytes. Consequently, these males are coordinated but sterile. Such centriole elongation is rescued by the continuous, full-length Ana1 sequence. We define a region that when deleted within otherwise intact Ana1 does not permit primary spermatocyte centrioles to elongate but still allows recruitment of Asl. Our findings point to differing demands upon the physical organization of Ana1 for the distinct processes of radial expansion and elongation of centrioles.
Collapse
Affiliation(s)
- Agota Nagy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125, USA
| | - Levente Kovacs
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125, USA
- Babeș-Bolyai University, Cluj-Napoca, Romania
| | - Helene Rangone
- Department of Genetics, University of Cambridge, CambridgeCB2 3EH, UK
| | - Jingyan Fu
- Department of Genetics, University of Cambridge, CambridgeCB2 3EH, UK
| | - Mark Ladinsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125, USA
| | - David M. Glover
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA91125, USA
- Department of Genetics, University of Cambridge, CambridgeCB2 3EH, UK
| |
Collapse
|
6
|
Nagy A, Kovacs L, Rangone H, Fu J, Ladinsky M, Glover DM. Interactions of N- and C-terminal parts of Ana1 permitting centriole duplication but not elongation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620588. [PMID: 39554154 PMCID: PMC11565839 DOI: 10.1101/2024.10.28.620588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The conserved process of centriole duplication requires establishment of a Sas6-centred cartwheel initiated by Plk4's phosphorylation of Ana1/STIL. Subsequently the centriole undergoes conversion to a centrosome requiring its radial expansion and elongation, mediated by a network requiring interactions between Cep135, Ana1/Cep295, and Asterless/Cep152. Here we show that mutant alleles encoding overlapping N- and C-terminal parts of Ana1 are capable of intragenic complementation to rescue radial expansion. This permits recruitment of Asl and thereby centriole duplication and mechanosensory cilia formation to restore the coordination defects of these mutants. This genetic combination also rescues centriole duplication in the male germ line but does not rescue the elongation of the triplet microtubule-containing centrioles of primary spermatocytes and consequently these males are coordinated but sterile. Such centriole elongation is rescued by the continuous, full-length Ana1 sequence. We define a region that when deleted within otherwise intact Ana1 does not permit primary spermatocyte centrioles to elongate but still allows recruitment of Asl. Our findings point to differing demands upon the physical organization of Ana1 for the distinct processes of radial expansion and elongation of centrioles.
Collapse
Affiliation(s)
- Agota Nagy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Levente Kovacs
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
- Babeş-Bolyai University, Cluj-Napoca, Romania
| | - Helene Rangone
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Jingyan Fu
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
- Present address: College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Mark Ladinsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - David M Glover
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| |
Collapse
|
7
|
Saju A, Chen PP, Weng TH, Tsai SY, Tanaka A, Tseng YT, Chang CC, Wang CH, Shimamoto Y, Hsia KC. HURP binding to the vinca domain of β-tubulin accounts for cancer drug resistance. Nat Commun 2024; 15:8844. [PMID: 39397030 PMCID: PMC11471760 DOI: 10.1038/s41467-024-53139-y] [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: 02/23/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024] Open
Abstract
Vinca alkaloids, a class of tubulin-binding agent, are widely used in treating cancer, yet the emerging resistance compromises their efficacy. Hepatoma up-regulated protein (HURP), a microtubule-associated protein displaying heightened expression across various cancer types, reduces cancer cells' sensitivity to vinca-alkaloid drugs upon overexpression. However, the molecular basis behind this drug resistance remains unknown. Here we discover a tubulin-binding domain within HURP, and establish its role in regulating microtubule growth. Cryo-EM analysis reveals interactions between HURP's tubulin-binding domain and the vinca domain on β-tubulin -- the site targeted by vinca alkaloid drugs. Importantly, HURP competes directly with vinorelbine, a vinca alkaloid-based chemotherapeutic agent, countering microtubule growth defects caused by vinorelbine both in vitro and in vivo. Our findings elucidate a mechanism driving drug resistance in HURP-overexpressing cancer cells and emphasize HURP tubulin-binding domain's role in mitotic spindle assembly. This underscores its potential as a therapeutic target to improve cancer treatment.
Collapse
Affiliation(s)
- Athira Saju
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Molecular and Cell Biology, Taiwan International Graduate Program and National Defense Medical Center, Taipei, Taiwan
| | - Po-Pang Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Institute of Biochemistry and Molecular biology, College of Life Sciences, National Yang-Ming Chiao-Tung University, Hsinchu, Taiwan
| | - Tzu-Han Weng
- Department of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Su-Yi Tsai
- Department of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Akihiro Tanaka
- Department of Chromosome Science, National Institute of Genetics, Shizuoka, Japan
| | - Yu-Ting Tseng
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chih-Chia Chang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chun-Hsiung Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Yuta Shimamoto
- Department of Chromosome Science, National Institute of Genetics, Shizuoka, Japan
| | - Kuo-Chiang Hsia
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
- Molecular and Cell Biology, Taiwan International Graduate Program and National Defense Medical Center, Taipei, Taiwan.
- Institute of Biochemistry and Molecular biology, College of Life Sciences, National Yang-Ming Chiao-Tung University, Hsinchu, Taiwan.
| |
Collapse
|
8
|
Pfister JA, Agostini L, Bournonville L, Sankaralingam P, Bell ZG, Hamel V, Guichard P, Biertümpfel C, Mizuno N, O’Connell KF. The C. elegans homolog of Sjögren's Syndrome Nuclear Antigen 1 is required for the structural integrity of the centriole and bipolar mitotic spindle assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.03.616528. [PMID: 39803516 PMCID: PMC11722412 DOI: 10.1101/2024.10.03.616528] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Centrioles play central roles in ciliogenesis and mitotic spindle assembly. Once assembled, centrioles exhibit long-term stability, a property essential for maintaining numerical control. How centriole stability is achieved and how it is lost in certain biological contexts are still not completely understood. In this study we show that SSNA-1, the Caenorhabditis elegans ortholog of Sjogren's Syndrome Nuclear Antigen 1, is a centriole constituent that localizes close to the microtubule outer wall, while also exhibiting a developmentally regulated association with centriole satellite-like structures. A complete deletion of the ssna-1 gene results in an embryonic lethal phenotype marked by the appearance of extra centrioles and spindle poles. We show that SSNA-1 genetically interacts with the centriole stability factor SAS-1 and is required post assembly for centriole structural integrity. In SSNA-1's absence, centrioles assemble but fracture leading to extra spindle poles. However, if the efficiency of cartwheel assembly is reduced, the absence of SSNA-1 results in daughter centriole loss and monopolar spindle formation, indicating that the cartwheel and SSNA-1 cooperate to stabilize the centriole during assembly. Our work thus shows that SSNA-1 contributes to centriole stability during and after assembly, thereby ensuring proper centriole number.
Collapse
Affiliation(s)
- Jason A. Pfister
- Laboratory of Biochemistry and Genetics, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lorenzo Agostini
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lorène Bournonville
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Prabhu Sankaralingam
- Laboratory of Biochemistry and Genetics, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Zachary G. Bell
- Laboratory of Biochemistry and Genetics, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Virginie Hamel
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Paul Guichard
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Christian Biertümpfel
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Naoko Mizuno
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kevin F. O’Connell
- Laboratory of Biochemistry and Genetics, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
9
|
Panda P, Ladinsky MS, Glover DM. 9-fold symmetry is not essential for centriole elongation and formation of new centriole-like structures. Nat Commun 2024; 15:4467. [PMID: 38796459 PMCID: PMC11127918 DOI: 10.1038/s41467-024-48831-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 05/14/2024] [Indexed: 05/28/2024] Open
Abstract
As daughter centrioles assemble during G2, they recruit conserved Ana3/RTTN followed by its partner Rcd4/PPP1R35. Together, this contributes to the subsequent recruitment of Ana1/CEP295, required for the centriole's conversion to a centrosome. Here, we show that Rcd4/PPP1R35 is also required to maintain 9-fold centriole symmetry in the Drosophila male germline; its absence causes microtubule triplets to disperse into a reduced number of doublet or singlet microtubules. rcd4-null mutant spermatocytes display skinny centrioles that elongate normally and localize centriolar components correctly. Mutant spermatocytes also have centrioles of normal girth that splay at their proximal ends when induced to elongate by Ana1 overexpression. Skinny and splayed spermatid centrioles can still recruit a proximal centriole-like (PCL) structure marking a capability to initiate features of centriole duplication in developing sperm. Thus, stable 9-fold symmetry of microtubule triplets is not essential for centriole growth, correct longitudinal association of centriole components, and aspects of centriole duplication.
Collapse
Affiliation(s)
- Pallavi Panda
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Mark S Ladinsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - David M Glover
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
10
|
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
|
11
|
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] [Download PDF] [Figures] [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
|
12
|
Köhrer S, Dittrich T, Schorb M, Weinhold N, Haberbosch I, Börmel M, Pajor G, Goldschmidt H, Müller-Tidow C, Raab MS, John L, Seckinger A, Brobeil A, Dreger P, Tornóczky T, Pajor L, Hegenbart U, Schönland SO, Schwab Y, Krämer A. High-throughput electron tomography identifies centriole over-elongation as an early event in plasma cell disorders. Leukemia 2023; 37:2468-2478. [PMID: 37821581 PMCID: PMC10681902 DOI: 10.1038/s41375-023-02056-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/14/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023]
Abstract
Plasma cell disorders are clonal outgrowths of pre-malignant or malignant plasma cells, characterized by extensive chromosomal aberrations. Centrosome abnormalities are a major driver of chromosomal instability in cancer but their origin, incidence, and composition in primary tumor cells is poorly understood. Using cutting-edge, semi-automated high-throughput electron tomography, we characterized at nanoscale 1386 centrioles in CD138pos plasma cells from eight healthy donors and 21 patients with plasma cell disorders, and 722 centrioles from different control populations. In plasma cells from healthy individuals, over-elongated centrioles accumulated with age. In plasma cell disorders, centriole over-elongation was notably frequent in early, pre-malignant disease stages, became less pronounced in overt multiple myeloma, and almost entirely disappeared in aggressive plasma cell leukemia. Centrioles in other types of patient-derived B cell neoplasms showed no over-elongation. In contrast to current belief, centriole length appears to be highly variable in long-lived, healthy plasma cells, and over-elongation and structural aberrations are common in this cell type. Our data suggest that structural centrosome aberrations accumulate with age in healthy CD138pos plasma cells and may thus play an important role in early aneuploidization as an oncogenic driver in plasma cell disorders.
Collapse
Affiliation(s)
- Sebastian Köhrer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Tobias Dittrich
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Amyloidosis Center, University of Heidelberg, Heidelberg, Germany
| | - Martin Schorb
- Electron Microscopy Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Niels Weinhold
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Isabella Haberbosch
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Mandy Börmel
- Electron Microscopy Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Gabor Pajor
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Pathology, University of Pécs Medical School and Clinic, Pécs, Hungary
| | - Hartmut Goldschmidt
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Internal Medicine V, GMMG-Studygroup at University of Heidelberg, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany
| | - Marc S Raab
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Lukas John
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Anja Seckinger
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Alexander Brobeil
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Tamás Tornóczky
- Department of Pathology, University of Pécs Medical School and Clinic, Pécs, Hungary
| | - László Pajor
- Department of Pathology, University of Pécs Medical School and Clinic, Pécs, Hungary
| | - Ute Hegenbart
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Amyloidosis Center, University of Heidelberg, Heidelberg, Germany
| | - Stefan O Schönland
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Amyloidosis Center, University of Heidelberg, Heidelberg, Germany
| | - Yannick Schwab
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- Electron Microscopy Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
13
|
Campanacci V, Gigant B. The C-terminus of stathmin-like proteins governs the stability of their complexes with tubulin. Biochem Biophys Res Commun 2023; 682:244-249. [PMID: 37826947 DOI: 10.1016/j.bbrc.2023.10.023] [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: 10/05/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023]
Abstract
Microtubule dynamics is modulated by many cellular factors including stathmin family proteins. Vertebrate stathmins sequester two αβ-tubulin heterodimers into a tight complex that cannot be incorporated in microtubules. Stathmins are regulated at the expression level during development and among tissues; they are also regulated by phosphorylation. Here, we study the dissociation kinetics of tubulin:stathmin assemblies in presence of different tubulin-binding proteins and identify a critical role of the C-terminus of the stathmin partner. Destabilizing this C-terminal region may represent an additional regulatory mechanism of the interaction with tubulin of stathmin proteins.
Collapse
Affiliation(s)
- Valérie Campanacci
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Benoît Gigant
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.
| |
Collapse
|
14
|
Carden S, Vitiello E, Rosa E Silva I, Holder J, Quarantotti V, Kishore K, Roamio Franklin VN, D'Santos C, Ochi T, van Breugel M, Gergely F. Proteomic profiling of centrosomes across multiple mammalian cell and tissue types by an affinity capture method. Dev Cell 2023; 58:2393-2410.e9. [PMID: 37852252 DOI: 10.1016/j.devcel.2023.09.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/27/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023]
Abstract
Centrosomes are the major microtubule-organizing centers in animals and play fundamental roles in many cellular processes. Understanding how their composition varies across diverse cell types and how it is altered in disease are major unresolved questions, yet currently available centrosome isolation protocols are cumbersome and time-consuming, and they lack scalability. Here, we report the development of centrosome affinity capture (CAPture)-mass spectrometry (MS), a powerful one-step purification method to obtain high-resolution centrosome proteomes from mammalian cells. Utilizing a synthetic peptide derived from CCDC61 protein, CAPture specifically isolates intact centrosomes. Importantly, as a bead-based affinity method, it enables rapid sample processing and multiplexing unlike conventional approaches. Our study demonstrates the power of CAPture-MS to elucidate cell-type-dependent heterogeneity in centrosome composition, dissect hierarchical interactions, and identify previously unknown centrosome components. Overall, CAPture-MS represents a transformative tool to unveil temporal, regulatory, cell-type- and tissue-specific changes in centrosome proteomes in health and disease.
Collapse
Affiliation(s)
- Sarah Carden
- CRUK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Elisa Vitiello
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | - James Holder
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Valentina Quarantotti
- CRUK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Kamal Kishore
- CRUK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | | | - Clive D'Santos
- CRUK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Takashi Ochi
- MRC Laboratory of Molecular Biology, Cambridge, UK; The Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, UK.
| | - Mark van Breugel
- MRC Laboratory of Molecular Biology, Cambridge, UK; School of Biological & Behavioural Sciences, Queen Mary University of London, London, UK.
| | - Fanni Gergely
- CRUK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Department of Biochemistry, University of Oxford, Oxford, UK.
| |
Collapse
|
15
|
Gopalakrishnan J, Feistel K, Friedrich BM, Grapin‐Botton A, Jurisch‐Yaksi N, Mass E, Mick DU, Müller R, May‐Simera H, Schermer B, Schmidts M, Walentek P, Wachten D. Emerging principles of primary cilia dynamics in controlling tissue organization and function. EMBO J 2023; 42:e113891. [PMID: 37743763 PMCID: PMC10620770 DOI: 10.15252/embj.2023113891] [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: 02/27/2023] [Revised: 08/07/2023] [Accepted: 09/08/2023] [Indexed: 09/26/2023] Open
Abstract
Primary cilia project from the surface of most vertebrate cells and are key in sensing extracellular signals and locally transducing this information into a cellular response. Recent findings show that primary cilia are not merely static organelles with a distinct lipid and protein composition. Instead, the function of primary cilia relies on the dynamic composition of molecules within the cilium, the context-dependent sensing and processing of extracellular stimuli, and cycles of assembly and disassembly in a cell- and tissue-specific manner. Thereby, primary cilia dynamically integrate different cellular inputs and control cell fate and function during tissue development. Here, we review the recently emerging concept of primary cilia dynamics in tissue development, organization, remodeling, and function.
Collapse
Affiliation(s)
- Jay Gopalakrishnan
- Institute for Human Genetics, Heinrich‐Heine‐UniversitätUniversitätsklinikum DüsseldorfDüsseldorfGermany
| | - Kerstin Feistel
- Department of Zoology, Institute of BiologyUniversity of HohenheimStuttgartGermany
| | | | - Anne Grapin‐Botton
- Cluster of Excellence Physics of Life, TU DresdenDresdenGermany
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at The University Hospital Carl Gustav Carus and Faculty of Medicine of the TU DresdenDresdenGermany
| | - Nathalie Jurisch‐Yaksi
- Department of Clinical and Molecular MedicineNorwegian University of Science and TechnologyTrondheimNorway
| | - Elvira Mass
- Life and Medical Sciences Institute, Developmental Biology of the Immune SystemUniversity of BonnBonnGermany
| | - David U Mick
- Center for Molecular Signaling (PZMS), Center of Human and Molecular Biology (ZHMB)Saarland School of MedicineHomburgGermany
| | - Roman‐Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD), Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
| | - Helen May‐Simera
- Institute of Molecular PhysiologyJohannes Gutenberg‐UniversityMainzGermany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD), Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
| | - Miriam Schmidts
- Pediatric Genetics Division, Center for Pediatrics and Adolescent MedicineUniversity Hospital FreiburgFreiburgGermany
- CIBSS‐Centre for Integrative Biological Signalling StudiesUniversity of FreiburgFreiburgGermany
| | - Peter Walentek
- CIBSS‐Centre for Integrative Biological Signalling StudiesUniversity of FreiburgFreiburgGermany
- Renal Division, Internal Medicine IV, Medical CenterUniversity of FreiburgFreiburgGermany
| | - Dagmar Wachten
- Institute of Innate Immunity, Biophysical Imaging, Medical FacultyUniversity of BonnBonnGermany
| |
Collapse
|
16
|
Kalbfuss N, Gönczy P. Towards understanding centriole elimination. Open Biol 2023; 13:230222. [PMID: 37963546 PMCID: PMC10645514 DOI: 10.1098/rsob.230222] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/14/2023] [Indexed: 11/16/2023] Open
Abstract
Centrioles are microtubule-based structures crucial for forming flagella, cilia and centrosomes. Through these roles, centrioles are critical notably for proper cell motility, signalling and division. Recent years have advanced significantly our understanding of the mechanisms governing centriole assembly and architecture. Although centrioles are typically very stable organelles, persisting over many cell cycles, they can also be eliminated in some cases. Here, we review instances of centriole elimination in a range of species and cell types. Moreover, we discuss potential mechanisms that enable the switch from a stable organelle to a vanishing one. Further work is expected to provide novel insights into centriole elimination mechanisms in health and disease, thereby also enabling scientists to readily manipulate organelle fate.
Collapse
Affiliation(s)
- Nils Kalbfuss
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
17
|
Jaiswal S, Sanghi S, Singh P. Separation-of-function MCPH-associated mutations in CPAP affect centriole number and length. J Cell Sci 2023; 136:jcs261297. [PMID: 37823337 DOI: 10.1242/jcs.261297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023] Open
Abstract
Centrioles are microtubule-based cylindrical ultrastructures characterized by their definite size and robustness. The molecular capping protein, CPAP (also known as CENPJ) engages its N-terminal region with the centriole microtubules to regulate their length. Nevertheless, the conserved C-terminal glycine-rich G-box of CPAP, which interacts with the centriole inner cartwheel protein STIL, is frequently mutated in primary microcephaly (MCPH) patients. Here, we show that two different MCPH-associated variants, E1235V and D1196N in the CPAP G-box, affect distinct functions at centrioles. The E1235V mutation reduces CPAP centriole recruitment and causes overly long centrioles. The D1196N mutation increases centriole numbers without affecting centriole localization. Both mutations prevent binding to STIL, which controls centriole duplication. Our work highlights the involvement of an alternative CEP152-dependent route for CPAP centriole localization. Molecular dynamics simulations suggest that E1235V leads to an increase in G-box flexibility, which could have implications on its molecular interactions. Collectively, we demonstrate that a CPAP region outside the microtubule-interacting domains influences centriole number and length, which translates to spindle defects and reduced cell viability. Our work provides new insights into the molecular causes of primary microcephaly.
Collapse
Affiliation(s)
- Sonal Jaiswal
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342037, Jodhpur, Rajasthan, India
| | - Srishti Sanghi
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342037, Jodhpur, Rajasthan, India
| | - Priyanka Singh
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342037, Jodhpur, Rajasthan, India
| |
Collapse
|
18
|
Ryniawec JM, Hannaford MR, Zibrat ME, Fagerstrom CJ, Galletta BJ, Aguirre SE, Guice BA, Dean SM, Rusan NM, Rogers GC. Cep104 is a component of the centriole distal tip complex that regulates centriole growth and contributes to Drosophila spermiogenesis. Curr Biol 2023; 33:4202-4216.e9. [PMID: 37729913 PMCID: PMC10591971 DOI: 10.1016/j.cub.2023.08.075] [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: 03/14/2023] [Revised: 07/21/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023]
Abstract
Proper centrosome number and function relies on the accurate assembly of centrioles, barrel-shaped structures that form the core duplicating elements of the organelle. The growth of centrioles is regulated in a cell cycle-dependent manner; while new daughter centrioles elongate during the S/G2/M phase, mature mother centrioles maintain their length throughout the cell cycle. Centriole length is controlled by the synchronized growth of the microtubules that ensheathe the centriole barrel. Although proteins exist that target the growing distal tips of centrioles, such as CP110 and Cep97, these proteins are generally thought to suppress centriolar microtubule growth, suggesting that distal tips may also contain unidentified counteracting factors that facilitate microtubule polymerization. Currently, a mechanistic understanding of how distal tip proteins balance microtubule growth and shrinkage to either promote daughter centriole elongation or maintain centriole length is lacking. Using a proximity-labeling screen in Drosophila cells, we identified Cep104 as a novel component of a group of evolutionarily conserved proteins that we collectively refer to as the distal tip complex (DTC). We found that Cep104 regulates centriole growth and promotes centriole elongation through its microtubule-binding TOG domain. Furthermore, analysis of Cep104 null flies revealed that Cep104 and Cep97 cooperate during spermiogenesis to align spermatids and coordinate individualization. Lastly, we mapped the complete DTC interactome and showed that Cep97 is the central scaffolding unit required to recruit DTC components to the distal tip of centrioles.
Collapse
Affiliation(s)
- John M Ryniawec
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Matthew R Hannaford
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Melanie E Zibrat
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Carey J Fagerstrom
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brian J Galletta
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sophia E Aguirre
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Bethany A Guice
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Spencer M Dean
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Nasser M Rusan
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Gregory C Rogers
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
19
|
Bieder A, Chandrasekar G, Wason A, Erkelenz S, Gopalakrishnan J, Kere J, Tapia-Páez I. Genetic and protein interaction studies between the ciliary dyslexia candidate genes DYX1C1 and DCDC2. BMC Mol Cell Biol 2023; 24:20. [PMID: 37237337 DOI: 10.1186/s12860-023-00483-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND DYX1C1 (DNAAF4) and DCDC2 are two of the most replicated dyslexia candidate genes in genetic studies. They both have demonstrated roles in neuronal migration, in cilia growth and function and they both are cytoskeletal interactors. In addition, they both have been characterized as ciliopathy genes. However, their exact molecular functions are still incompletely described. Based on these known roles, we asked whether DYX1C1 and DCDC2 interact on the genetic and the protein level. RESULTS Here, we report the physical protein-protein interaction of DYX1C1 and DCDC2 as well as their respective interactions with the centrosomal protein CPAP (CENPJ) on exogenous and endogenous levels in different cell models including brain organoids. In addition, we show a synergistic genetic interaction between dyx1c1 and dcdc2b in zebrafish exacerbating the ciliary phenotype. Finally, we show a mutual effect on transcriptional regulation among DYX1C1 and DCDC2 in a cellular model. CONCLUSIONS In summary, we describe the physical and functional interaction between the two genes DYX1C1 and DCDC2. These results contribute to the growing understanding of the molecular roles of DYX1C1 and DCDC2 and set the stage for future functional studies.
Collapse
Affiliation(s)
- Andrea Bieder
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | | | - Arpit Wason
- Center for Molecular Medicine, Institute for Biochemistry I of the University of Cologne, Cologne, Germany
| | - Steffen Erkelenz
- Institute of Human Genetics, Universitätsklinikum, Heinrich Heine University, Düsseldorf, Germany
| | - Jay Gopalakrishnan
- Institute of Human Genetics, Universitätsklinikum, Heinrich Heine University, Düsseldorf, Germany
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Molecular Neurology Research Program, University of Helsinki, Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Isabel Tapia-Páez
- Department of Medicine, Solna, Karolinska Institutet, Solnavägen 30, SE-171 76, Solna, Sweden.
| |
Collapse
|
20
|
Wiecek AJ, Cutty SJ, Kornai D, Parreno-Centeno M, Gourmet LE, Tagliazucchi GM, Jacobson DH, Zhang P, Xiong L, Bond GL, Barr AR, Secrier M. Genomic hallmarks and therapeutic implications of G0 cell cycle arrest in cancer. Genome Biol 2023; 24:128. [PMID: 37221612 PMCID: PMC10204193 DOI: 10.1186/s13059-023-02963-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 05/07/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Therapy resistance in cancer is often driven by a subpopulation of cells that are temporarily arrested in a non-proliferative G0 state, which is difficult to capture and whose mutational drivers remain largely unknown. RESULTS We develop methodology to robustly identify this state from transcriptomic signals and characterise its prevalence and genomic constraints in solid primary tumours. We show that G0 arrest preferentially emerges in the context of more stable, less mutated genomes which maintain TP53 integrity and lack the hallmarks of DNA damage repair deficiency, while presenting increased APOBEC mutagenesis. We employ machine learning to uncover novel genomic dependencies of this process and validate the role of the centrosomal gene CEP89 as a modulator of proliferation and G0 arrest capacity. Lastly, we demonstrate that G0 arrest underlies unfavourable responses to various therapies exploiting cell cycle, kinase signalling and epigenetic mechanisms in single-cell data. CONCLUSIONS We propose a G0 arrest transcriptional signature that is linked with therapeutic resistance and can be used to further study and clinically track this state.
Collapse
Affiliation(s)
- Anna J. Wiecek
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Stephen J. Cutty
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Daniel Kornai
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Mario Parreno-Centeno
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Lucie E. Gourmet
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| | | | - Daniel H. Jacobson
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
- UCL Cancer Institute, Paul O’Gorman Building, University College London, London, UK
| | - Ping Zhang
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Lingyun Xiong
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Gareth L. Bond
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Alexis R. Barr
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- Cell Cycle Control Team, MRC London Institute of Medical Sciences (LMS), London, UK
| | - Maria Secrier
- UCL Genetics Institute, Department of Genetics, Evolution and Environment, University College London, London, UK
| |
Collapse
|
21
|
Uzbekov R, Singina GN, Shedova EN, Banliat C, Avidor-Reiss T, Uzbekova S. Centrosome Formation in the Bovine Early Embryo. Cells 2023; 12:1335. [PMID: 37174735 PMCID: PMC10177215 DOI: 10.3390/cells12091335] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/21/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
Centrosome formation during early development in mice and rats occurs due to the appearance of centrioles de novo. In contrast, in humans and other non-rodent mammals, centrioles are thought to be derived from spermatozoa. Ultrastructural study of zygotes and early embryos of cattle at full series of ultrathin sections show that the proximal centriole of the spermatozoon disappears by the end of the first cleavage division. Centrioles appear in two to four cell embryos in fertilized oocytes and in parthenogenetic embryos. Centriole formation includes the appearance of atypical centrioles with randomly arranged triplets and centrioles with microtubule triplets of various lengths. After the third cleavage, four centriolar cylinders appear for the first time in the blastomeres while each embryo still has two atypical centrioles. Our results showed that the mechanisms of centriole formation in different groups of mammals are universal, differing only in the stage of development in which they occur.
Collapse
Affiliation(s)
- Rustem Uzbekov
- Laboratory of Cell Biology and Electron Microscopy, Faculty of Medicine, University of Tours, 37032 Tours, France
- Faculty of Bioengineering and Bioinformatics, Moscow State University, 119992 Moscow, Russia
| | - Galina N. Singina
- Laboratory of Experimental Embryology, L.K. Ernst Federal Research Center for Animal Husbandry, Moscow Region, 142132 Podolsk, Russia
| | - Ekaterina N. Shedova
- Laboratory of Experimental Embryology, L.K. Ernst Federal Research Center for Animal Husbandry, Moscow Region, 142132 Podolsk, Russia
| | - Charles Banliat
- Ecole Supérieure d’agricultures (ESA), Unité de Recherche sur les Systèmes D’élevage (URSE), 55 rue Rabelais BP, 30748 Angers, France
| | - Tomer Avidor-Reiss
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Svetlana Uzbekova
- UMR Physiologie de la Reproduction et des Comportements (PRC), INRAE, CNRS, Université de Tours, IFCE, 37380 Nouzilly, France
| |
Collapse
|
22
|
Weier AK, Homrich M, Ebbinghaus S, Juda P, Miková E, Hauschild R, Zhang L, Quast T, Mass E, Schlitzer A, Kolanus W, Burgdorf S, Gruß OJ, Hons M, Wieser S, Kiermaier E. Multiple centrosomes enhance migration and immune cell effector functions of mature dendritic cells. J Cell Biol 2022; 221:e202107134. [PMID: 36214847 PMCID: PMC9555069 DOI: 10.1083/jcb.202107134] [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: 07/24/2021] [Revised: 03/01/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022] Open
Abstract
Centrosomes play a crucial role during immune cell interactions and initiation of the immune response. In proliferating cells, centrosome numbers are tightly controlled and generally limited to one in G1 and two prior to mitosis. Defects in regulating centrosome numbers have been associated with cell transformation and tumorigenesis. Here, we report the emergence of extra centrosomes in leukocytes during immune activation. Upon antigen encounter, dendritic cells pass through incomplete mitosis and arrest in the subsequent G1 phase leading to tetraploid cells with accumulated centrosomes. In addition, cell stimulation increases expression of polo-like kinase 2, resulting in diploid cells with two centrosomes in G1-arrested cells. During cell migration, centrosomes tightly cluster and act as functional microtubule-organizing centers allowing for increased persistent locomotion along gradients of chemotactic cues. Moreover, dendritic cells with extra centrosomes display enhanced secretion of inflammatory cytokines and optimized T cell responses. Together, these results demonstrate a previously unappreciated role of extra centrosomes for regular cell and tissue homeostasis.
Collapse
Affiliation(s)
- Ann-Kathrin Weier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Mirka Homrich
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Stephanie Ebbinghaus
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Pavel Juda
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Eliška Miková
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Robert Hauschild
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Lili Zhang
- Life and Medical Sciences Institute, Quantitative Systems Biology, University of Bonn, Bonn, Germany
| | - Thomas Quast
- Life and Medical Sciences Institute, Molecular Immunology and Cell Biology, University of Bonn, Bonn, Germany
| | - Elvira Mass
- Life and Medical Sciences Institute, Developmental Biology of the Immune System, University of Bonn, Bonn, Germany
| | - Andreas Schlitzer
- Life and Medical Sciences Institute, Quantitative Systems Biology, University of Bonn, Bonn, Germany
| | - Waldemar Kolanus
- Life and Medical Sciences Institute, Molecular Immunology and Cell Biology, University of Bonn, Bonn, Germany
| | - Sven Burgdorf
- Life and Medical Sciences Institute, Cellular Immunology, University of Bonn, Bonn, Germany
| | - Oliver J. Gruß
- Institute of Genetics, University of Bonn, Bonn, Germany
| | - Miroslav Hons
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Stefan Wieser
- Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Castelldefels, Spain
| | - Eva Kiermaier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| |
Collapse
|
23
|
Forni G, Mikheyev AS, Luchetti A, Mantovani B. Gene transcriptional profiles in gonads of Bacillus taxa (Phasmida) with different cytological mechanisms of automictic parthenogenesis. ZOOLOGICAL LETTERS 2022; 8:14. [PMID: 36435814 PMCID: PMC9701443 DOI: 10.1186/s40851-022-00197-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
The evolution of automixis - i.e., meiotic parthenogenesis - requires several features, including ploidy restoration after meiosis and maintenance of fertility. Characterizing the relative contribution of novel versus pre-existing genes and the similarities in their expression and sequence evolution is fundamental to understand the evolution of reproductive novelties. Here we identify gonads-biased genes in two Bacillus automictic stick-insects and compare their expression profile and sequence evolution with a bisexual congeneric species. The two parthenogens restore ploidy through different cytological mechanisms: in Bacillus atticus, nuclei derived from the first meiotic division fuse to restore a diploid egg nucleus, while in Bacillus rossius, diploidization occurs in some cells of the haploid blastula through anaphase restitution. Parthenogens' gonads transcriptional program is found to be largely assembled from genes that were already present before the establishment of automixis. The three species transcriptional profiles largely reflect their phyletic relationships, yet we identify a shared core of genes with gonad-biased patterns of expression in parthenogens which are either male gonads-biased in the sexual species or are not differentially expressed there. At the sequence level, just a handful of gonads-biased genes were inferred to have undergone instances of positive selection exclusively in the parthenogen species. This work is the first to explore the molecular underpinnings of automixis in a comparative framework: it delineates how reproductive novelties can be sustained by genes whose origin precedes the establishment of the novelty itself and shows that different meiotic mechanisms of reproduction can be associated with a shared molecular ground plan.
Collapse
Affiliation(s)
- Giobbe Forni
- Dip. Scienze Biologiche, Geologiche e Ambientali (BiGeA), University of Bologna, 40126, Bologna, Italy
- Dip. Scienze Agrarie e Ambientali, University of Milano, Milano, Italy
| | - Alexander S Mikheyev
- Australian National University, ACT, Canberra, 2600, Australia
- Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Okinawa, 904-0495, Japan
| | - Andrea Luchetti
- Dip. Scienze Biologiche, Geologiche e Ambientali (BiGeA), University of Bologna, 40126, Bologna, Italy.
| | - Barbara Mantovani
- Dip. Scienze Biologiche, Geologiche e Ambientali (BiGeA), University of Bologna, 40126, Bologna, Italy
| |
Collapse
|
24
|
Dittrich T, Köhrer S, Schorb M, Haberbosch I, Börmel M, Goldschmidt H, Pajor G, Müller-Tidow C, Raab MS, Hegenbart U, Schönland SO, Schwab Y, Krämer A. A high-throughput electron tomography workflow reveals over-elongated centrioles in relapsed/refractory multiple myeloma. CELL REPORTS METHODS 2022; 2:100322. [PMID: 36452870 PMCID: PMC9701608 DOI: 10.1016/j.crmeth.2022.100322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/24/2022] [Accepted: 10/06/2022] [Indexed: 06/17/2023]
Abstract
Electron microscopy is the gold standard to characterize centrosomal ultrastructure. However, production of significant morphometrical data is highly limited by acquisition time. We therefore developed a generalizable, semi-automated high-throughput electron tomography strategy to study centrosome aberrations in sparse patient-derived cancer cells at nanoscale. As proof of principle, we present electron tomography data on 455 centrioles of CD138pos plasma cells from one patient with relapsed/refractory multiple myeloma and CD138neg bone marrow mononuclear cells from three healthy donors as a control. Plasma cells from the myeloma patient displayed 122 over-elongated centrioles (48.8%). Particularly mother centrioles also harbored gross structural abnormalities, including fragmentation and disturbed microtubule cylinder formation, while control centrioles were phenotypically unremarkable. These data demonstrate the feasibility of our scalable high-throughput electron tomography strategy to study structural centrosome aberrations in primary tumor cells. Moreover, our electron tomography workflow and data provide a resource for the characterization of cell organelles beyond centrosomes.
Collapse
Affiliation(s)
- Tobias Dittrich
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), and Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
- Amyloidosis Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Sebastian Köhrer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), and Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Martin Schorb
- Electron Microscopy Core Facility, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Isabella Haberbosch
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), and Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
| | - Mandy Börmel
- Electron Microscopy Core Facility, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Hartmut Goldschmidt
- Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), University of Heidelberg, 69120 Heidelberg, Germany
| | - Gabor Pajor
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), and Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), University of Heidelberg, 69120 Heidelberg, Germany
| | - Marc S. Raab
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), and Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
| | - Ute Hegenbart
- Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
- Amyloidosis Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Stefan O. Schönland
- Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
- Amyloidosis Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Yannick Schwab
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
- Electron Microscopy Core Facility, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), and Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
25
|
Vijayan K, Arang N, Wei L, Morrison R, Geiger R, Parks KR, Lewis AJ, Mast FD, Douglass AN, Kain HS, Aitchison JD, Johnson JS, Aderem A, Kaushansky A. A genome-wide CRISPR-Cas9 screen identifies CENPJ as a host regulator of altered microtubule organization during Plasmodium liver infection. Cell Chem Biol 2022; 29:1419-1433.e5. [PMID: 35738280 PMCID: PMC9481707 DOI: 10.1016/j.chembiol.2022.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 02/03/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022]
Abstract
Prior to initiating symptomatic malaria, a single Plasmodium sporozoite infects a hepatocyte and develops into thousands of merozoites, in part by scavenging host resources, likely delivered by vesicles. Here, we demonstrate that host microtubules (MTs) dynamically reorganize around the developing liver stage (LS) parasite to facilitate vesicular transport to the parasite. Using a genome-wide CRISPR-Cas9 screen, we identified host regulators of cytoskeleton organization, vesicle trafficking, and ER/Golgi stress that regulate LS development. Foci of γ-tubulin localized to the parasite periphery; depletion of centromere protein J (CENPJ), a novel regulator identified in the screen, exacerbated this re-localization and increased infection. We demonstrate that the Golgi acts as a non-centrosomal MT organizing center (ncMTOC) by positioning γ-tubulin and stimulating MT nucleation at parasite periphery. Together, these data support a model where the Plasmodium LS recruits host Golgi to form MT-mediated conduits along which host organelles are recruited to PVM and support parasite development.
Collapse
Affiliation(s)
- Kamalakannan Vijayan
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Nadia Arang
- Center for Infectious Disease Research, Seattle, WA, USA
| | - Ling Wei
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Robert Morrison
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Rechel Geiger
- MSTP Program, University of Washington, Seattle, WA, USA
| | - K Rachael Parks
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Adam J Lewis
- Center for Infectious Disease Research, Seattle, WA, USA
| | - Fred D Mast
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Alyse N Douglass
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Heather S Kain
- Center for Infectious Disease Research, Seattle, WA, USA
| | - John D Aitchison
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Biochemistry, University of Washington, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Alan Aderem
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Alexis Kaushansky
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
| |
Collapse
|
26
|
Aydogan MG, Hankins LE, Steinacker TL, Mofatteh M, Saurya S, Wainman A, Wong SS, Lu X, Zhou FY, Raff JW. Centriole distal-end proteins CP110 and Cep97 influence centriole cartwheel growth at the proximal end. J Cell Sci 2022; 135:jcs260015. [PMID: 35707992 PMCID: PMC9450887 DOI: 10.1242/jcs.260015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/13/2022] [Indexed: 11/20/2022] Open
Abstract
Centrioles are composed of a central cartwheel tethered to nine-fold symmetric microtubule (MT) blades. The centriole cartwheel and MTs are thought to grow from opposite ends of these organelles, so it is unclear how they coordinate their assembly. We previously showed that in Drosophila embryos an oscillation of Polo-like kinase 4 (Plk4) helps to initiate and time the growth of the cartwheel at the proximal end. Here, in the same model, we show that CP110 and Cep97 form a complex close to the distal-end of the centriole MTs whose levels rise and fall as the new centriole MTs grow, in a manner that appears to be entrained by the core cyclin-dependent kinase (Cdk)-Cyclin oscillator that drives the nuclear divisions in these embryos. These CP110 and Cep97 dynamics, however, do not appear to time the period of centriole MT growth directly. Instead, we find that changing the levels of CP110 and Cep97 appears to alter the Plk4 oscillation and the growth of the cartwheel at the proximal end. These findings reveal an unexpected potential crosstalk between factors normally concentrated at opposite ends of the growing centrioles, which might help to coordinate centriole growth. This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Mustafa G. Aydogan
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Laura E. Hankins
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | | | - Mohammad Mofatteh
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Saroj Saurya
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Alan Wainman
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Siu-Shing Wong
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Xin Lu
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Felix Y. Zhou
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Jordan W. Raff
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| |
Collapse
|
27
|
Chen F, Wu J, Iwanski MK, Jurriens D, Sandron A, Pasolli M, Puma G, Kromhout JZ, Yang C, Nijenhuis W, Kapitein LC, Berger F, Akhmanova A. Self-assembly of pericentriolar material in interphase cells lacking centrioles. eLife 2022; 11:77892. [PMID: 35787744 PMCID: PMC9307276 DOI: 10.7554/elife.77892] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022] Open
Abstract
The major microtubule-organizing center (MTOC) in animal cells, the centrosome, comprises a pair of centrioles surrounded by pericentriolar material (PCM), which nucleates and anchors microtubules. Centrosome assembly depends on PCM binding to centrioles, PCM self-association and dynein-mediated PCM transport, but the self-assembly properties of PCM components in interphase cells are poorly understood. Here, we used experiments and modeling to study centriole-independent features of interphase PCM assembly. We showed that when centrioles are lost due to PLK4 depletion or inhibition, dynein-based transport and self-clustering of PCM proteins are sufficient to form a single compact MTOC, which generates a dense radial microtubule array. Interphase self-assembly of PCM components depends on γ-tubulin, pericentrin, CDK5RAP2 and ninein, but not NEDD1, CEP152, or CEP192. Formation of a compact acentriolar MTOC is inhibited by AKAP450-dependent PCM recruitment to the Golgi or by randomly organized CAMSAP2-stabilized microtubules, which keep PCM mobile and prevent its coalescence. Linking of CAMSAP2 to a minus-end-directed motor leads to the formation of an MTOC, but MTOC compaction requires cooperation with pericentrin-containing self-clustering PCM. Our data reveal that interphase PCM contains a set of components that can self-assemble into a compact structure and organize microtubules, but PCM self-organization is sensitive to motor- and microtubule-based rearrangement.
Collapse
Affiliation(s)
- Fangrui Chen
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Jingchao Wu
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | | | - Daphne Jurriens
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Arianna Sandron
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Milena Pasolli
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Gianmarco Puma
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | | | - Chao Yang
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Wilco Nijenhuis
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | | | - Florian Berger
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Anna Akhmanova
- Department of Biology, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
28
|
Centrosome Defects in Hematological Malignancies: Molecular Mechanisms and Therapeutic Insights. BLOOD SCIENCE 2022; 4:143-151. [DOI: 10.1097/bs9.0000000000000127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/07/2022] [Indexed: 11/26/2022] Open
|
29
|
Tkach JM, Philip R, Sharma A, Strecker J, Durocher D, Pelletier L. Global cellular response to chemical perturbation of PLK4 activity and abnormal centrosome number. eLife 2022; 11:e73944. [PMID: 35758262 PMCID: PMC9236612 DOI: 10.7554/elife.73944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 06/04/2022] [Indexed: 11/13/2022] Open
Abstract
Centrosomes act as the main microtubule organizing center (MTOC) in metazoans. Centrosome number is tightly regulated by limiting centriole duplication to a single round per cell cycle. This control is achieved by multiple mechanisms, including the regulation of the protein kinase PLK4, the most upstream facilitator of centriole duplication. Altered centrosome numbers in mouse and human cells cause p53-dependent growth arrest through poorly defined mechanisms. Recent work has shown that the E3 ligase TRIM37 is required for cell cycle arrest in acentrosomal cells. To gain additional insights into this process, we undertook a series of genome-wide CRISPR/Cas9 screens to identify factors important for growth arrest triggered by treatment with centrinone B, a selective PLK4 inhibitor. We found that TRIM37 is a key mediator of growth arrest after partial or full PLK4 inhibition. Interestingly, PLK4 cellular mobility decreased in a dose-dependent manner after centrinone B treatment. In contrast to recent work, we found that growth arrest after PLK4 inhibition correlated better with PLK4 activity than with mitotic length or centrosome number. These data provide insights into the global response to changes in centrosome number and PLK4 activity and extend the role for TRIM37 in regulating the abundance, localization, and function of centrosome proteins.
Collapse
Affiliation(s)
- Johnny M Tkach
- Lunenfeld-Tanenbaum Research Institute, Sinai Health SystemTorontoCanada
| | - Reuben Philip
- Lunenfeld-Tanenbaum Research Institute, Sinai Health SystemTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| | - Amit Sharma
- Lunenfeld-Tanenbaum Research Institute, Sinai Health SystemTorontoCanada
| | - Jonathan Strecker
- Lunenfeld-Tanenbaum Research Institute, Sinai Health SystemTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| | - Daniel Durocher
- Lunenfeld-Tanenbaum Research Institute, Sinai Health SystemTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| | - Laurence Pelletier
- Lunenfeld-Tanenbaum Research Institute, Sinai Health SystemTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| |
Collapse
|
30
|
Zhang Z, Zhang W. Epigenetic Signatures of Centrosomes Are Novel Targets in Cancer Diagnosis: Insights from an Analysis of the Cancer Genome Atlas. EPIGENOMES 2022; 6:14. [PMID: 35735471 PMCID: PMC9222712 DOI: 10.3390/epigenomes6020014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/28/2022] [Accepted: 05/29/2022] [Indexed: 02/04/2023] Open
Abstract
The centrosome plays a central role for cellular signaling and is critical for several fundamental cellular processes in human cells. Centrosome abnormalities have been linked to multiple solid tumors and hematological malignancies. We sought to explore the potential role of the DNA methylation, a critical epigenetic modification, of centrosome-related genes in different cancers. The 450K array DNA methylation data and RNA-seq data were downloaded for ~4000 tumor samples and ~500 normal controls from The Cancer Genome Atlas (TCGA) project, covering 11 major cancer types. Cancers with more than 30 normal controls were retained for analysis. Differentially modified CpGs of centrosome genes were identified, and cancer-specific epigenetic models were developed using a machine-learning algorithm for each cancer type. The association between the methylation level of differential CpGs and the corresponding gene expression, as well as the co-localization of the differential CpGs and cis-regulatory elements were evaluated. In total, 2761 CpGs located on 160 centrosome genes for 6 cancers were included in the analysis. Cancer-specific models demonstrated a high accuracy in terms of the area under the receiver operating characteristic (ROC) curve (AUC > 0.9) in five cancers and showed tissue specificity. This study enhanced our understanding of the epigenetic mechanisms underlying the DNA methylation of centrosome-related genes in cancers, and showed the potential of these epigenetic modifications as novel cancer biomarkers.
Collapse
Affiliation(s)
- Zhou Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
- The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
31
|
Sánchez-Arcila JC, Jensen KDC. Forward Genetics in Apicomplexa Biology: The Host Side of the Story. Front Cell Infect Microbiol 2022; 12:878475. [PMID: 35646724 PMCID: PMC9133346 DOI: 10.3389/fcimb.2022.878475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Forward genetic approaches have been widely used in parasitology and have proven their power to reveal the complexities of host-parasite interactions in an unbiased fashion. Many aspects of the parasite's biology, including the identification of virulence factors, replication determinants, antibiotic resistance genes, and other factors required for parasitic life, have been discovered using such strategies. Forward genetic approaches have also been employed to understand host resistance mechanisms to parasitic infection. Here, we will introduce and review all forward genetic approaches that have been used to identify host factors involved with Apicomplexa infections, which include classical genetic screens and QTL mapping, GWAS, ENU mutagenesis, overexpression, RNAi and CRISPR-Cas9 library screens. Collectively, these screens have improved our understanding of host resistance mechanisms, immune regulation, vaccine and drug designs for Apicomplexa parasites. We will also discuss how recent advances in molecular genetics give present opportunities to further explore host-parasite relationships.
Collapse
Affiliation(s)
- Juan C. Sánchez-Arcila
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
| | - Kirk D. C. Jensen
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
- Health Science Research Institute, University of California, Merced, Merced, CA, United States
| |
Collapse
|
32
|
Structural convergence for tubulin binding of CPAP and vinca domain microtubule inhibitors. Proc Natl Acad Sci U S A 2022; 119:e2120098119. [PMID: 35507869 PMCID: PMC9171608 DOI: 10.1073/pnas.2120098119] [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] [Indexed: 11/23/2022] Open
Abstract
Microtubules are dynamic assemblies of αβ-tubulin that are involved in key cellular functions, including cell division and intracellular transport. Microtubule dynamics is inhibited by several families of small molecules, some of which are used in oncology. The extent to which these compounds target the binding sites of cellular partners of tubulin remains poorly characterized. We show here that a region of the CPAP protein binds to the so-called vinca domain of β-tubulin in a way very similar to that of peptide-like inhibitors produced by bacteria and fungi. Therefore, our work identifies a structural convergence for tubulin binding between inhibitors and a regulator of microtubule dynamics. Microtubule dynamics is regulated by various cellular proteins and perturbed by small-molecule compounds. To what extent the mechanism of the former resembles that of the latter is an open question. We report here structures of tubulin bound to the PN2-3 domain of CPAP, a protein controlling the length of the centrioles. We show that an α-helix of the PN2-3 N-terminal region binds and caps the longitudinal surface of the tubulin β subunit. Moreover, a PN2-3 N-terminal stretch lies in a β-tubulin site also targeted by fungal and bacterial peptide-like inhibitors of the vinca domain, sharing a very similar binding mode with these compounds. Therefore, our results identify several characteristic features of cellular partners that bind to this site and highlight a structural convergence of CPAP with small-molecule inhibitors of microtubule assembly.
Collapse
|
33
|
Vásquez-Limeta A, Lukasik K, Kong D, Sullenberger C, Luvsanjav D, Sahabandu N, Chari R, Loncarek J. CPAP insufficiency leads to incomplete centrioles that duplicate but fragment. J Cell Biol 2022; 221:213119. [PMID: 35404385 PMCID: PMC9007748 DOI: 10.1083/jcb.202108018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/13/2022] [Accepted: 02/28/2022] [Indexed: 11/22/2022] Open
Abstract
Centrioles are structures that assemble centrosomes. CPAP is critical for centrosome assembly, and its mutations are found in patients with diseases such as primary microcephaly. CPAP’s centrosomal localization, its dynamics, and the consequences of its insufficiency in human cells are poorly understood. Here we use human cells genetically engineered for fast degradation of CPAP, in combination with superresolution microscopy, to address these uncertainties. We show that three independent centrosomal CPAP populations are dynamically regulated during the cell cycle. We confirm that CPAP is critical for assembly of human centrioles, but not for recruitment of pericentriolar material on already assembled centrioles. Further, we reveal that CPAP insufficiency leads to centrioles with incomplete microtubule triplets that can convert to centrosomes, duplicate, and form mitotic spindle poles, but fragment owing to loss of cohesion between microtubule blades. These findings further our basic understanding of the role of CPAP in centrosome biogenesis and help understand how CPAP aberrations can lead to human diseases.
Collapse
Affiliation(s)
- Alejandra Vásquez-Limeta
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Kimberly Lukasik
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Dong Kong
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Catherine Sullenberger
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Delgermaa Luvsanjav
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Natalie Sahabandu
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| | - Raj Chari
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Jadranka Loncarek
- Laboratory of Protein Dynamics and Signaling, National Institutes of Health, National Cancer Institute, Center for Cancer Research, Frederick, MD
| |
Collapse
|
34
|
Takumi K, Kitagawa D. Experimental and Natural Induction of de novo Centriole Formation. Front Cell Dev Biol 2022; 10:861864. [PMID: 35445021 PMCID: PMC9014216 DOI: 10.3389/fcell.2022.861864] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/14/2022] [Indexed: 01/11/2023] Open
Abstract
In cycling cells, new centrioles are assembled in the vicinity of pre-existing centrioles. Although this canonical centriole duplication is a tightly regulated process in animal cells, centrioles can also form in the absence of pre-existing centrioles; this process is termed de novo centriole formation. De novo centriole formation is triggered by the removal of all pre-existing centrioles in the cell in various manners. Moreover, overexpression of polo-like kinase 4 (Plk4), a master regulatory kinase for centriole biogenesis, can induce de novo centriole formation in some cell types. Under these conditions, structurally and functionally normal centrioles can be formed de novo. While de novo centriole formation is normally suppressed in cells with intact centrioles, depletion of certain suppressor proteins leads to the ectopic formation of centriole-related protein aggregates in the cytoplasm. It has been shown that de novo centriole formation also occurs naturally in some species. For instance, during the multiciliogenesis of vertebrate epithelial cells, massive de novo centriole amplification occurs to form numerous motile cilia. In this review, we summarize the previous findings on de novo centriole formation, particularly under experimental conditions, and discuss its regulatory mechanisms.
Collapse
Affiliation(s)
- Kasuga Takumi
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| | - Daiju Kitagawa
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
35
|
Aggresome assembly at the centrosome is driven by CP110–CEP97–CEP290 and centriolar satellites. Nat Cell Biol 2022; 24:483-496. [PMID: 35411088 PMCID: PMC9033585 DOI: 10.1038/s41556-022-00869-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/10/2022] [Indexed: 12/30/2022]
Abstract
Protein degradation is critical to maintaining cellular homeostasis, and perturbation of the ubiquitin proteasome system leads to the accumulation of protein aggregates. These aggregates are either directed towards autophagy for destruction or sequestered into an inclusion, termed the aggresome, at the centrosome. Utilizing high-resolution quantitative analysis, here, we define aggresome assembly at the centrosome in human cells. Centriolar satellites are proteinaceous granules implicated in the trafficking of proteins to the centrosome. During aggresome assembly, satellites were required for the growth of the aggresomal structure from an initial ring of phosphorylated HSP27 deposited around the centrioles. The seeding of this phosphorylated HSP27 ring depended on the centrosomal proteins CP110, CEP97 and CEP290. Owing to limiting amounts of CP110, senescent cells, which are characterized by the accumulation of protein aggregates, were defective in aggresome formation. Furthermore, satellites and CP110–CEP97–CEP290 were required for the aggregation of mutant huntingtin. Together, these data reveal roles for CP110–CEP97–CEP290 and satellites in the control of cellular proteostasis and the aggregation of disease-relevant proteins. Prosser et al. report that centriolar satellite and centrosomal proteins seed aggresomes, perinuclear inclusions of misfolded proteins, and may play a role in aggresome formation during senescence and huntingtin aggregation.
Collapse
|
36
|
An HL, Kuo HC, Tang TK. Modeling Human Primary Microcephaly With hiPSC-Derived Brain Organoids Carrying CPAP-E1235V Disease-Associated Mutant Protein. Front Cell Dev Biol 2022; 10:830432. [PMID: 35309908 PMCID: PMC8924525 DOI: 10.3389/fcell.2022.830432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
The centrosome is composed of a pair of centrioles and serves as the major microtubule-organizing center (MTOC) in cells. Centrosome dysfunction has been linked to autosomal recessive primary microcephaly (MCPH), which is a rare human neurodevelopmental disorder characterized by small brain size with intellectual disability. Recently, several mouse models carrying mutated genes encoding centrosomal proteins have been generated to address the genotype-phenotype relationships in MCPH. However, several human-specific features were not observed in the mouse models during brain development. Herein, we generated isogenic hiPSCs carrying the gene encoding centrosomal CPAP-E1235V mutant protein using the CRISPR-Cas9 genome editing system, and examined the phenotypic features of wild-type and mutant hiPSCs and their derived brain organoids. Our results showed that the CPAP-E1235V mutant perturbed the recruitment of several centriolar proteins involved in centriole elongation, including CEP120, CEP295, CENTROBIN, POC5, and POC1B, onto nascent centrioles, resulting in the production of short centrioles but long cilia. Importantly, our wild-type hiPSC-derived brain organoid recapitulated many cellular events seen in the developing human brain, including neuronal differentiation and cortical spatial lamination. Interestingly, hiPSC-CPAP-E1235V-derived brain organoids induced p53-dependent neuronal cell death, resulting in the production of smaller brain organoids that mimic the microcephaly phenotype. Furthermore, we observed that the CPAP-E1235V mutation altered the spindle orientation of neuronal progenitor cells and induced premature neuronal differentiation. In summary, we have shown that the hiPSC-derived brain organoid coupled with CRISPR/Cas9 gene editing technology can recapitulate the centrosome/centriole-associated MCPH pathological features. Possible mechanisms for MCPH with centriole/centrosome dysfunction are discussed.
Collapse
Affiliation(s)
- Hsiao-Lung An
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hung-Chih Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Tang K Tang
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
37
|
Estrogens—Origin of Centrosome Defects in Human Cancer? Cells 2022; 11:cells11030432. [PMID: 35159242 PMCID: PMC8833882 DOI: 10.3390/cells11030432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/22/2022] Open
Abstract
Estrogens are associated with a variety of diseases and play important roles in tumor development and progression. Centrosome defects are hallmarks of human cancers and contribute to ongoing chromosome missegragation and aneuploidy that manifest in genomic instability and tumor progression. Although several mechanisms underlie the etiology of centrosome aberrations in human cancer, upstream regulators are hardly known. Accumulating experimental and clinical evidence points to an important role of estrogens in deregulating centrosome homeostasis and promoting karyotype instability. Here, we will summarize existing literature of how natural and synthetic estrogens might contribute to structural and numerical centrosome defects, genomic instability and human carcinogenesis.
Collapse
|
38
|
Keep Calm and Carry on with Extra Centrosomes. Cancers (Basel) 2022; 14:cancers14020442. [PMID: 35053604 PMCID: PMC8774008 DOI: 10.3390/cancers14020442] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/01/2022] [Accepted: 01/03/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Precise chromosome segregation during mitosis is a vital event orchestrated by formation of bipolar spindle poles. Supernumerary centrosomes, caused by centrosome amplification, deteriorates mitotic processes, resulting in segregation defects leading to chromosomal instability (CIN). Centrosome amplification is frequently observed in various types of cancer and considered as a significant contributor to destabilization of chromosomes. This review provides a comprehensive overview of causes and consequences of centrosome amplification thoroughly describing molecular mechanisms. Abstract Aberrations in the centrosome number and structure can readily be detected at all stages of tumor progression and are considered hallmarks of cancer. Centrosome anomalies are closely linked to chromosome instability and, therefore, are proposed to be one of the driving events of tumor formation and progression. This concept, first posited by Boveri over 100 years ago, has been an area of interest to cancer researchers. We have now begun to understand the processes by which these numerical and structural anomalies may lead to cancer, and vice-versa: how key events that occur during carcinogenesis could lead to amplification of centrosomes. Despite the proliferative advantages that having extra centrosomes may confer, their presence can also lead to loss of essential genetic material as a result of segregational errors and cancer cells must deal with these deadly consequences. Here, we review recent advances in the current literature describing the mechanisms by which cancer cells amplify their centrosomes and the methods they employ to tolerate the presence of these anomalies, focusing particularly on centrosomal clustering.
Collapse
|
39
|
Shin B, Kim MS, Lee Y, Jung GI, Rhee K. Generation and Fates of Supernumerary Centrioles in Dividing Cells. Mol Cells 2021; 44:699-705. [PMID: 34711687 PMCID: PMC8560585 DOI: 10.14348/molcells.2021.0220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/25/2021] [Accepted: 09/30/2021] [Indexed: 11/27/2022] Open
Abstract
The centrosome is a subcellular organelle from which a cilium assembles. Since centrosomes function as spindle poles during mitosis, they have to be present as a pair in a cell. How the correct number of centrosomes is maintained in a cell has been a major issue in the fields of cell cycle and cancer biology. Centrioles, the core of centrosomes, assemble and segregate in close connection to the cell cycle. Abnormalities in centriole numbers are attributed to decoupling from cell cycle regulation. Interestingly, supernumerary centrioles are commonly observed in cancer cells. In this review, we discuss how supernumerary centrioles are generated in diverse cellular conditions. We also discuss how the cells cope with supernumerary centrioles during the cell cycle.
Collapse
Affiliation(s)
- Byungho Shin
- Department of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Myung Se Kim
- Department of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Yejoo Lee
- Department of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Gee In Jung
- Department of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Kunsoo Rhee
- Department of Biological Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
40
|
Jung GI, Rhee K. Triple deletion of TP53, PCNT, and CEP215 promotes centriole amplification in the M phase. Cell Cycle 2021; 20:1500-1517. [PMID: 34233584 DOI: 10.1080/15384101.2021.1950386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Supernumerary centrioles are frequently observed in diverse types of cancer cells. In this study, we investigated the mechanism underlying the generation of supernumerary centrioles during the M phase. We generated the TP53;PCNT;CEP215 triple knockout (KO) cells and determined the configurations of the centriole during the cell cycle. The triple KO cells exhibited a precocious separation of centrioles and unscheduled centriole assembly in the M phase. Supernumerary centrioles in the triple KO cells were present throughout the cell cycle; however, among all the centrioles, only two maintained an intact composition, including CEP135, CEP192, CEP295 and CEP152. Intact centrioles were formed during the S phase and the rest of the centrioles may be generated during the M phase. M-phase-assembled centrioles lacked the ability to organize microtubules in the interphase; however, a fraction of them may acquire pericentriolar material to organize microtubules during the M phase. Taken together, our work reveals the heterogeneity of the supernumerary centrioles in the triple KO cells. .
Collapse
Affiliation(s)
- Gee In Jung
- Department of Biological Sciences, Seoul National University, Seoul, Korea
| | - Kunsoo Rhee
- Department of Biological Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
41
|
Gudi R, Palanisamy V, Vasu C. Centrosomal P4.1-associated protein (CPAP) positively regulates endocytic vesicular transport and lysosome targeting of EGFR. Sci Rep 2021; 11:12689. [PMID: 34135376 PMCID: PMC8209166 DOI: 10.1038/s41598-021-91818-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/01/2021] [Indexed: 11/09/2022] Open
Abstract
Centrosomal P4.1-associated protein (CPAP) plays a critical role in restricting the centriole length in human cells. Here, we report a novel, positive regulatory influence for CPAP on endocytic vesicular transport (EVT) and lysosome targeting of internalized-cell surface receptor EGFR. We observed that higher CPAP levels cause an increase in the abundance of multi-vesicular body (MVB) and EGFR is detectable in CPAP-overexpression induced puncta. The surface and cellular levels of EGFR are higher under CPAP deficiency and lower under CPAP overexpression. While ligand-engagement induced internalization or routing of EGFR into early endosomes is not influenced by cellular levels of CPAP, we found that targeting of ligand-activated, internalized EGFR to lysosome is impacted by CPAP levels. Transport of ligand-bound EGFR from early endosome to late endosome/MVB and lysosome is diminished in CPAP-depleted cells. Moreover, CPAP depleted cells appear to show a diminished ability to form MVB structures upon EGFR activation. These observations suggest a positive regulatory effect of CPAP on EVT of ligand-bound EGFR-like cell surface receptors to MVB and lysosome. Overall, identification of a non-centriolar function of CPAP in endocytic trafficking provides new insights in understanding the non-canonical cellular functions of CPAP.
Collapse
Affiliation(s)
- Radhika Gudi
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA.
| | - Viswanathan Palanisamy
- Department of Biochemistry, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Chenthamarakshan Vasu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
42
|
Human Microcephaly Protein RTTN Is Required for Proper Mitotic Progression and Correct Spindle Position. Cells 2021; 10:cells10061441. [PMID: 34207628 PMCID: PMC8229632 DOI: 10.3390/cells10061441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 01/16/2023] Open
Abstract
Autosomal recessive primary microcephaly (MCPH) is a complex neurodevelopmental disorder characterized by a small brain size with mild to moderate intellectual disability. We previously demonstrated that human microcephaly RTTN played an important role in regulating centriole duplication during interphase, but the role of RTTN in mitosis is not fully understood. Here, we show that RTTN is required for normal mitotic progression and correct spindle position. The depletion of RTTN induces the dispersion of the pericentriolar protein γ-tubulin and multiple mitotic abnormalities, including monopolar, abnormal bipolar, and multipolar spindles. Importantly, the loss of RTTN altered NuMA/p150Glued congression to the spindle poles, perturbed NuMA cortical localization, and reduced the number and the length of astral microtubules. Together, our results provide a new insight into how RTTN functions in mitosis.
Collapse
|
43
|
Doornbos C, Roepman R. Moonlighting of mitotic regulators in cilium disassembly. Cell Mol Life Sci 2021; 78:4955-4972. [PMID: 33860332 PMCID: PMC8233288 DOI: 10.1007/s00018-021-03827-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/03/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023]
Abstract
Correct timing of cellular processes is essential during embryological development and to maintain the balance between healthy proliferation and tumour formation. Assembly and disassembly of the primary cilium, the cell’s sensory signalling organelle, are linked to cell cycle timing in the same manner as spindle pole assembly and chromosome segregation. Mitotic processes, ciliary assembly, and ciliary disassembly depend on the centrioles as microtubule-organizing centres (MTOC) to regulate polymerizing and depolymerizing microtubules. Subsequently, other functional protein modules are gathered to potentiate specific protein–protein interactions. In this review, we show that a significant subset of key mitotic regulator proteins is moonlighting at the cilium, among which PLK1, AURKA, CDC20, and their regulators. Although ciliary assembly defects are linked to a variety of ciliopathies, ciliary disassembly defects are more often linked to brain development and tumour formation. Acquiring a better understanding of the overlap in regulators of ciliary disassembly and mitosis is essential in finding therapeutic targets for the different diseases and types of tumours associated with these regulators.
Collapse
Affiliation(s)
- Cenna Doornbos
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ronald Roepman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands. .,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
44
|
Gudi RR, Janakiraman H, Howe PH, Palanisamy V, Vasu C. Loss of CPAP causes sustained EGFR signaling and epithelial-mesenchymal transition in oral cancer. Oncotarget 2021; 12:807-822. [PMID: 33889303 PMCID: PMC8057274 DOI: 10.18632/oncotarget.27932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 03/22/2021] [Indexed: 11/25/2022] Open
Abstract
Higher epidermal growth factor receptor (EGFR) signaling can contribute to tumor metastasis and resistance to therapies in oral squamous cell carcinoma (OSCC). EGFR signaling can promote epithelial-mesenchymal transition (EMT) in OSCC. EMT is a process by which epithelial cells acquire invasive properties and it can contribute to tumor metastasis. Not only do the abnormal functions of microtubule and microtubule-organizing centers (MTOC) such as centrosomes lead to cancers, but also the malignant tissues are characterized by aberrant centriolar features and amplified centrosomes. Microtubule inhibition therapies increase the sensitivity to EGFR targeting drugs in various cancers. In this study, we show that the loss of expression of a microtubule/tubulin binding protein, centrosomal protein 4.1-associated protein (CPAP), which is critical for centriole biogenesis and normal functioning of the centrosome, caused an increase in the EGFR levels and its signaling and, enhanced the EMT features and invasiveness of OSCC cells. Further, depletion of CPAP enhanced the tumorigenicity of these cells in a xeno-transplant model. Importantly, CPAP loss-associated EMT features and invasiveness of multiple OSCC cells were attenuated upon depletion of EGFR in them. On the other hand, we found that CPAP protein levels were higher in EGF treated OSCC cells as well as in oral cancer tissues, suggesting that the frequently reported aberrant centriolar features of tumors are potentially a consequence, but not the cause, of tumor progression. Overall, our novel observations show that, in addition to its known indispensable role in centrosome biogenesis, CPAP also plays a vital role in suppressing tumorigenesis in OSCC by facilitating EGFR homeostasis.
Collapse
Affiliation(s)
- Radhika R Gudi
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | - Philip H Howe
- Department of Biochemistry, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Viswanathan Palanisamy
- Department of Biochemistry, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Chenthamarakshan Vasu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
45
|
Fatalska A, Stepinac E, Richter M, Kovacs L, Pietras Z, Puchinger M, Dong G, Dadlez M, Glover DM. The dimeric Golgi protein Gorab binds to Sas6 as a monomer to mediate centriole duplication. eLife 2021; 10:e57241. [PMID: 33704067 PMCID: PMC8009671 DOI: 10.7554/elife.57241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 03/11/2021] [Indexed: 12/30/2022] Open
Abstract
The duplication and ninefold symmetry of the Drosophila centriole requires that the cartwheel molecule, Sas6, physically associates with Gorab, a trans-Golgi component. How Gorab achieves these disparate associations is unclear. Here, we use hydrogen-deuterium exchange mass spectrometry to define Gorab's interacting surfaces that mediate its subcellular localization. We identify a core stabilization sequence within Gorab's C-terminal coiled-coil domain that enables homodimerization, binding to Rab6, and thereby trans-Golgi localization. By contrast, part of the Gorab monomer's coiled-coil domain undergoes an antiparallel interaction with a segment of the parallel coiled-coil dimer of Sas6. This stable heterotrimeric complex can be visualized by electron microscopy. Mutation of a single leucine residue in Sas6's Gorab-binding domain generates a Sas6 variant with a sixteenfold reduced binding affinity for Gorab that cannot support centriole duplication. Thus, Gorab dimers at the Golgi exist in equilibrium with Sas6-associated monomers at the centriole to balance Gorab's dual role.
Collapse
Affiliation(s)
- Agnieszka Fatalska
- Department of Genetics, University of CambridgeCambridgeUnited Kingdom
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
- Institute of Biochemistry and Biophysics, Polish Academy of SciencesWarsawPoland
| | - Emma Stepinac
- Department of Medical Biochemistry, Max Perutz Labs, Medical University of ViennaViennaAustria
| | - Magdalena Richter
- Department of Genetics, University of CambridgeCambridgeUnited Kingdom
| | - Levente Kovacs
- Department of Genetics, University of CambridgeCambridgeUnited Kingdom
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| | - Zbigniew Pietras
- Institute of Biochemistry and Biophysics, Polish Academy of SciencesWarsawPoland
| | - Martin Puchinger
- Department of Structural and Computational Biology, Max Perutz Labs, University of ViennaViennaAustria
| | - Gang Dong
- Department of Medical Biochemistry, Max Perutz Labs, Medical University of ViennaViennaAustria
| | - Michal Dadlez
- Institute of Biochemistry and Biophysics, Polish Academy of SciencesWarsawPoland
| | - David M Glover
- Department of Genetics, University of CambridgeCambridgeUnited Kingdom
- Division of Biology and Biological Engineering, California Institute of TechnologyPasadenaUnited States
| |
Collapse
|
46
|
Balestra FR, Domínguez-Calvo A, Wolf B, Busso C, Buff A, Averink T, Lipsanen-Nyman M, Huertas P, Ríos RM, Gönczy P. TRIM37 prevents formation of centriolar protein assemblies by regulating Centrobin. eLife 2021; 10:62640. [PMID: 33491649 PMCID: PMC7870141 DOI: 10.7554/elife.62640] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/22/2021] [Indexed: 12/17/2022] Open
Abstract
TRIM37 is an E3 ubiquitin ligase mutated in Mulibrey nanism, a disease with impaired organ growth and increased tumor formation. TRIM37 depletion from tissue culture cells results in supernumerary foci bearing the centriolar protein Centrin. Here, we characterize these centriolar protein assemblies (Cenpas) to uncover the mechanism of action of TRIM37. We find that an atypical de novo assembly pathway can generate Cenpas that act as microtubule-organizing centers (MTOCs), including in Mulibrey patient cells. Correlative light electron microscopy reveals that Cenpas are centriole-related or electron-dense structures with stripes. TRIM37 regulates the stability and solubility of Centrobin, which accumulates in elongated entities resembling the striped electron dense structures upon TRIM37 depletion. Furthermore, Cenpas formation upon TRIM37 depletion requires PLK4, as well as two parallel pathways relying respectively on Centrobin and PLK1. Overall, our work uncovers how TRIM37 prevents Cenpas formation, which would otherwise threaten genome integrity.
Collapse
Affiliation(s)
- Fernando R Balestra
- Departamento de Genética, Universidad de Sevilla, Sevilla, Spain.,Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Andrés Domínguez-Calvo
- Departamento de Genética, Universidad de Sevilla, Sevilla, Spain.,Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Benita Wolf
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Coralie Busso
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Alizée Buff
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Tessa Averink
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Marita Lipsanen-Nyman
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pablo Huertas
- Departamento de Genética, Universidad de Sevilla, Sevilla, Spain.,Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Rosa M Ríos
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
47
|
Sharma A, Olieric N, Steinmetz MO. Centriole length control. Curr Opin Struct Biol 2020; 66:89-95. [PMID: 33220554 DOI: 10.1016/j.sbi.2020.10.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/10/2020] [Accepted: 10/13/2020] [Indexed: 10/22/2022]
Abstract
Centrioles are microtubule-based structures involved in cell division and ciliogenesis. Centriole formation is a highly regulated cellular process and aberrations in centriole structure, size or numbers have implications in multiple human pathologies. In this review, we propose that the proteins that control centriole length can be subdivided into two classes based on their antagonistic activities on centriolar microtubules, which we refer to as 'centriole elongation activators' (CEAs) and 'centriole elongation inhibitors' (CEIs). We discuss and illustrate the structure-function relationship of CEAs and CEIs as well as their interaction networks. Based on our current knowledge, we formulate some outstanding open questions in the field and present possible routes for future studies.
Collapse
Affiliation(s)
- Ashwani Sharma
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, CH-5232 Villigen, Switzerland.
| | - Natacha Olieric
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, CH-5232 Villigen, Switzerland.
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, CH-5232 Villigen, Switzerland; University of Basel, Biozentrum, CH-4056 Basel, Switzerland.
| |
Collapse
|
48
|
Fatalska A, Dzhindzhev NS, Dadlez M, Glover DM. Interaction interface in the C-terminal parts of centriole proteins Sas6 and Ana2. Open Biol 2020; 10:200221. [PMID: 33171067 PMCID: PMC7729032 DOI: 10.1098/rsob.200221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022] Open
Abstract
The centriole is a ninefold symmetrical structure found at the core of centrosomes and, as a basal body, at the base of cilia, whose conserved duplication is regulated by Plk4 kinase. Plk4 phosphorylates a single serine residue at the N-terminus of Ana2 to promote Ana2's loading to the site of procentriole formation. Four conserved serines in Ana2's STAN motif are then phosphorylated by Plk4, enabling Sas6 recruitment. Crystallographic data indicate that the coiled-coil domain of Ana2 forms a tetramer but the structure of full-length Ana2 has not been solved. Here, we have employed hydrogen-deuterium exchange coupled with mass spectrometry (HDX-MS) to uncover the conformational dynamics of Ana2, revealing the high flexibility of this protein with one rigid region. To determine the elusive nature of the interaction surfaces between Ana2 and Sas6, we have confirmed complex formation between the phosphomimetic form of Ana2 (Ana2-4D) and Sas6 in vitro and in vivo. Analysis of this complex by HDX-MS identifies short critical regions required for this interaction, which lie in the C-terminal parts of both proteins. Mutational studies confirmed the relevance of these regions for the Ana2-Sas6 interaction. The Sas6 site required for Ana2 binding is distinct from the site required for Sas6 to bind Gorab and Sas6 is able to bind both these protein partners simultaneously.
Collapse
Affiliation(s)
- Agnieszka Fatalska
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | | | - Michal Dadlez
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - David M. Glover
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
49
|
Yamazoe T, Nagai T, Umeda S, Sugaya Y, Mizuno K. Roles of TOG and jelly-roll domains of centrosomal protein CEP104 in its functions in cilium elongation and Hedgehog signaling. J Biol Chem 2020; 295:14723-14736. [PMID: 32820051 DOI: 10.1074/jbc.ra120.013334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/11/2020] [Indexed: 12/26/2022] Open
Abstract
Primary cilia are generated through the extension of the microtubule-based axoneme. Centrosomal protein 104 (CEP104) localizes to the tip of the elongating axoneme, and CEP104 mutations are linked to a ciliopathy, Joubert syndrome. Thus, CEP104 has been implicated in ciliogenesis. However, the mechanism by which CEP104 regulates ciliogenesis remains elusive. We report here that CEP104 is critical for cilium elongation but not for initiating ciliogenesis. We also demonstrated that the tumor-overexpressed gene (TOG) domain of CEP104 exhibits microtubule-polymerizing activity and that this activity is essential for the cilium-elongating activity of CEP104. Knockdown/rescue experiments showed that the N-terminal jelly-roll (JR) fold partially contributes to cilium-elongating activity of CEP104, but neither the zinc-finger region nor the SXIP motif is required for this activity. CEP104 binds to a centriole-capping protein, CP110, through the zinc-finger region and to a microtubule plus-end-binding protein, EB1, through the SXIP motif, indicating that the binding of CP110 and EB1 is dispensable for the cilium-elongating activity of CEP104. Moreover, CEP104 depletion does not affect CP110 removal from the mother centriole, which suggests that CEP104 functions after the removal of CP110. Last, we also showed that CEP104 is required for the ciliary entry of Smoothened and export of GPR161 upon Hedgehog signal activation and that the TOG domain plays a critical role in this activity. Our results define the roles of the individual domains of CEP104 in its functions in cilium elongation and Hedgehog signaling and should enhance our understanding of the mechanism underlying CEP104 mutation-associated ciliopathies.
Collapse
Affiliation(s)
- Takashi Yamazoe
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Tomoaki Nagai
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan; Department of Biochemistry, Fukushima Medical University School of Medicine, Fukushima, Japan.
| | - Shinya Umeda
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Yuko Sugaya
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Kensaku Mizuno
- Department of Molecular and Chemical Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan; Institute of Liberal Arts and Sciences, Tohoku University, Kawauchi, Sendai, Miyagi, Japan.
| |
Collapse
|
50
|
Principal Postulates of Centrosomal Biology. Version 2020. Cells 2020; 9:cells9102156. [PMID: 32987651 PMCID: PMC7598677 DOI: 10.3390/cells9102156] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022] Open
Abstract
The centrosome, which consists of two centrioles surrounded by pericentriolar material, is a unique structure that has retained its main features in organisms of various taxonomic groups from unicellular algae to mammals over one billion years of evolution. In addition to the most noticeable function of organizing the microtubule system in mitosis and interphase, the centrosome performs many other cell functions. In particular, centrioles are the basis for the formation of sensitive primary cilia and motile cilia and flagella. Another principal function of centrosomes is the concentration in one place of regulatory proteins responsible for the cell's progression along the cell cycle. Despite the existing exceptions, the functioning of the centrosome is subject to general principles, which are discussed in this review.
Collapse
|