1
|
Tsai YC, Kuo TN, Chao YY, Lin RC, Chien HH, Peng IT, Tsai YF, Su PJ, Wang CY. Pericentriolar material 1 aggregation maintains cell survival upon prolonged replication stress. Arch Biochem Biophys 2025; 768:110383. [PMID: 40090437 DOI: 10.1016/j.abb.2025.110383] [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: 08/02/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025]
Abstract
The centrosome is essential for maintaining cell shape and facilitating cell division. Thus, precise control of centrosome copy numbers is crucial for proper chromosome segregation. Pericentriolar material 1 (PCM1) is a scaffold component of centriolar satellites-electron-dense granules dispersed around the centrosome-that regulate the centrosome or primary cilia. It has been shown that disrupting PCM1 aggregation by treating cells with sodium orthovanadate inhibits centrosome amplification. However, sodium orthovanadate is a protein tyrosine phosphatase and may have off-target effects on the centrosome. To further confirm the role of PCM1 aggregation in promoting centrosome amplification, we disrupted PCM1 aggregation by interfering with microtubule networks, inhibiting the dynactin motor complex, or depleting PCM1. Centrosome copy numbers were then examined under conditions of prolonged replication stress. Our data suggest that PCM1 aggregation does not promote centrosome amplification in osteosarcoma U2-OS or pancreatic ductal adenocarcinoma PANC-1 cell lines. Instead, we found that centrosome amplification promoted PCM1 aggregation in a PLK4-dependent manner. Furthermore, we observed that PCM1 depletion inhibited U2-OS cell survival under prolonged replication stress. Prolonged replication stress induced DNA damage signaling via the ATM-CHK1 axis and autophagy to maintain cell survival, while PCM1 depletion alleviated ATM, CHK1, and autophagy activity, thereby reducing cell survival. Our findings propose that PCM1 does not facilitate centrosome amplification but instead induces activation of the ATM-CHK1 axis and autophagy to sustain osteosarcoma cell viability during prolonged replication stress.
Collapse
Affiliation(s)
- Yung-Chieh Tsai
- Department of Obstetrics and Gynecology, Chi-Mei Medical Center, Tainan, Taiwan; Department of Sport Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Tian-Ni Kuo
- Department of Obstetrics and Gynecology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yu-Ying Chao
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ruei-Ci Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Han-Hsiang Chien
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - I-Ting Peng
- Department of Obstetrics and Gynecology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yueh-Fong Tsai
- Department of Obstetrics and Gynecology, An Nan Hospital, China Medical University, Tainan, Taiwan.
| | - Ping-Jui Su
- Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan.
| | - Chia-Yih Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
2
|
Begar E, Seyrek E, Firat‐Karalar EN. Navigating centriolar satellites: the role of PCM1 in cellular and organismal processes. FEBS J 2025; 292:688-708. [PMID: 38825736 PMCID: PMC11839937 DOI: 10.1111/febs.17194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/20/2024] [Accepted: 05/22/2024] [Indexed: 06/04/2024]
Abstract
Centriolar satellites are ubiquitous membrane-less organelles that play critical roles in numerous cellular and organismal processes. They were initially discovered through electron microscopy as cytoplasmic granules surrounding centrosomes in vertebrate cells. These structures remained enigmatic until the identification of pericentriolar material 1 protein (PCM1) as their molecular marker, which has enabled their in-depth characterization. Recently, centriolar satellites have come into the spotlight due to their links to developmental and neurodegenerative disorders. This review presents a comprehensive summary of the major advances in centriolar satellite biology, with a focus on studies that investigated their biology associated with the essential scaffolding protein PCM1. We begin by exploring the molecular, cellular, and biochemical properties of centriolar satellites, laying the groundwork for a deeper understanding of their functions and mechanisms at both cellular and organismal levels. We then examine the implications of their dysregulation in various diseases, particularly highlighting their emerging roles in neurodegenerative and developmental disorders, as revealed by organismal models of PCM1. We conclude by discussing the current state of knowledge and posing questions about the adaptable nature of these organelles, thereby setting the stage for future research.
Collapse
Affiliation(s)
- Efe Begar
- Department of Molecular Biology and GeneticsKoç UniversityIstanbulTurkey
| | - Ece Seyrek
- Department of Molecular Biology and GeneticsKoç UniversityIstanbulTurkey
| | - Elif Nur Firat‐Karalar
- Department of Molecular Biology and GeneticsKoç UniversityIstanbulTurkey
- School of MedicineKoç UniversityIstanbulTurkey
| |
Collapse
|
3
|
Agostini L, Pfister J, Basnet N, Ding J, Zhang R, Biertümpfel C, O'Connell KF, Mizuno N. Structural insights into SSNA1 self-assembly and its microtubule binding for centriole maintenance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623454. [PMID: 39803484 PMCID: PMC11722292 DOI: 10.1101/2024.11.13.623454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
SSNA-1 is a fibrillar protein localized at the area where dynamic microtubule remodeling occurs including centrosomes. Despite the important activities of SSNA1 to microtubules such as nucleation, co-polymerization, and lattice sharing microtubule branching, the underlying molecular mechanism have remained unclear due to a lack of structural information. Here, we determined the cryo-EM structure of C. elegans SSNA-1 at 4.55 Å resolution and evaluated its role during embryonic development in C. elegans. We found that SSNA1 forms an anti-parallel coiled-coil, and its self-assembly is facilitated by the overhangs of 16 residues at its C-terminus, which dock on the adjacent coiled-coil to form a triple-stranded helical junction. Notably, the microtubule-binding region is within the triple-stranded junction, highlighting that self-assembly of SSNA-1 facilitates effective microtubule interaction by creating hubs along a fibril. Furthermore, our genetical analysis elucidated that deletion of SSNA-1 resulted in a significant reduction in embryonic viability and the formation of multipolar spindles during cell division. Interestingly, when the ability of SSNA-1 self-assembly was impaired, embryonic viability stayed low, comparable to that of the knockout strain. Our study provides molecular insights into the self-assembly mechanisms of SSNA-1, shedding light on its role in controlling microtubule binding and cell division through the regulation of centriole stability.
Collapse
Affiliation(s)
- Lorenzo Agostini
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Dr., Bethesda, MD, 20892, USA
| | - Jason Pfister
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 8 Center Dr., Bethesda, MD, 20892, USA
| | - Nirakar Basnet
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Dr., Bethesda, MD, 20892, USA
| | - Jienyu Ding
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Dr., Bethesda, MD, 20892, USA
| | - Rui Zhang
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Christian Biertümpfel
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Dr., Bethesda, MD, 20892, USA
| | - Kevin F O'Connell
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 8 Center Dr., Bethesda, MD, 20892, USA
| | - Naoko Mizuno
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Dr., Bethesda, MD, 20892, USA
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 50 South Dr., Bethesda, MD, 20892, USA
| |
Collapse
|
4
|
Chinbold B, Kwon HM, Park R. TonEBP inhibits ciliogenesis by controlling aurora kinase A and regulating centriolar satellite integrity. Cell Commun Signal 2024; 22:348. [PMID: 38961488 PMCID: PMC11221002 DOI: 10.1186/s12964-024-01721-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/20/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Primary cilia on the surface of eukaryotic cells serve as sensory antennas for the reception and transmission in various cell signaling pathways. They are dynamic organelles that rapidly form during differentiation and cell cycle exit. Defects in these organelles cause a group of wide-ranging disorders called ciliopathies. Tonicity-responsive enhancer-binding protein (TonEBP) is a pleiotropic stress protein that mediates various physiological and pathological cellular responses. TonEBP is well-known for its role in adaptation to a hypertonic environment, to which primary cilia have been reported to contribute. Furthermore, TonEBP is involved in a wide variety of other signaling pathways, such as Sonic Hedgehog and WNT signaling, that promote primary ciliogenesis, suggesting a possible regulatory role. However, the functional relationship between TonEBP and primary ciliary formation remains unclear. METHODS TonEBP siRNAs and TonEBP-mCherry plasmids were used to examine their effects on cell ciliation rates, assembly and disassembly processes, and regulators. Serum starvation was used as a condition to induce ciliogenesis. RESULTS We identified a novel pericentriolar localization for TonEBP. The results showed that TonEBP depletion facilitates the formation of primary cilia, whereas its overexpression results in fewer ciliated cells. Moreover, TonEBP controlled the expression and activity of aurora kinase A, a major negative regulator of ciliogenesis. Additionally, TonEBP overexpression inhibited the loss of CP110 from the mother centrioles during the early stages of primary cilia assembly. Finally, TonEBP regulated the localization of PCM1 and AZI1, which are necessary for primary cilia formation. CONCLUSIONS This study proposes a novel role for TonEBP as a pericentriolar protein that regulates the integrity of centriolar satellite components. This regulation has shown to have a negative effect on ciliogenesis. Investigations into cilium assembly and disassembly processes suggest that TonEBP acts upstream of the aurora kinase A - histone deacetylase 6 signaling pathway and affects basal body formation to control ciliogenesis. Taken together, our data proposes previously uncharacterized regulation of primary cilia assembly by TonEBP.
Collapse
Affiliation(s)
- Batchingis Chinbold
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Hyug Moo Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
5
|
Renaud CC, Nicolau CA, Maghe C, Trillet K, Jardine J, Escot S, David N, Gavard J, Bidère N. Necrosulfonamide causes oxidation of PCM1 and impairs ciliogenesis and autophagy. iScience 2024; 27:109580. [PMID: 38600973 PMCID: PMC11004361 DOI: 10.1016/j.isci.2024.109580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 01/25/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024] Open
Abstract
Centriolar satellites are high-order assemblies, scaffolded by the protein PCM1, that gravitate as particles around the centrosome and play pivotal roles in fundamental cellular processes notably ciliogenesis and autophagy. Despite stringent control mechanisms involving phosphorylation and ubiquitination, the landscape of post-translational modifications shaping these structures remains elusive. Here, we report that necrosulfonamide (NSA), a small molecule known for binding and inactivating the pivotal effector of cell death by necroptosis MLKL, intersects with centriolar satellites, ciliogenesis, and autophagy independently of MLKL. NSA functions as a potent redox cycler and triggers the oxidation and aggregation of PCM1 alongside select partners, while minimally impacting the overall distribution of centriolar satellites. Additionally, NSA-mediated ROS production disrupts ciliogenesis and leads to the accumulation of autophagy markers, partially alleviated by PCM1 deletion. Together, these results identify PCM1 as a redox sensor protein and provide new insights into the interplay between centriolar satellites and autophagy.
Collapse
Affiliation(s)
- Clotilde C.N. Renaud
- Team SOAP, CRCINA, Nantes University, INSERM, CNRS, Université d’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Carolina Alves Nicolau
- Team SOAP, CRCINA, Nantes University, INSERM, CNRS, Université d’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Clément Maghe
- Team SOAP, CRCINA, Nantes University, INSERM, CNRS, Université d’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Kilian Trillet
- Team SOAP, CRCINA, Nantes University, INSERM, CNRS, Université d’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Jane Jardine
- Team SOAP, CRCINA, Nantes University, INSERM, CNRS, Université d’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Sophie Escot
- Laboratoire d’Optique et de Biosciences LOB, Ecole Polytechnique, Palaiseau, France
| | - Nicolas David
- Laboratoire d’Optique et de Biosciences LOB, Ecole Polytechnique, Palaiseau, France
| | - Julie Gavard
- Team SOAP, CRCINA, Nantes University, INSERM, CNRS, Université d’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Institut de Cancérologie de l’Ouest (ICO), Saint-Herblain, France
| | - Nicolas Bidère
- Team SOAP, CRCINA, Nantes University, INSERM, CNRS, Université d’Angers, Nantes, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| |
Collapse
|
6
|
Vicente JJ, Wagenbach M, Decarreau J, Zelter A, MacCoss MJ, Davis TN, Wordeman L. The kinesin motor Kif9 regulates centriolar satellite positioning and mitotic progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587821. [PMID: 38617353 PMCID: PMC11014612 DOI: 10.1101/2024.04.03.587821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Centrosomes are the principal microtubule-organizing centers of the cell and play an essential role in mitotic spindle function. Centrosome biogenesis is achieved by strict control of protein acquisition and phosphorylation prior to mitosis. Defects in this process promote fragmentation of pericentriolar material culminating in multipolar spindles and chromosome missegregation. Centriolar satellites, membrane-less aggrupations of proteins involved in the trafficking of proteins toward and away from the centrosome, are thought to contribute to centrosome biogenesis. Here we show that the microtubule plus-end directed kinesin motor Kif9 localizes to centriolar satellites and regulates their pericentrosomal localization during interphase. Lack of Kif9 leads to aggregation of satellites closer to the centrosome and increased centrosomal protein degradation that disrupts centrosome maturation and results in chromosome congression and segregation defects during mitosis. Our data reveal roles for Kif9 and centriolar satellites in the regulation of cellular proteostasis and mitosis.
Collapse
|
7
|
Ryu S, Ko D, Shin B, Rhee K. The intercentriolar fibers function as docking sites of centriolar satellites for cilia assembly. J Cell Biol 2024; 223:e202105065. [PMID: 38416111 PMCID: PMC10901237 DOI: 10.1083/jcb.202105065] [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: 05/12/2021] [Revised: 08/09/2023] [Accepted: 01/15/2024] [Indexed: 02/29/2024] Open
Abstract
Two mother centrioles in an animal cell are linked by intercentriolar fibers that have CROCC/rootletin as their main building block. Here, we investigated the regulatory role of intercentriolar/rootlet fibers in cilia assembly. The cilia formation rates were significantly reduced in the CEP250/C-NAP1 and CROCC/rootletin knockout (KO) cells, irrespective of the departure of the young mother centrioles from the basal bodies. In addition, centriolar satellites were dispersed throughout the cytoplasm in the CEP250 and CROCC KO cells. We observed that PCM1 directly binds to CROCC. Their interaction is critical not only for the accumulation of centriolar satellites near the centrosomes/basal bodies but also for cilia formation. Finally, we observed that the centriolar satellite proteins are localized at the intercentriolar/rootlet fibers in the kidney epithelial cells. Based on these findings, we propose that the intercentriolar/rootlet fibers function as docking sites for centriolar satellites near the centrosomes/basal bodies and facilitate the cilia assembly process.
Collapse
Affiliation(s)
- Sungjin Ryu
- Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Donghee Ko
- Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Byungho Shin
- Department of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Kunsoo Rhee
- Department of Biological Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
8
|
Atmakuru PS, Dhawan J. The cilium-centrosome axis in coupling cell cycle exit and cell fate. J Cell Sci 2023; 136:308872. [PMID: 37144419 DOI: 10.1242/jcs.260454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
The centrosome is an evolutionarily conserved, ancient organelle whose role in cell division was first described over a century ago. The structure and function of the centrosome as a microtubule-organizing center, and of its extracellular extension - the primary cilium - as a sensory antenna, have since been extensively studied, but the role of the cilium-centrosome axis in cell fate is still emerging. In this Opinion piece, we view cellular quiescence and tissue homeostasis from the vantage point of the cilium-centrosome axis. We focus on a less explored role in the choice between distinct forms of mitotic arrest - reversible quiescence and terminal differentiation, which play distinct roles in tissue homeostasis. We outline evidence implicating the centrosome-basal body switch in stem cell function, including how the cilium-centrosome complex regulates reversible versus irreversible arrest in adult skeletal muscle progenitors. We then highlight exciting new findings in other quiescent cell types that suggest signal-dependent coupling of nuclear and cytoplasmic events to the centrosome-basal body switch. Finally, we propose a framework for involvement of this axis in mitotically inactive cells and identify future avenues for understanding how the cilium-centrosome axis impacts central decisions in tissue homeostasis.
Collapse
Affiliation(s)
- Priti S Atmakuru
- CSIR Centre for Cellular and Molecular Biology, Hyderabad 500 007, India
| | - Jyotsna Dhawan
- CSIR Centre for Cellular and Molecular Biology, Hyderabad 500 007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
9
|
Hall EA, Kumar D, Prosser SL, Yeyati PL, Herranz-Pérez V, García-Verdugo JM, Rose L, McKie L, Dodd DO, Tennant PA, Megaw R, Murphy LC, Ferreira MF, Grimes G, Williams L, Quidwai T, Pelletier L, Reiter JF, Mill P. Centriolar satellites expedite mother centriole remodeling to promote ciliogenesis. eLife 2023; 12:e79299. [PMID: 36790165 PMCID: PMC9998092 DOI: 10.7554/elife.79299] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 02/14/2023] [Indexed: 02/16/2023] Open
Abstract
Centrosomes are orbited by centriolar satellites, dynamic multiprotein assemblies nucleated by Pericentriolar material 1 (PCM1). To study the requirement for centriolar satellites, we generated mice lacking PCM1, a crucial component of satellites. Pcm1-/- mice display partially penetrant perinatal lethality with survivors exhibiting hydrocephalus, oligospermia, and cerebellar hypoplasia, and variably expressive phenotypes such as hydronephrosis. As many of these phenotypes have been observed in human ciliopathies and satellites are implicated in cilia biology, we investigated whether cilia were affected. PCM1 was dispensable for ciliogenesis in many cell types, whereas Pcm1-/- multiciliated ependymal cells and human PCM1-/- retinal pigmented epithelial 1 (RPE1) cells showed reduced ciliogenesis. PCM1-/- RPE1 cells displayed reduced docking of the mother centriole to the ciliary vesicle and removal of CP110 and CEP97 from the distal mother centriole, indicating compromised early ciliogenesis. Similarly, Pcm1-/- ependymal cells exhibited reduced removal of CP110 from basal bodies in vivo. We propose that PCM1 and centriolar satellites facilitate efficient trafficking of proteins to and from centrioles, including the departure of CP110 and CEP97 to initiate ciliogenesis, and that the threshold to trigger ciliogenesis differs between cell types.
Collapse
Affiliation(s)
- Emma A Hall
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Dhivya Kumar
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of CaliforniaSan FranciscoUnited States
| | - Suzanna L Prosser
- Lunenfeld-Tanenbaum Research Institute, Sinai Health SystemTorontoCanada
| | - Patricia L Yeyati
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Vicente Herranz-Pérez
- Cavanilles Institute of Biodiversity and Evolutionary Biology, University of ValenciaValenciaSpain
- Predepartamental Unit of Medicine, Jaume I UniversityCastelló de la PlanaSpain
| | | | - Lorraine Rose
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Lisa McKie
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Daniel O Dodd
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Peter A Tennant
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Roly Megaw
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Laura C Murphy
- Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Marisa F Ferreira
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Graeme Grimes
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Lucy Williams
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Tooba Quidwai
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Laurence Pelletier
- Lunenfeld-Tanenbaum Research Institute, Sinai Health SystemTorontoCanada
- Department of Molecular Genetics, University of TorontoUniversity of TorontoCanada
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of CaliforniaSan FranciscoUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Pleasantine Mill
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
10
|
Jewett CE, McCurdy BL, O'Toole ET, Stemm-Wolf AJ, Given KS, Lin CH, Olsen V, Martin W, Reinholdt L, Espinosa JM, Sullivan KD, Macklin WB, Prekeris R, Pearson CG. Trisomy 21 induces pericentrosomal crowding delaying primary ciliogenesis and mouse cerebellar development. eLife 2023; 12:e78202. [PMID: 36656118 PMCID: PMC9851619 DOI: 10.7554/elife.78202] [Citation(s) in RCA: 4] [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: 02/26/2022] [Accepted: 12/06/2022] [Indexed: 01/20/2023] Open
Abstract
Trisomy 21, the genetic cause of Down syndrome, disrupts primary cilia formation and function, in part through elevated Pericentrin, a centrosome protein encoded on chromosome 21. Yet how trisomy 21 and elevated Pericentrin disrupt cilia-related molecules and pathways, and the in vivo phenotypic relevance remain unclear. Utilizing ciliogenesis time course experiments combined with light microscopy and electron tomography, we reveal that chromosome 21 polyploidy elevates Pericentrin and microtubules away from the centrosome that corral MyosinVA and EHD1, delaying ciliary membrane delivery and mother centriole uncapping essential for ciliogenesis. If given enough time, trisomy 21 cells eventually ciliate, but these ciliated cells demonstrate persistent trafficking defects that reduce transition zone protein localization and decrease sonic hedgehog signaling in direct anticorrelation with Pericentrin levels. Consistent with cultured trisomy 21 cells, a mouse model of Down syndrome with elevated Pericentrin has fewer primary cilia in cerebellar granule neuron progenitors and thinner external granular layers at P4. Our work reveals that elevated Pericentrin from trisomy 21 disrupts multiple early steps of ciliogenesis and creates persistent trafficking defects in ciliated cells. This pericentrosomal crowding mechanism results in signaling deficiencies consistent with the neurological phenotypes found in individuals with Down syndrome.
Collapse
Affiliation(s)
- Cayla E Jewett
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Bailey L McCurdy
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Eileen T O'Toole
- Molecular, Cellular, and Developmental Biology, University of Colorado BoulderBoulderUnited States
| | - Alexander J Stemm-Wolf
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Katherine S Given
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Carrie H Lin
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Valerie Olsen
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | | | | | - Joaquín M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Pharmacology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Chad G Pearson
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical CampusAuroraUnited States
| |
Collapse
|
11
|
Nguyen A, Goetz SC. TTBK2 controls cilium stability by regulating distinct modules of centrosomal proteins. Mol Biol Cell 2022; 34:ar8. [PMID: 36322399 PMCID: PMC9816645 DOI: 10.1091/mbc.e22-08-0373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The serine-threonine kinase tau tubulin kinase 2 (TTBK2) is a key regulator of the assembly of primary cilia, which are vital signaling organelles. TTBK2 is also implicated in the stability of the assembled cilium through mechanisms that remain to be defined. Here we use mouse embryonic fibroblasts derived from Ttbk2fl/fl, UBC-CreERT+ embryos (hereafter Ttbk2cmut) to dissect the role of TTBK2 in cilium stability. This system depletes TTBK2 levels after cilia formation, allowing us to assess the molecular changes to the assembled cilium over time. As a consequence of Ttbk2 deletion, the ciliary axoneme is destabilized and primary cilia are lost within 48-72 h following recombination. Axoneme destabilization involves an increased frequency of cilia breaks and a reduction in axonemal microtubule modifications. Cilia loss was delayed by using inhibitors that affect actin-based trafficking. At the same time, we find that TTBK2 is required to regulate the composition of the centriolar satellites and to maintain the basal body pools of intraflagellar transport proteins. Altogether, our results reveal parallel pathways by which TTBK2 maintains cilium stability.
Collapse
Affiliation(s)
- Abraham Nguyen
- Molecular Cancer Biology Program, Duke University School of Medicine, Durham, NC 27710,Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710
| | - Sarah C. Goetz
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710,*Address correspondence to: Sarah C. Goetz ()
| |
Collapse
|
12
|
Whole-genome optical mapping to elucidate myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions. Leuk Res 2022; 123:106972. [DOI: 10.1016/j.leukres.2022.106972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
|
13
|
Willekers S, Tessadori F, van der Vaart B, Henning HH, Stucchi R, Altelaar M, Roelen BAJ, Akhmanova A, Bakkers J. The centriolar satellite protein Cfap53 facilitates formation of the zygotic microtubule organizing center in the zebrafish embryo. Development 2022; 149:dev198762. [PMID: 35980365 PMCID: PMC9481976 DOI: 10.1242/dev.198762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 07/20/2022] [Indexed: 12/02/2023]
Abstract
In embryos of most animal species, the zygotic centrosome is assembled by the centriole derived from the sperm cell and pericentriolar proteins present in the oocyte. This zygotic centrosome acts as a microtubule organizing center (MTOC) to assemble the sperm aster and mitotic spindle. As MTOC formation has been studied mainly in adult cells, very little is known about the formation of the zygotic MTOC. Here, we show that zebrafish (Danio rerio) embryos lacking either maternal or paternal Cfap53, a centriolar satellite protein, arrest during the first cell cycle. Although Cfap53 is dispensable for sperm aster function, it aids proper formation of the mitotic spindle. During cell division, Cfap53 colocalizes with γ-tubulin and with other centrosomal and centriolar satellite proteins at the MTOC. Furthermore, we find that γ-tubulin localization at the MTOC is impaired in the absence of Cfap53. Based on these results, we propose a model in which Cfap53 deposited in the oocyte and the sperm participates in the organization of the zygotic MTOC to allow mitotic spindle formation.
Collapse
Affiliation(s)
- Sven Willekers
- Hubrecht Institute-KNAW, Utrecht 3584 CT, The Netherlands
| | | | - Babet van der Vaart
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Heiko H. Henning
- Equine Sciences, Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CM, The Netherlands
| | - Riccardo Stucchi
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Bernard A. J. Roelen
- Embryology, Anatomy and Physiology, Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW, Utrecht 3584 CT, The Netherlands
- Department of Pediatric Cardiology, Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584 EA, The Netherlands
| |
Collapse
|
14
|
McCurdy BL, Jewett CE, Stemm-Wolf AJ, Duc HN, Joshi M, Espinosa JM, Prekeris R, Pearson CG. Trisomy 21 increases microtubules and disrupts centriolar satellite localization. Mol Biol Cell 2022; 33:br11. [PMID: 35476505 PMCID: PMC9635274 DOI: 10.1091/mbc.e21-10-0517-t] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 11/11/2022] Open
Abstract
Trisomy 21, the source of Down syndrome, causes a 0.5-fold protein increase of the chromosome 21-resident gene Pericentrin (PCNT) and reduces primary cilia formation and signaling. We investigate how PCNT imbalances disrupt cilia. Using isogenic RPE-1 cells with increased chromosome 21 dosage, we find PCNT accumulates around the centrosome as a cluster of enlarged cytoplasmic puncta that localize along microtubules (MTs) and at MT ends. Cytoplasmic PCNT puncta impact the density, stability, and localization of the MT trafficking network required for primary cilia. The PCNT puncta appear to sequester cargo peripheral to centrosomes in what we call pericentrosomal crowding. The centriolar satellite proteins PCM1, CEP131, and CEP290, important for ciliogenesis, accumulate at enlarged PCNT puncta in trisomy 21 cells. Reducing PCNT when chromosome 21 ploidy is elevated is sufficient to decrease PCNT puncta and pericentrosomal crowding, reestablish a normal density of MTs around the centrosome, and restore ciliogenesis to wild-type levels. A transient reduction in MTs also decreases pericentrosomal crowding and partially rescues ciliogenesis in trisomy 21 cells, indicating that increased PCNT leads to defects in the MT network deleterious to normal centriolar satellite distribution. We propose that chromosome 21 aneuploidy disrupts MT-dependent intracellular trafficking required for primary cilia.
Collapse
Affiliation(s)
- Bailey L. McCurdy
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045-2537
| | - Cayla E. Jewett
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045-2537
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO 80045-2537
| | - Alexander J. Stemm-Wolf
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045-2537
| | - Huy Nguyen Duc
- Functional Genomics Facility, University of Colorado School of Medicine, Aurora, CO 80045-2537
| | - Molishree Joshi
- Functional Genomics Facility, University of Colorado School of Medicine, Aurora, CO 80045-2537
| | - Joaquin M. Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO 80045-2537
- Functional Genomics Facility, University of Colorado School of Medicine, Aurora, CO 80045-2537
- Department of Pharmacology, University of Colorado School of Medicine, University of Colorado School of Medicine, Aurora, CO 80045-2537
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045-2537
| | - Chad G. Pearson
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045-2537
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, Aurora, CO 80045-2537
| |
Collapse
|
15
|
Komarasamy TV, Adnan NAA, James W, Balasubramaniam VRMT. Zika Virus Neuropathogenesis: The Different Brain Cells, Host Factors and Mechanisms Involved. Front Immunol 2022; 13:773191. [PMID: 35371036 PMCID: PMC8966389 DOI: 10.3389/fimmu.2022.773191] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/21/2022] [Indexed: 12/16/2022] Open
Abstract
Zika virus (ZIKV), despite being discovered six decades earlier, became a major health concern only after an epidemic in French Polynesia and an increase in the number of microcephaly cases in Brazil. Substantial evidence has been found to support the link between ZIKV and neurological complications in infants. The virus targets various cells in the brain, including radial glial cells, neural progenitor cells (NPCs), astrocytes, microglial and glioblastoma stem cells. It affects the brain cells by exploiting different mechanisms, mainly through apoptosis and cell cycle dysregulation. The modulation of host immune response and the inflammatory process has also been demonstrated to play a critical role in ZIKV induced neurological complications. In addition to that, different ZIKV strains have exhibited specific neurotropism and unique molecular mechanisms. This review provides a comprehensive and up-to-date overview of ZIKV-induced neuroimmunopathogenesis by dissecting its main target cells in the brain, and the underlying cellular and molecular mechanisms. We highlighted the roles of the different ZIKV host factors and how they exploit specific host factors through various mechanisms. Overall, it covers key components for understanding the crosstalk between ZIKV and the brain.
Collapse
Affiliation(s)
- Thamil Vaani Komarasamy
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Nur Amelia Azreen Adnan
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - William James
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Vinod R M T Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
16
|
Expanding the phenotype of males with OFD1 pathogenic variants-a case report and literature review. Eur J Med Genet 2022; 65:104496. [PMID: 35398350 PMCID: PMC10369588 DOI: 10.1016/j.ejmg.2022.104496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/31/2022] [Accepted: 03/29/2022] [Indexed: 11/23/2022]
Abstract
Pathogenic variants in the OFD1 gene have been classically associated with the Orofaciodigital syndrome type 1 in females, a condition previously considered to be X-linked dominant with male embryonic lethality. However, an increasing number of males with pathogenic OFD1 variants who survived beyond the neonatal period have now been reported in the literature. Although each new report has added to the ever-broadening spectrum of clinical findings seen in males, many questions about genotype-phenotype correlations and disease mechanism remain. Herein, we describe a 9-year-old male child with a novel hemizygous pathogenic OFD1 variant identified by exome sequencing and a unique combination of findings, not previously reported, including presence of both a hypothalamic hamartoma and the molar tooth sign. His clinical features overlap multiple ciliopathy phenotypes, blurring the boundaries of distinct ciliopathy gene-disease relationships. This case provides further evidence for the consideration of a broad OFD1-relateddisorder spectrum in affected males rather than multiple distinct phenotypes. Additionally, a review of previously published cases of the disorder in males support the inclusion of the OFD1 gene in the differential diagnosis and work up for all individuals who present with primary ciliopathy-type features, regardless of their gender. We also highlight current information about OFD1 variant types and pathogenesis and explore how these could mechanistically drive some of the observed phenotypic differences.
Collapse
|
17
|
Wang L, Paudyal SC, Kang Y, Owa M, Liang FX, Spektor A, Knaut H, Sánchez I, Dynlacht BD. Regulators of tubulin polyglutamylation control nuclear shape and cilium disassembly by balancing microtubule and actin assembly. Cell Res 2022; 32:190-209. [PMID: 34782749 PMCID: PMC8807603 DOI: 10.1038/s41422-021-00584-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 10/05/2021] [Indexed: 02/03/2023] Open
Abstract
Cytoskeletal networks play an important role in regulating nuclear morphology and ciliogenesis. However, the role of microtubule (MT) post-translational modifications in nuclear shape regulation and cilium disassembly has not been explored. Here we identified a novel regulator of the tubulin polyglutamylase complex (TPGC), C11ORF49/CSTPP1, that regulates cytoskeletal organization, nuclear shape, and cilium disassembly. Mechanistically, loss of C11ORF49/CSTPP1 impacts the assembly and stability of the TPGC, which modulates long-chain polyglutamylation levels on microtubules (MTs) and thereby balances the binding of MT-associated proteins and actin nucleators. As a result, loss of TPGC leads to aberrant, enhanced assembly of MTs that penetrate the nucleus, which in turn leads to defects in nuclear shape, and disorganization of cytoplasmic actin that disrupts the YAP/TAZ pathway and cilium disassembly. Further, we showed that C11ORF49/CSTPP1-TPGC plays mechanistically distinct roles in the regulation of nuclear shape and cilium disassembly. Remarkably, disruption of C11ORF49/CSTPP1-TPGC also leads to developmental defects in vivo. Our findings point to an unanticipated nexus that links tubulin polyglutamylation with nuclear shape and ciliogenesis.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY, USA.
| | - Sharad C Paudyal
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yuchen Kang
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY, USA
| | - Mikito Owa
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY, USA
| | - Feng-Xia Liang
- Microscopy Laboratory, Division of Advanced Research Technologies, NYU Langone Health, New York, NY, USA
| | - Alexander Spektor
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Holger Knaut
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - Irma Sánchez
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY, USA
| | - Brian D Dynlacht
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
18
|
Renaud CCN, Bidère N. Function of Centriolar Satellites and Regulation by Post-Translational Modifications. Front Cell Dev Biol 2021; 9:780502. [PMID: 34888313 PMCID: PMC8650133 DOI: 10.3389/fcell.2021.780502] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/09/2021] [Indexed: 11/13/2022] Open
Abstract
Centriolar satellites are small membrane-less granules that gravitate around the centrosome. Recent advances in defining the satellite proteome and interactome have unveiled hundreds of new satellite components thus illustrating the complex nature of these particles. Although initially linked to the homeostasis of centrosome and the formation of primary cilia, these composite and highly dynamic structures appear to participate in additional cellular processes, such as proteostasis, autophagy, and cellular stress. In this review, we first outline the main features and many roles of centriolar satellites. We then discuss how post-translational modifications, such as phosphorylation and ubiquitination, shape their composition and functions. This is of particular interest as interfering with these processes may provide ways to manipulate these structures.
Collapse
Affiliation(s)
| | - Nicolas Bidère
- CNRS, CRCINA, INSERM, Université de Nantes, Nantes, France
| |
Collapse
|
19
|
Li M, Zhang J, Zhou H, Xiang R. Primary Cilia-Related Pathways Moderate the Development and Therapy Resistance of Glioblastoma. Front Oncol 2021; 11:718995. [PMID: 34513696 PMCID: PMC8426355 DOI: 10.3389/fonc.2021.718995] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/04/2021] [Indexed: 12/21/2022] Open
Abstract
As microtubule-based structures, primary cilia are typically present on the cells during the G0 or G1-S/G2 phase of the cell cycle and are closely related to the development of the central nervous system. The presence or absence of this special organelle may regulate the central nervous system tumorigenesis (e.g., glioblastoma) and several degenerative diseases. Additionally, the development of primary cilia can be regulated by several pathways. Conversely, primary cilia are able to regulate a few signaling transduction pathways. Therefore, development of the central nervous system tumors in conjunction with abnormal cilia can be regulated by up- or downregulation of the pathways related to cilia and ciliogenesis. Here, we review some pathways related to ciliogenesis and tumorigenesis, aiming to provide a potential target for developing new therapies at genetic and molecular levels.
Collapse
Affiliation(s)
- Minghao Li
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiaxun Zhang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Haonan Zhou
- School of Life Sciences, Central South University, Changsha, China
| | - Rong Xiang
- School of Life Sciences, Central South University, Changsha, China.,Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, Changsha, China
| |
Collapse
|
20
|
Kumar D, Rains A, Herranz-Pérez V, Lu Q, Shi X, Swaney DL, Stevenson E, Krogan NJ, Huang B, Westlake C, Garcia-Verdugo JM, Yoder BK, Reiter JF. A ciliopathy complex builds distal appendages to initiate ciliogenesis. J Cell Biol 2021; 220:e202011133. [PMID: 34241634 PMCID: PMC8276316 DOI: 10.1083/jcb.202011133] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 05/12/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022] Open
Abstract
Cells inherit two centrioles, the older of which is uniquely capable of generating a cilium. Using proteomics and superresolved imaging, we identify a module that we term DISCO (distal centriole complex). The DISCO components CEP90, MNR, and OFD1 underlie human ciliopathies. This complex localizes to both distal centrioles and centriolar satellites, proteinaceous granules surrounding centrioles. Cells and mice lacking CEP90 or MNR do not generate cilia, fail to assemble distal appendages, and do not transduce Hedgehog signals. Disrupting the satellite pools does not affect distal appendage assembly, indicating that it is the centriolar populations of MNR and CEP90 that are critical for ciliogenesis. CEP90 recruits the most proximal known distal appendage component, CEP83, to root distal appendage formation, an early step in ciliogenesis. In addition, MNR, but not CEP90, restricts centriolar length by recruiting OFD1. We conclude that DISCO acts at the distal centriole to support ciliogenesis by restraining centriole length and assembling distal appendages, defects in which cause human ciliopathies.
Collapse
Affiliation(s)
- Dhivya Kumar
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Addison Rains
- Department of Cell, Developmental, and Integrative Biology, University of Alabama, Birmingham, AL
| | - Vicente Herranz-Pérez
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, Biomedical Research Networking Center on Neurodegenerative Diseases, Valencia, Spain
- Predepartamental Unit of Medicine, Faculty of Health Sciences, Universitat Jaume I, Castelló de la Plana, Spain
| | - Quanlong Lu
- Laboratory of Cellular and Developmental Signaling, Center for Cancer Research, National Cancer Institute Frederick, Frederick, MD
| | - Xiaoyu Shi
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA
| | - Danielle L. Swaney
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
- California Institute for Quantitative Biosciences, Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA
- J. David Gladstone Institutes, San Francisco, CA
| | - Erica Stevenson
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
- California Institute for Quantitative Biosciences, Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA
- J. David Gladstone Institutes, San Francisco, CA
| | - Nevan J. Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
- California Institute for Quantitative Biosciences, Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA
- J. David Gladstone Institutes, San Francisco, CA
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub, San Francisco, CA
| | - Christopher Westlake
- Laboratory of Cellular and Developmental Signaling, Center for Cancer Research, National Cancer Institute Frederick, Frederick, MD
| | - Jose Manuel Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, Biomedical Research Networking Center on Neurodegenerative Diseases, Valencia, Spain
| | - Bradley K. Yoder
- Department of Cell, Developmental, and Integrative Biology, University of Alabama, Birmingham, AL
| | - Jeremy F. Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub, San Francisco, CA
| |
Collapse
|
21
|
Wensel TG, Potter VL, Moye A, Zhang Z, Robichaux MA. Structure and dynamics of photoreceptor sensory cilia. Pflugers Arch 2021; 473:1517-1537. [PMID: 34050409 PMCID: PMC11216635 DOI: 10.1007/s00424-021-02564-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
The rod and cone photoreceptor cells of the vertebrate retina have highly specialized structures that enable them to carry out their function of light detection over a broad range of illumination intensities with optimized spatial and temporal resolution. Most prominent are their unusually large sensory cilia, consisting of outer segments packed with photosensitive disc membranes, a connecting cilium with many features reminiscent of the primary cilium transition zone, and a pair of centrioles forming a basal body which serves as the platform upon which the ciliary axoneme is assembled. These structures form a highway through which an enormous flux of material moves on a daily basis to sustain the continual turnover of outer segment discs and the energetic demands of phototransduction. After decades of study, the details of the fine structure and distribution of molecular components of these structures are still incompletely understood, but recent advances in cellular imaging techniques and animal models of inherited ciliary defects are yielding important new insights. This knowledge informs our understanding both of the mechanisms of trafficking and assembly and of the pathophysiological mechanisms of human blinding ciliopathies.
Collapse
Affiliation(s)
- Theodore G Wensel
- Vera and Marrs McLean Department of Biochemistry and Molecular Biology and Developmental Biology Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Valencia L Potter
- Vera and Marrs McLean Department of Biochemistry and Molecular Biology and Developmental Biology Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
- Medical Scientist Training Program (MSTP), Baylor College of Medicine, Houston, TX, 77030, USA
| | - Abigail Moye
- Vera and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zhixian Zhang
- Vera and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Michael A Robichaux
- Departments of Ophthalmology and Biochemistry, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
22
|
Liu WN, Wu KX, Wang XT, Lin LR, Tong ML, Liu LL. LncRNA- ENST00000421645 promotes T cells to secrete IFN-γ by sponging PCM1 in neurosyphilis. Epigenomics 2021; 13:1187-1203. [PMID: 34382410 DOI: 10.2217/epi-2021-0163] [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: 12/23/2022] Open
Abstract
Aim: Neurosyphilis patients exhibited significant expression of long noncoding RNA (lncRNA) in peripheral blood T lymphocytes. In this study, we further clarified the role of lncRNA-ENST00000421645 in the pathogenic mechanism of neurosyphilis. Methods: lncRNA-ENST00000421645 was transfected into Jurkat-E6-1 cells, namely lentivirus (Lv)-1645 cells. RNA pull-down assay, flow cytometry, RT-qPCR, ELISA (Neobioscience Technology Co Ltd, Shenzhen, China) and RNA immunoprecipitation chip assay were used to analyze the function of lncRNA-ENST00000421645. Results: The expression of IFN-γ in Lv-1645 cells was significantly increased compared to that in Jurkat-E6-1 cells stimulated by phorbol-12-myristate-13-acetate (PMA). Then, it was suggested that lncRNA-ENST00000421645 interacts with PCM1 protein. Silencing PCM1 significantly increased the level of IFN-γ in Lv-1645 cells stimulated by PMA. Conclusion: This study revealed that lncRNA-ENST00000421645 mediates the production of IFN-γ by sponging PCM1 protein after PMA stimulation.
Collapse
Affiliation(s)
- Wen-Na Liu
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medical, Xiamen University, Xiamen, Fujian Province, China.,Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science & Technology of China, Chengdu, China
| | - Kai-Xuan Wu
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medical, Xiamen University, Xiamen, Fujian Province, China
| | - Xiao-Tong Wang
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medical, Xiamen University, Xiamen, Fujian Province, China
| | - Li-Rong Lin
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medical, Xiamen University, Xiamen, Fujian Province, China
| | - Man-Li Tong
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medical, Xiamen University, Xiamen, Fujian Province, China
| | - Li-Li Liu
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medical, Xiamen University, Xiamen, Fujian Province, China
| |
Collapse
|
23
|
Nickson CM, Fabbrizi MR, Carter RJ, Hughes JR, Kacperek A, Hill MA, Parsons JL. USP9X Is Required to Maintain Cell Survival in Response to High-LET Radiation. Front Oncol 2021; 11:671431. [PMID: 34277417 PMCID: PMC8281306 DOI: 10.3389/fonc.2021.671431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/15/2021] [Indexed: 12/26/2022] Open
Abstract
Ionizing radiation (IR) principally acts through induction of DNA damage that promotes cell death, although the biological effects of IR are more broad ranging. In fact, the impact of IR of higher-linear energy transfer (LET) on cell biology is generally not well understood. Critically, therefore, the cellular enzymes and mechanisms responsible for enhancing cell survival following high-LET IR are unclear. To this effect, we have recently performed siRNA screening to identify deubiquitylating enzymes that control cell survival specifically in response to high-LET α-particles and protons, in comparison to low-LET X-rays and protons. From this screening, we have now thoroughly validated that depletion of the ubiquitin-specific protease 9X (USP9X) in HeLa and oropharyngeal squamous cell carcinoma (UMSCC74A) cells using small interfering RNA (siRNA), leads to significantly decreased survival of cells after high-LET radiation. We consequently investigated the mechanism through which this occurs, and demonstrate that an absence of USP9X has no impact on DNA damage repair post-irradiation nor on apoptosis, autophagy, or senescence. We discovered that USP9X is required to stabilize key proteins (CEP55 and CEP131) involved in centrosome and cilia formation and plays an important role in controlling pericentrin-rich foci, particularly in response to high-LET protons. This was also confirmed directly by demonstrating that depletion of CEP55/CEP131 led to both enhanced radiosensitivity of cells to high-LET protons and amplification of pericentrin-rich foci. Our evidence supports the importance of USP9X in maintaining centrosome function and biogenesis and which is crucial particularly in the cellular response to high-LET radiation.
Collapse
Affiliation(s)
- Catherine M. Nickson
- Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Maria Rita Fabbrizi
- Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Rachel J. Carter
- Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Jonathan R. Hughes
- Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Andrzej Kacperek
- Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, United Kingdom
| | - Mark A. Hill
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Gray Laboratories, Oxford, United Kingdom
| | - Jason L. Parsons
- Cancer Research Centre, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, United Kingdom
| |
Collapse
|
24
|
Nita A, Abraham SP, Krejci P, Bosakova M. Oncogenic FGFR Fusions Produce Centrosome and Cilia Defects by Ectopic Signaling. Cells 2021; 10:1445. [PMID: 34207779 PMCID: PMC8227969 DOI: 10.3390/cells10061445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/27/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
A single primary cilium projects from most vertebrate cells to guide cell fate decisions. A growing list of signaling molecules is found to function through cilia and control ciliogenesis, including the fibroblast growth factor receptors (FGFR). Aberrant FGFR activity produces abnormal cilia with deregulated signaling, which contributes to pathogenesis of the FGFR-mediated genetic disorders. FGFR lesions are also found in cancer, raising a possibility of cilia involvement in the neoplastic transformation and tumor progression. Here, we focus on FGFR gene fusions, and discuss the possible mechanisms by which they function as oncogenic drivers. We show that a substantial portion of the FGFR fusion partners are proteins associated with the centrosome cycle, including organization of the mitotic spindle and ciliogenesis. The functions of centrosome proteins are often lost with the gene fusion, leading to haploinsufficiency that induces cilia loss and deregulated cell division. We speculate that this complements the ectopic FGFR activity and drives the FGFR fusion cancers.
Collapse
Affiliation(s)
- Alexandru Nita
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
| | - Sara P. Abraham
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
- Institute of Animal Physiology and Genetics of the CAS, 60200 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| | - Michaela Bosakova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic; (A.N.); (S.P.A.); (P.K.)
- Institute of Animal Physiology and Genetics of the CAS, 60200 Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, 65691 Brno, Czech Republic
| |
Collapse
|
25
|
Odabasi E, Batman U, Firat-Karalar EN. Unraveling the mysteries of centriolar satellites: time to rewrite the textbooks about the centrosome/cilium complex. Mol Biol Cell 2021; 31:866-872. [PMID: 32286929 PMCID: PMC7185976 DOI: 10.1091/mbc.e19-07-0402] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Centriolar satellites are membraneless granules that localize and move around centrosomes and cilia. Once referred to as structures with no obvious function, research in the past decade has identified satellites as key regulators of a wide range of cellular and organismal processes. Importantly, these studies have revealed a substantial overlap between functions, proteomes, and disease links of satellites with centrosomes and cilia. Therefore, satellites are now accepted as the “third component” of the vertebrate centrosome/cilium complex, which profoundly changes the way we think about the assembly, maintenance, and remodeling of the complex at the cellular and organismal levels. In this perspective, we first provide an overview of the cellular and structural complexities of centriolar satellites. We then describe the progress in the identification of the satellite interactome, which have paved the way to a molecular understanding of their mechanism of action and assembly mechanisms. After exploring current insights into their functions as recently described by loss-of-function studies and comparative evolutionary approaches, we discuss major unanswered questions regarding their functional and compositional diversity and their functions outside centrosomes and cilia.
Collapse
Affiliation(s)
- Ezgi Odabasi
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| | - Umut Batman
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| | | |
Collapse
|
26
|
Gurkaslar HK, Culfa E, Arslanhan MD, Lince-Faria M, Firat-Karalar EN. CCDC57 Cooperates with Microtubules and Microcephaly Protein CEP63 and Regulates Centriole Duplication and Mitotic Progression. Cell Rep 2021; 31:107630. [PMID: 32402286 DOI: 10.1016/j.celrep.2020.107630] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 03/08/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022] Open
Abstract
Centrosomes function in key cellular processes ranging from cell division to cellular signaling. Their dysfunction is linked to cancer and developmental disorders. Here, we identify CCDC57 as a pleiotropic regulator of centriole duplication, mitosis, and ciliogenesis. Combining proximity mapping with superresolution imaging, we show that CCDC57 localizes to the proximal end of centrioles and interacts with the microcephaly protein CEP63, centriolar satellite proteins, and microtubules. Loss of CCDC57 causes defects in centriole duplication and results in a failure to localize CEP63 and CEP152 to the centrosome. Additionally, CCDC57 depletion perturbs mitotic progression both in wild-type and centriole-less cells. Importantly, its centrosome-targeting region is required for its interaction with CEP63 and functions during centriole duplication and cilium assembly, whereas the microtubule-targeting region is required for its mitotic functions. Together, our results identify CCDC57 as a critical interface between centrosome and microtubule-mediated cellular processes that are deregulated in microcephaly.
Collapse
Affiliation(s)
- H Kubra Gurkaslar
- Department of Molecular Biology and Genetics, Koç University, Sarıyer, İstanbul 34450, Turkey
| | - Efraim Culfa
- Department of Molecular Biology and Genetics, Koç University, Sarıyer, İstanbul 34450, Turkey
| | - Melis D Arslanhan
- Department of Molecular Biology and Genetics, Koç University, Sarıyer, İstanbul 34450, Turkey
| | - Mariana Lince-Faria
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, Oeiras 2780-156, Portugal
| | - Elif Nur Firat-Karalar
- Department of Molecular Biology and Genetics, Koç University, Sarıyer, İstanbul 34450, Turkey.
| |
Collapse
|
27
|
Piette BL, Alerasool N, Lin ZY, Lacoste J, Lam MHY, Qian WW, Tran S, Larsen B, Campos E, Peng J, Gingras AC, Taipale M. Comprehensive interactome profiling of the human Hsp70 network highlights functional differentiation of J domains. Mol Cell 2021; 81:2549-2565.e8. [PMID: 33957083 DOI: 10.1016/j.molcel.2021.04.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/22/2022]
Abstract
Hsp70s comprise a deeply conserved chaperone family that has a central role in maintaining protein homeostasis. In humans, Hsp70 client specificity is provided by 49 different co-factors known as J domain proteins (JDPs). However, the cellular function and client specificity of JDPs have largely remained elusive. We have combined affinity purification-mass spectrometry (AP-MS) and proximity-dependent biotinylation (BioID) to characterize the interactome of all human JDPs and Hsp70s. The resulting network suggests specific functions for many uncharacterized JDPs, and we establish a role of conserved JDPs DNAJC9 and DNAJC27 in histone chaperoning and ciliogenesis, respectively. Unexpectedly, we find that the J domain of DNAJC27 but not of other JDPs can fully replace the function of endogenous DNAJC27, suggesting a previously unappreciated role for J domains themselves in JDP specificity. More broadly, our work expands the role of the Hsp70-regulated proteostasis network and provides a platform for further discovery of JDP-dependent functions.
Collapse
Affiliation(s)
- Benjamin L Piette
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Nader Alerasool
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Zhen-Yuan Lin
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Jessica Lacoste
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Mandy Hiu Yi Lam
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Wesley Wei Qian
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Stephanie Tran
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Brett Larsen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Eric Campos
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Jian Peng
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada.
| | - Mikko Taipale
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada.
| |
Collapse
|
28
|
Primary cilia and the DNA damage response: linking a cellular antenna and nuclear signals. Biochem Soc Trans 2021; 49:829-841. [PMID: 33843966 DOI: 10.1042/bst20200751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 11/17/2022]
Abstract
The maintenance of genome stability involves integrated biochemical activities that detect DNA damage or incomplete replication, delay the cell cycle, and direct DNA repair activities on the affected chromatin. These processes, collectively termed the DNA damage response (DDR), are crucial for cell survival and to avoid disease, particularly cancer. Recent work has highlighted links between the DDR and the primary cilium, an antenna-like, microtubule-based signalling structure that extends from a centriole docked at the cell surface. Ciliary dysfunction gives rise to a range of complex human developmental disorders termed the ciliopathies. Mutations in ciliopathy genes have been shown to impact on several functions that relate to centrosome integrity, DNA damage signalling, responses to problems in DNA replication and the control of gene expression. This review covers recent findings that link cilia and the DDR and explores the various roles played by key genes in these two contexts. It outlines how proteins encoded by ciliary genes impact checkpoint signalling, DNA replication and repair, gene expression and chromatin remodelling. It discusses how these diverse activities may integrate nuclear responses with those that affect a structure of the cell periphery. Additional directions for exploration of the interplay between these pathways are highlighted, with a focus on new ciliary gene candidates that alter genome stability.
Collapse
|
29
|
Owa M, Dynlacht B. A non-canonical function for Centromere-associated protein-E controls centrosome integrity and orientation of cell division. Commun Biol 2021; 4:358. [PMID: 33742057 PMCID: PMC7979751 DOI: 10.1038/s42003-021-01861-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 02/17/2021] [Indexed: 12/27/2022] Open
Abstract
Centromere-associated protein-E (CENP-E) is a kinesin motor localizing at kinetochores. Although its mitotic functions have been well studied, it has been challenging to investigate direct consequences of CENP-E removal using conventional methods because CENP-E depletion resulted in mitotic arrest. In this study, we harnessed an auxin-inducible degron system to achieve acute degradation of CENP-E. We revealed a kinetochore-independent role for CENP-E that removes pericentriolar material 1 (PCM1) from centrosomes in late S/early G2 phase. After acute loss of CENP-E, centrosomal Polo-like kinase 1 (Plk1) localization is abrogated through accumulation of PCM1, resulting in aberrant phosphorylation and destabilization of centrosomes, which triggers shortened astral microtubules and oblique cell divisions. Furthermore, we also observed centrosome and cell division defects in cells from a microcephaly patient with mutations in CENPE. Orientation of cell division is deregulated in some microcephalic patients, and our unanticipated findings provide additional insights into how microcephaly can result from centrosomal defects.
Collapse
Affiliation(s)
- Mikito Owa
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY, USA.
| | - Brian Dynlacht
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
30
|
Castellano-Pellicena I, Morrison CG, Bell M, O’Connor C, Tobin DJ. Melanin Distribution in Human Skin: Influence of Cytoskeletal, Polarity, and Centrosome-Related Machinery of Stratum basale Keratinocytes. Int J Mol Sci 2021; 22:ijms22063143. [PMID: 33808676 PMCID: PMC8003549 DOI: 10.3390/ijms22063143] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/13/2021] [Accepted: 03/14/2021] [Indexed: 12/12/2022] Open
Abstract
Melanin granules cluster within supra-nuclear caps in basal keratinocytes (KCs) of the human epidermis, where they protect KC genomic DNA against ultraviolet radiation (UVR) damage. While much is known about melanogenesis in melanocytes (MCs) and a moderate amount about melanin transfer from MC to KC, we know little about the fate of melanin once inside KCs. We recently reported that melanin fate in progenitor KCs is regulated by rare asymmetric organelle movement during mitosis. Here, we explore the role of actin, microtubules, and centrosome-associated machinery in distributing melanin within KCs. Short-term cultures of human skin explants were treated with cytochalasin-B and nocodazole to target actin filaments and microtubules, respectively. Treatment effects on melanin distribution were assessed by the Warthin-Starry stain, on centrosome-associated proteins by immunofluorescence microscopy, and on co-localisation with melanin granules by brightfield microscopy. Cytochalasin-B treatment disassembled supra-nuclear melanin caps, while nocodazole treatment moved melanin from the apical to basal KC domain. Centrosome and centriolar satellite-associated proteins showed a high degree of co-localisation with melanin. Thus, once melanin granules are transferred to KCs, their preferred apical distribution appears to be facilitated by coordinated movement of centrosomes and centriolar satellites. This mechanism may control melanin's strategic position within UVR-exposed KCs.
Collapse
Affiliation(s)
- Irene Castellano-Pellicena
- The Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Ciaran G. Morrison
- Centre for Chromosome Biology, School of Natural Sciences, National University of Ireland Galway, H91 W2TY Galway, Ireland;
| | - Mike Bell
- Walgreens Boots Alliance, Nottingham NG90 1BS, UK; (M.B.); (C.O.)
| | - Clare O’Connor
- Walgreens Boots Alliance, Nottingham NG90 1BS, UK; (M.B.); (C.O.)
| | - Desmond J. Tobin
- The Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland;
- The Conway Institute of Biomolecular and Biomedical Research, University College Dublin, D04 V1W8 Dublin, Ireland
- Correspondence: ; Tel.: +353-(0)-1-716-6262
| |
Collapse
|
31
|
Kyun ML, Kim SO, Lee HG, Hwang JA, Hwang J, Soung NK, Cha-Molstad H, Lee S, Kwon YT, Kim BY, Lee KH. Wnt3a Stimulation Promotes Primary Ciliogenesis through β-Catenin Phosphorylation-Induced Reorganization of Centriolar Satellites. Cell Rep 2021; 30:1447-1462.e5. [PMID: 32023461 DOI: 10.1016/j.celrep.2020.01.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/22/2019] [Accepted: 01/06/2020] [Indexed: 01/10/2023] Open
Abstract
Primary cilium is an antenna-like microtubule-based cellular sensing structure. Abnormal regulation of the dynamic assembly and disassembly cycle of primary cilia is closely related to ciliopathy and cancer. The Wnt signaling pathway plays a major role in embryonic development and tissue homeostasis, and defects in Wnt signaling are associated with a variety of human diseases, including cancer. In this study, we provide direct evidence of Wnt3a-induced primary ciliogenesis, which includes a continuous pathway showing that the stimulation of Wnt3a, a canonical Wnt ligand, promotes the generation of β-catenin p-S47 epitope by CK1δ, and these events lead to the reorganization of centriolar satellites resulting in primary ciliogenesis. We have also confirmed the application of our findings in MCF-7/ADR cells, a multidrug-resistant tumor cell model. Thus, our data provide a Wnt3a-induced primary ciliogenesis pathway and may provide a clue on how to overcome multidrug resistance in cancer treatment.
Collapse
Affiliation(s)
- Mi-Lang Kyun
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Korea
| | - Sun-Ok Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Korea
| | - Jeong-Ah Hwang
- Research Institute of Medical Sciences, Department of Physiology, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Joonsung Hwang
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea
| | - Nak-Kyun Soung
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea
| | - Hyunjoo Cha-Molstad
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea
| | - Sangku Lee
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea
| | - Yong Tae Kwon
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea.
| | - Bo Yeon Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Korea.
| | - Kyung Ho Lee
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 30 Yeongudanji-ro, Ochang, Cheongwon, Chungbuk 28116, Republic of Korea.
| |
Collapse
|
32
|
Wu Q, Yu X, Liu L, Sun S, Sun S. Centrosome-phagy: implications for human diseases. Cell Biosci 2021; 11:49. [PMID: 33663596 PMCID: PMC7934278 DOI: 10.1186/s13578-021-00557-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/16/2021] [Indexed: 01/11/2023] Open
Abstract
Autophagy is a prominent mechanism to preserve homeostasis and the response to intracellular or extracellular stress. Autophagic degradation can be selectively targeted to dysfunctional subcellular compartments. Centrosome homeostasis is pivotal for healthy proliferating cells, but centrosome aberration is a hallmark of diverse human disorders. Recently, a process called centrosome-phagy has been identified. The process involves a panel of centrosomal proteins and centrosome-related pathways that mediate the specific degradation of centrosomal components via the autophagic machinery. Although autophagy normally mediates centrosome homeostasis, autophagy defects facilitate ageing and multiple human diseases, such as ciliopathies and cancer, which benefit from centrosome aberration. Here, we discuss the molecular systems that trigger centrosome-phagy and its role in human disorders.
Collapse
Affiliation(s)
- Qi Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, 238 Ziyang Road, Wuhan, 430060, Hubei, People's Republic of China
| | - Xin Yu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, 238 Ziyang Road, Wuhan, 430060, Hubei, People's Republic of China
| | - Le Liu
- Center of Ultramicroscopic Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, 238 Ziyang Road, Wuhan, 430060, Hubei, People's Republic of China.
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, 238 Ziyang Road, Wuhan, 430060, Hubei, People's Republic of China.
| |
Collapse
|
33
|
Kwon OS, Mishra R, Safieddine A, Coleno E, Alasseur Q, Faucourt M, Barbosa I, Bertrand E, Spassky N, Le Hir H. Exon junction complex dependent mRNA localization is linked to centrosome organization during ciliogenesis. Nat Commun 2021; 12:1351. [PMID: 33649372 PMCID: PMC7921557 DOI: 10.1038/s41467-021-21590-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 01/14/2021] [Indexed: 12/17/2022] Open
Abstract
Exon junction complexes (EJCs) mark untranslated spliced mRNAs and are crucial for the mRNA lifecycle. An imbalance in EJC dosage alters mouse neural stem cell (mNSC) division and is linked to human neurodevelopmental disorders. In quiescent mNSC and immortalized human retinal pigment epithelial (RPE1) cells, centrioles form a basal body for ciliogenesis. Here, we report that EJCs accumulate at basal bodies of mNSC or RPE1 cells and decline when these cells differentiate or resume growth. A high-throughput smFISH screen identifies two transcripts accumulating at centrosomes in quiescent cells, NIN and BICD2. In contrast to BICD2, the localization of NIN transcripts is EJC-dependent. NIN mRNA encodes a core component of centrosomes required for microtubule nucleation and anchoring. We find that EJC down-regulation impairs both pericentriolar material organization and ciliogenesis. An EJC-dependent mRNA trafficking towards centrosome and basal bodies might contribute to proper mNSC division and brain development. Exon junction complexes (EJCs) that mark untranslated mRNA are involved in transport, translation and nonsense-mediated mRNA decay. Here the authors show centrosomal localization of EJCs which appears to be required for both the localization of NIN mRNA around centrosomes and ciliogenesis.
Collapse
Affiliation(s)
- Oh Sung Kwon
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Rahul Mishra
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France.,Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Adham Safieddine
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Equipe labélisée Ligue Nationale Contre le Cancer, University of Montpellier, CNRS, Montpellier, France
| | - Emeline Coleno
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Equipe labélisée Ligue Nationale Contre le Cancer, University of Montpellier, CNRS, Montpellier, France
| | - Quentin Alasseur
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Isabelle Barbosa
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Edouard Bertrand
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Equipe labélisée Ligue Nationale Contre le Cancer, University of Montpellier, CNRS, Montpellier, France
| | - Nathalie Spassky
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Hervé Le Hir
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France.
| |
Collapse
|
34
|
Zhao H, Chen Q, Li F, Cui L, Xie L, Huang Q, Liang X, Zhou J, Yan X, Zhu X. Fibrogranular materials function as organizers to ensure the fidelity of multiciliary assembly. Nat Commun 2021; 12:1273. [PMID: 33627667 PMCID: PMC7904937 DOI: 10.1038/s41467-021-21506-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Multicilia are delicate motile machineries, and how they are accurately assembled is poorly understood. Here, we show that fibrogranular materials (FGMs), large arrays of electron-dense granules specific to multiciliated cells, are essential for their ultrastructural fidelity. Pcm1 forms the granular units that further network into widespread FGMs, which are abundant in spherical FGM cores. FGM cores selectively concentrate multiple important centriole-related proteins as clients, including Cep131 that specifically decorates a foot region of ciliary central pair (CP) microtubules. FGMs also tightly contact deuterosome-procentriole complexes. Disruption of FGMs in mouse cells undergoing multiciliogenesis by Pcm1 RNAi markedly deregulates centriolar targeting of FGM clients, elongates CP-foot, and alters deuterosome size, number, and distribution. Although the multicilia are produced in correct numbers, they display abnormal ultrastructure and motility. Our results suggest that FGMs organize deuterosomes and centriole-related proteins to facilitate the faithful assembly of basal bodies and multiciliary axonemes.
Collapse
Affiliation(s)
- Huijie Zhao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qingxia Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Fan Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lihong Cui
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Lele Xie
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qiongping Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xin Liang
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jun Zhou
- Institute of Biomedical Sciences, College of Life Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Normal University, Jinan, 250014, China
| | - Xiumin Yan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xueliang Zhu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China. .,School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
35
|
CFAP53 regulates mammalian cilia-type motility patterns through differential localization and recruitment of axonemal dynein components. PLoS Genet 2020; 16:e1009232. [PMID: 33347437 PMCID: PMC7817014 DOI: 10.1371/journal.pgen.1009232] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 01/20/2021] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Motile cilia can beat with distinct patterns, but how motility variations are regulated remain obscure. Here, we have studied the role of the coiled-coil protein CFAP53 in the motility of different cilia-types in the mouse. While node (9+0) cilia of Cfap53 mutants were immotile, tracheal and ependymal (9+2) cilia retained motility, albeit with an altered beat pattern. In node cilia, CFAP53 mainly localized at the base (centriolar satellites), whereas it was also present along the entire axoneme in tracheal cilia. CFAP53 associated tightly with microtubules and interacted with axonemal dyneins and TTC25, a dynein docking complex component. TTC25 and outer dynein arms (ODAs) were lost from node cilia, but were largely maintained in tracheal cilia of Cfap53-/- mice. Thus, CFAP53 at the base of node cilia facilitates axonemal transport of TTC25 and dyneins, while axonemal CFAP53 in 9+2 cilia stabilizes dynein binding to microtubules. Our study establishes how differential localization and function of CFAP53 contributes to the unique motion patterns of two important mammalian cilia-types. Motile cilia in various kinds of tissues and cell-types drive fluid flow over epithelia or facilitate cellular locomotion. There are two types of motile cilia. Motile cilia with a 9+2 configuration of microtubules are found on tracheal epithelial cells and brain ependymal cells, and exhibit planar beating with effective and recovery strokes. On the other hand, 9+0 motile cilia are found in the embryonic node, show rotational movement and are involved in establishing left-right asymmetry of visceral organs. However, it is not well understood how these two types of motile cilia exhibit their characteristic motion patterns. We have uncovered distinct roles and subcellular localization of the CFAP53 protein in 9+0 versus the 9+2 motile cilia of the mouse. Our data provide novel insights into the molecular basis of motility differences that characterize these two types of mammalian motile cilia.
Collapse
|
36
|
Kumar D, Reiter J. How the centriole builds its cilium: of mothers, daughters, and the acquisition of appendages. Curr Opin Struct Biol 2020; 66:41-48. [PMID: 33160100 DOI: 10.1016/j.sbi.2020.09.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/08/2020] [Accepted: 09/13/2020] [Indexed: 12/23/2022]
Abstract
Centrioles are microtubule-based structures in eukaryotic cells. From organizing the microtubule cytoskeleton during interphase to focusing the mitotic spindle during mitosis, centrioles are busy at all stages of the cell cycle. One crucial interphase function of centrioles is to assemble cilia, microtubular projections that can either be motile or nonmotile. Motile cilia function in sperm locomotion and propulsion of extracellular fluids, as in mucus flow in the lung. Immotile primary cilia are critical for some forms of intercellular signaling. Here, we review how procentrioles mature into daughter and, then, mother centrioles. We highlight key steps in ciliogenesis, including the acquisition of appendages by the mother centriole, as well as the distal centriole, an understudied domain critical for ciliogenesis. Importantly, several genes mutated in ciliopathies encode distal centriolar components. We propose that understanding how centrioles are remodeled to support cilium assembly will provide insights into the molecular etiologies of ciliopathies.
Collapse
Affiliation(s)
- Dhivya Kumar
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Jeremy Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
37
|
Devi R, Pelletier L, Prosser SL. Charting the complex composite nature of centrosomes, primary cilia and centriolar satellites. Curr Opin Struct Biol 2020; 66:32-40. [PMID: 33130249 DOI: 10.1016/j.sbi.2020.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 10/24/2022]
Abstract
The centrosome and its associated structures of the primary cilium and centriolar satellites have been established as central players in a plethora of cellular processes ranging from cell division to cellular signaling. Consequently, defects in the structure or function of these organelles are linked to a diverse range of human diseases, including cancer, microcephaly, ciliopathies, and neurodegeneration. To understand the molecular mechanisms underpinning these diseases, the biology of centrosomes, cilia, and centriolar satellites has to be elucidated. Central to solving this conundrum is the identification, localization, and functional analysis of all the proteins that reside and interact with these organelles. In this review, we discuss the technological breakthroughs that are dissecting the molecular players of these enigmatic organelles with unprecedented spatial and temporal resolution.
Collapse
Affiliation(s)
- Raksha Devi
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, Ontario, M5G 1X5, Canada
| | - Laurence Pelletier
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, Ontario, M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| | - Suzanna L Prosser
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, Ontario, M5G 1X5, Canada.
| |
Collapse
|
38
|
Vishnoi N, Dhanasekeran K, Chalfant M, Surovstev I, Khokha MK, Lusk CP. Differential turnover of Nup188 controls its levels at centrosomes and role in centriole duplication. J Cell Biol 2020; 219:133835. [PMID: 32211895 PMCID: PMC7055002 DOI: 10.1083/jcb.201906031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 11/18/2019] [Accepted: 01/09/2020] [Indexed: 02/07/2023] Open
Abstract
NUP188 encodes a scaffold component of the nuclear pore complex (NPC) and has been implicated as a congenital heart disease gene through an ill-defined function at centrioles. Here, we explore the mechanisms that physically and functionally segregate Nup188 between the pericentriolar material (PCM) and NPCs. Pulse-chase fluorescent labeling indicates that Nup188 populates centrosomes with newly synthesized protein that does not exchange with NPCs even after mitotic NPC breakdown. In addition, the steady-state levels of Nup188 are controlled by the sensitivity of the PCM pool, but not the NPC pool, to proteasomal degradation. Proximity-labeling and super-resolution microscopy show that Nup188 is vicinal to the inner core of the interphase centrosome. Consistent with this, we demonstrate direct binding between Nup188 and Cep152. We further show that Nup188 functions in centriole duplication at or upstream of Sas6 loading. Together, our data establish Nup188 as a component of PCM needed to duplicate the centriole with implications for congenital heart disease mechanisms.
Collapse
Affiliation(s)
- Nidhi Vishnoi
- Department of Cell Biology, Yale School of Medicine, New Haven, CT
| | | | | | - Ivan Surovstev
- Department of Cell Biology, Yale School of Medicine, New Haven, CT
| | - Mustafa K Khokha
- Pediatric Genomics Discovery Program, Departments of Pediatrics and Genetics, Yale School of Medicine, New Haven, CT
| | - C Patrick Lusk
- Department of Cell Biology, Yale School of Medicine, New Haven, CT
| |
Collapse
|
39
|
Douanne T, André-Grégoire G, Thys A, Trillet K, Gavard J, Bidère N. CYLD Regulates Centriolar Satellites Proteostasis by Counteracting the E3 Ligase MIB1. Cell Rep 2020; 27:1657-1665.e4. [PMID: 31067453 DOI: 10.1016/j.celrep.2019.04.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 03/18/2019] [Accepted: 04/05/2019] [Indexed: 12/25/2022] Open
Abstract
The tumor suppressor CYLD is a deubiquitinating enzyme that removes non-degradative ubiquitin linkages bound to a variety of signal transduction adaptors. CYLD participates in the formation of primary cilia, a microtubule-based structure that protrudes from the cell body to act as a "sensing antenna." Yet, how exactly CYLD regulates ciliogenesis is not fully understood. Here, we conducted an unbiased proteomic screen of CYLD binding partners and identified components of the centriolar satellites. These small granular structures, tethered to the scaffold protein pericentriolar matrix protein 1 (PCM1), gravitate toward the centrosome and orchestrate ciliogenesis. CYLD knockdown promotes PCM1 degradation and the subsequent dismantling of the centriolar satellites. We found that CYLD marshals the centriolar satellites by deubiquitinating and preventing the E3 ligase Mindbomb 1 (MIB1) from marking PCM1 for proteasomal degradation. These results link CYLD to the regulation of centriolar satellites proteostasis and provide insight into how reversible ubiquitination finely tunes ciliogenesis.
Collapse
Affiliation(s)
- Tiphaine Douanne
- CRCINA, Team SOAP, INSERM, CNRS, Université de Nantes, Université d'Angers, IRS-UN blg, Room 405, 8 quai Moncousu, 44007 Nantes, France
| | - Gwennan André-Grégoire
- CRCINA, Team SOAP, INSERM, CNRS, Université de Nantes, Université d'Angers, IRS-UN blg, Room 405, 8 quai Moncousu, 44007 Nantes, France; Institut de Cancérologie de l'Ouest, Site René Gauducheau, Saint-Herblain, France
| | - An Thys
- CRCINA, Team SOAP, INSERM, CNRS, Université de Nantes, Université d'Angers, IRS-UN blg, Room 405, 8 quai Moncousu, 44007 Nantes, France
| | - Kilian Trillet
- CRCINA, Team SOAP, INSERM, CNRS, Université de Nantes, Université d'Angers, IRS-UN blg, Room 405, 8 quai Moncousu, 44007 Nantes, France
| | - Julie Gavard
- CRCINA, Team SOAP, INSERM, CNRS, Université de Nantes, Université d'Angers, IRS-UN blg, Room 405, 8 quai Moncousu, 44007 Nantes, France; Institut de Cancérologie de l'Ouest, Site René Gauducheau, Saint-Herblain, France
| | - Nicolas Bidère
- CRCINA, Team SOAP, INSERM, CNRS, Université de Nantes, Université d'Angers, IRS-UN blg, Room 405, 8 quai Moncousu, 44007 Nantes, France.
| |
Collapse
|
40
|
Danielsson F, Mahdessian D, Axelsson U, Sullivan D, Uhlén M, Andersen JS, Thul PJ, Lundberg E. Spatial Characterization of the Human Centrosome Proteome Opens Up New Horizons for a Small but Versatile Organelle. Proteomics 2020; 20:e1900361. [PMID: 32558245 DOI: 10.1002/pmic.201900361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/29/2020] [Indexed: 12/27/2022]
Abstract
After a century of research, the human centrosome continues to fascinate. Based on immunofluorescence and confocal microscopy, an extensive inventory of the protein components of the human centrosome, and the centriolar satellites, with the important contribution of over 300 novel proteins localizing to these compartments is presented. A network of candidate centrosome proteins involved in ubiquitination, including six interaction partners of the Kelch-like protein 21, and an additional network of protein phosphatases, together supporting the suggested role of the centrosome as an interactive hub for cell signaling, is identified. Analysis of multi-localization across cellular organelles analyzed within the Human Protein Atlas (HPA) project shows how multi-localizing proteins are particularly overrepresented in centriolar satellites, supporting the dynamic nature and wide range of functions for this compartment. In summary, the spatial dissection of the human centrosome and centriolar satellites described here provides a comprehensive knowledgebase for further exploration of their proteomes.
Collapse
Affiliation(s)
- Frida Danielsson
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Solna, 17121, Sweden
| | - Diana Mahdessian
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Solna, 17121, Sweden
| | - Ulrika Axelsson
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Solna, 17121, Sweden
| | - Devin Sullivan
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Solna, 17121, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Solna, 17121, Sweden.,Department of Protein Science, KTH - Royal Institute of Technology, Stockholm, 106 91, Sweden
| | - Jens S Andersen
- Center for Experimental Bioinformatics, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, 5230, Denmark
| | - Peter J Thul
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Solna, 17121, Sweden
| | - Emma Lundberg
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, Solna, 17121, Sweden
| |
Collapse
|
41
|
Acute inhibition of centriolar satellite function and positioning reveals their functions at the primary cilium. PLoS Biol 2020; 18:e3000679. [PMID: 32555591 PMCID: PMC7326281 DOI: 10.1371/journal.pbio.3000679] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/30/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
Centriolar satellites are dynamic, membraneless granules composed of over 200 proteins. They store, modify, and traffic centrosome and primary cilium proteins, and help to regulate both the biogenesis and some functions of centrosomes and cilium. In most cell types, satellites cluster around the perinuclear centrosome, but their integrity and cellular distribution are dynamically remodeled in response to different stimuli, such as cell cycle cues. Dissecting the specific and temporal functions and mechanisms of satellites and how these are influenced by their cellular positioning and dynamics has been challenging using genetic approaches, particularly in ciliated and proliferating cells. To address this, we developed a chemical-based trafficking assay to rapidly and efficiently redistribute satellites to either the cell periphery or center, and fuse them into stable clusters in a temporally controlled way. Induced satellite clustering at either the periphery or center resulted in antagonistic changes in the pericentrosomal levels of a subset of proteins, revealing a direct and selective role for their positioning in protein targeting and sequestration. Systematic analysis of the interactome of peripheral satellite clusters revealed enrichment of proteins implicated in cilium biogenesis and mitosis. Importantly, induction of peripheral satellite targeting in ciliated cells revealed a function for satellites not just for efficient cilium assembly but also in the maintenance of steady-state cilia and in cilia disassembly by regulating the structural integrity of the ciliary axoneme. Finally, perturbing satellite distribution and dynamics inhibited their mitotic dissolution, and mitotic progression was perturbed only in cells with centrosomal satellite clustering. Collectively, our results for the first time showed a direct link between satellite functions and their pericentrosomal clustering, suggested new mechanisms underlying satellite functions during cilium assembly, and provided a new tool for probing temporal satellite functions in different contexts What happens when centriolar satellites are not in the right place at the right time? By redistributing satellites to the periphery or center of the cell and assessing the consequences of their mispositioning, this study reveals novel functions for satellites during mitosis, cilium maintenance, and cilium disassembly and suggests new mechanisms.
Collapse
|
42
|
Zhang C, Li C, Siu GKY, Luo X, Yu S. Distinct Roles of TRAPPC8 and TRAPPC12 in Ciliogenesis via Their Interactions With OFD1. Front Cell Dev Biol 2020; 8:148. [PMID: 32258032 PMCID: PMC7090148 DOI: 10.3389/fcell.2020.00148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/21/2020] [Indexed: 01/08/2023] Open
Abstract
The transport protein particle (TRAPP) complex was initially identified as a tethering factor for COPII vesicle. Subsequently, three forms (TRAPPI, II, and III) have been found and TRAPPIII has been reported to serve as a regulator in autophagy. This study investigates a new role of mammalian TRAPPIII in ciliogenesis. We found a ciliopathy protein, oral-facial-digital syndrome 1 (OFD1), interacting with the TRAPPIII-specific subunits TRAPPC8 and TRAPPC12. TRAPPC8 is necessary for the association of OFD1 with pericentriolar material 1 (PCM1). Its depletion reduces the extent of colocalized signals between OFD1 and PCM1, but does not compromise the structural integrity of centriolar satellites. The interaction between TRAPPC8 and OFD1 inhibits that between OFD1 and TRAPPC12, suggesting different roles of TRAPPIII-specific subunits in ciliogenesis and explaining the differences in cilium lengths in TRAPPC8-depleted and TRAPPC12-depleted hTERT-RPE1 cells. On the other hand, TRAPPC12 depletion causes increased ciliary length because TRAPPC12 is required for the disassembly of primary cilia. Overall, this study has revealed different roles of TRAPPC8 and TRAPPC12 in the assembly of centriolar satellites and demonstrated a possible tethering role of TRAPPIII during ciliogenesis.
Collapse
Affiliation(s)
- Caiyun Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, China
| | - Chunman Li
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen, China
| | - Gavin Ka Yu Siu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, China
| | - Xiaomin Luo
- School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, China
| | - Sidney Yu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Sha Tin, China
| |
Collapse
|
43
|
Cianfanelli V, Cecconi F. Doryphagy: when selective autophagy safeguards centrosome integrity. Mol Cell Oncol 2020; 7:1719021. [PMID: 32158930 DOI: 10.1080/23723556.2020.1719021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 10/25/2022]
Abstract
Although centrosome abnormalities are frequent in cancer, the mechanisms responsible for their accumulation are poorly understood. Here we comment on our recent publication identifying a new type of selective autophagy, named doryphagy, which preserves centrosome organization through targeting Centriolar Satellites (CS). Thus, doryphagy prevents inaccurate mitosis and genomic instability.
Collapse
Affiliation(s)
- Valentina Cianfanelli
- Unit of Cell Stress and Survival, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Francesco Cecconi
- Unit of Cell Stress and Survival, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark.,Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.,Department of Biology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
44
|
Prosser SL, Pelletier L. Centriolar satellite biogenesis and function in vertebrate cells. J Cell Sci 2020; 133:133/1/jcs239566. [PMID: 31896603 DOI: 10.1242/jcs.239566] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Centriolar satellites are non-membranous cytoplasmic granules that concentrate in the vicinity of the centrosome, the major microtubule-organizing centre (MTOC) in animal cells. Originally assigned as conduits for the transport of proteins towards the centrosome and primary cilium, the complexity of satellites is starting to become apparent. Recent studies defined the satellite proteome and interactomes, placing hundreds of proteins from diverse pathways in association with satellites. In addition, studies on cells lacking satellites have revealed that the centrosome can assemble in their absence, whereas studies on acentriolar cells have demonstrated that satellite assembly is independent from an intact MTOC. A role for satellites in ciliogenesis is well established; however, their contribution to other cellular functions is poorly understood. In this Review, we discuss the developments in our understanding of centriolar satellite assembly and function, and why satellites are rapidly becoming established as governors of multiple cellular processes. We highlight the composition and biogenesis of satellites and what is known about the regulation of these aspects. Furthermore, we discuss the evolution from thinking of satellites as mere facilitators of protein trafficking to the centrosome to thinking of them being key regulators of protein localization and cellular proteostasis for a diverse set of pathways, making them of broader interest to fields beyond those focused on centrosomes and ciliogenesis.
Collapse
Affiliation(s)
- Suzanna L Prosser
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada
| | - Laurence Pelletier
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada .,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
45
|
Wen F, Armstrong N, Hou W, Cruz-Cosme R, Obwolo LA, Ishizuka K, Ullah H, Luo MH, Sawa A, Tang Q. Zika virus increases mind bomb 1 levels, causing degradation of pericentriolar material 1 (PCM1) and dispersion of PCM1-containing granules from the centrosome. J Biol Chem 2019; 294:18742-18755. [PMID: 31666336 DOI: 10.1074/jbc.ra119.010973] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/28/2019] [Indexed: 12/25/2022] Open
Abstract
The centrosome is a cytoplasmic nonenveloped organelle functioning as one of the microtubule-organizing centers and composing a centriole center surrounded by pericentriolar material (PCM) granules. PCM consists of many centrosomal proteins, including PCM1 and centrosomal protein 131 (CEP131), and helps maintain centrosome stability. Zika virus (ZIKV) is a flavivirus of the family Flaviviridae whose RNA and viral particles are replicated in the cytoplasm. However, how ZIKV interacts with host cell components during its productive infection stage is incompletely understood. Here, using several primate cell lines, we report that ZIKV infection disrupts and disperses the PCM granules. We demonstrate that PCM1- and CEP131-containing granules are dispersed in ZIKV-infected cells, whereas the centrioles remain intact. We found that ZIKV does not significantly alter cellular skeletal proteins, and, hence, these proteins may not be involved in the interaction between ZIKV and centrosomal proteins. Moreover, ZIKV infection decreased PCM1 and CEP131 protein, but not mRNA, levels. We further found that the protease inhibitor MG132 prevents the decrease in PCM1 and CEP131 levels and centriolar satellite dispersion. Therefore, we hypothesized that ZIKV infection induces proteasomal PCM1 and CEP131 degradation and thereby disrupts the PCM granules. Supporting this hypothesis, we show that ZIKV infection increases levels of mind bomb 1 (MIB1), previously demonstrated to be an E3 ubiquitin ligase for PCM1 and CEP131 and that ZIKV fails to degrade or disperse PCM in MIB1-ko cells. Our results imply that ZIKV infection activates MIB1-mediated ubiquitination that degrades PCM1 and CEP131, leading to PCM granule dispersion.
Collapse
Affiliation(s)
- Fayuan Wen
- Department of Microbiology, Howard University College of Medicine, Washington, D. C. 20059
| | - Najealicka Armstrong
- Department of Microbiology, Howard University College of Medicine, Washington, D. C. 20059
| | - Wangheng Hou
- Department of Microbiology, Howard University College of Medicine, Washington, D. C. 20059
| | - Ruth Cruz-Cosme
- Department of Microbiology, Howard University College of Medicine, Washington, D. C. 20059
| | - Lilian Akello Obwolo
- Department of Microbiology, Howard University College of Medicine, Washington, D. C. 20059
| | - Koko Ishizuka
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Hemayet Ullah
- Department of Biology, Howard University, Washington, D. C. 20059
| | - Min-Hua Luo
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, D. C. 20059.
| |
Collapse
|
46
|
Wang P, Xia J, Zhang L, Zhao S, Li S, Wang H, Cheng S, Li H, Yin W, Pei D, Shu X. SNX17 Recruits USP9X to Antagonize MIB1-Mediated Ubiquitination and Degradation of PCM1 during Serum-Starvation-Induced Ciliogenesis. Cells 2019; 8:cells8111335. [PMID: 31671755 PMCID: PMC6912348 DOI: 10.3390/cells8111335] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/15/2019] [Accepted: 10/27/2019] [Indexed: 12/12/2022] Open
Abstract
Centriolar satellites are non-membrane cytoplasmic granules that deliver proteins to centrosome during centrosome biogenesis and ciliogenesis. Centriolar satellites are highly dynamic during cell cycle or ciliogenesis and how they are regulated remains largely unknown. We report here that sorting nexin 17 (SNX17) regulates the homeostasis of a subset of centriolar satellite proteins including PCM1, CEP131, and OFD1 during serum-starvation-induced ciliogenesis. Mechanistically, SNX17 recruits the deubiquitinating enzyme USP9X to antagonize the mindbomb 1 (MIB1)-induced ubiquitination and degradation of PCM1. SNX17 deficiency leads to enhanced degradation of USP9X as well as PCM1 and disrupts ciliogenesis upon serum starvation. On the other hand, SNX17 is dispensable for the homeostasis of PCM1 and USP9X in serum-containing media. These findings reveal a SNX17/USP9X mediated pathway essential for the homeostasis of centriolar satellites under serum starvation, and provide insight into the mechanism of USP9X in ciliogenesis, which may lead to a better understating of USP9X-deficiency-related human diseases such as X-linked mental retardation and neurodegenerative diseases.
Collapse
Affiliation(s)
- Pengtao Wang
- School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou 510530, China.
- Hefei Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
| | - Jianhong Xia
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou 510530, China.
| | - Leilei Zhang
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou 510530, China.
| | - Shaoyang Zhao
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou 510530, China.
| | - Shengbiao Li
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou 510530, China.
| | - Haiyun Wang
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou 510530, China.
| | - Shan Cheng
- Hefei Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
| | - Heying Li
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou 510530, China.
| | - Wenguang Yin
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou 510530, China.
| | - Duanqing Pei
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou 510530, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510005, China.
| | - Xiaodong Shu
- CAS Key Laboratory of Regenerative Biology, South China Institutes for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou 510530, China.
- Hefei Institute of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510005, China.
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
47
|
Centrosomal and ciliary targeting of CCDC66 requires cooperative action of centriolar satellites, microtubules and molecular motors. Sci Rep 2019; 9:14250. [PMID: 31582766 PMCID: PMC6776500 DOI: 10.1038/s41598-019-50530-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
Mammalian centrosomes and cilia play key roles in many cellular processes and their deregulation is linked to cancer and ciliopathies. Spatiotemporal regulation of their biogenesis and function in response to physiological stimuli requires timely protein targeting. This can occur by different pathways, including microtubule-dependent active transport and via centriolar satellites, which are key regulators of cilia assembly and signaling. How satellites mediate their functions and their relationship with other targeting pathways is currently unclear. To address this, we studied retinal degeneration gene product CCDC66, which localizes to centrosomes, cilia, satellites and microtubules and functions in ciliogenesis. FRAP experiments showed that its centrosomal pool was dynamic and the ciliary pool associated with the ciliary axoneme and was stable. Centrosomal CCDC66 abundance and dynamics required microtubule-dependent active transport and tethering, and was inhibited by sequestration at satellites. Systematic quantitation of satellite dynamics identified only a small fraction to display microtubule-based bimodal motility, consistent with trafficking function. Majority displayed diffusive motility with unimodal persistence, supporting sequestration function. Together, our findings reveal new mechanisms of communication between membrane-less compartments.
Collapse
|
48
|
Fokin Artem I, Zhapparova Olga N, Burakov Anton V, Nadezhdina Elena S. Centrosome-derived microtubule radial array, PCM-1 protein, and primary cilia formation. PROTOPLASMA 2019; 256:1361-1373. [PMID: 31079229 DOI: 10.1007/s00709-019-01385-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 04/16/2019] [Indexed: 06/09/2023]
Abstract
In animal cells, the centrosome nucleates and anchors microtubules (MT), forming their radial array. During interphase centrosome-derived MT, aster can either team up with other MT network or function in an autonomous manner. What is the function of the centrosome-derived MT aster? We suggested that it might play an important role in the formation of the primary cilium, the organelle obligatorily associated with the centrosome. PCM-1 (PeriCentriolar Matrix 1) protein, which participates in the organization of the primary cilium, is a part of pericentiolar satellites. They are transported to the centrosome along MTs by the motor protein dynein in a complex with its cofactor dynactin. Previously, we showed that SLK/LOSK phosphorylated the p150Glued subunit of dynactin, thus promoting its centrosomal targeting followed by its participation in the retention of microtubules. Here, we found that under the repression of SLK/LOSK activity, the PCM-1 protein lost its pericentrosomal localization and was being dispersed throughout the cytoplasm. Despite that the alanine and glutamine mutants of p150Glued had opposite effects on PCM-1 localization, they associated with PCM-1 to the same extent. The occurrence of primary cilia also significantly decreased when SLK/LOSK was repressed. These defects also correlated with a disturbance of the long-range transport in cells, whereas dynein-depending motility was intact. Treatment with the GSK-3β kinase inhibitor also resulted in the loss of the centrosome-derived MT aster, dispersion of PCM-1 over the cytoplasm, and reduction of primary cilia occurrence. Thus, kinases involved in the centrosome-derived MT aster regulation can indirectly control the formation of primary cilia in cells.
Collapse
Affiliation(s)
- I Fokin Artem
- A.N. Belozersky Institute for Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Vorobjevy Gory, 1 bld.73, Moscow, Russian Federation, 119991
| | - N Zhapparova Olga
- A.N. Belozersky Institute for Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Vorobjevy Gory, 1 bld.73, Moscow, Russian Federation, 119991
| | - V Burakov Anton
- A.N. Belozersky Institute for Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Vorobjevy Gory, 1 bld.73, Moscow, Russian Federation, 119991
| | - S Nadezhdina Elena
- Department of Cell Biology of Institute of Protein Research, Russian Academy of Science, Vavilova ul., 34, Moscow, Russian Federation, 117334.
| |
Collapse
|
49
|
Miller M, Wang Y, Bromberg Y. What went wrong with variant effect predictor performance for the PCM1 challenge. Hum Mutat 2019; 40:1486-1494. [PMID: 31268618 PMCID: PMC6744297 DOI: 10.1002/humu.23832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/03/2019] [Accepted: 05/31/2019] [Indexed: 12/31/2022]
Abstract
The recent years have seen a drastic increase in the amount of available genomic sequences. Alongside this explosion, hundreds of computational tools were developed to assess the impact of observed genetic variation. Critical Assessment of Genome Interpretation (CAGI) provides a platform to evaluate the performance of these tools in experimentally relevant contexts. In the CAGI-5 challenge assessing the 38 missense variants affecting the human Pericentriolar material 1 protein (PCM1), our SNAP-based submission was the top performer, although it did worse than expected from other evaluations. Here, we compare the CAGI-5 submissions, and 24 additional commonly used variant effect predictors, to analyze the reasons for this observation. We identified per residue conservation, structural, and functional PCM1 characteristics, which may be responsible. As expected, predictors had a hard time distinguishing effect variants in nonconserved positions. They were also better able to call effect variants in a structurally rich region than in a less-structured one; in the latter, they more often correctly identified benign than effect variants. Curiously, most of the protein was predicted to be functionally robust to mutation-a feature that likely makes it a harder problem for generalized variant effect predictors.
Collapse
Affiliation(s)
- Maximilian Miller
- Department of Biochemistry and Microbiology, Rutgers University, 76 Lipman Dr, New Brunswick, NJ 08873, USA
| | - Yanran Wang
- Department of Biochemistry and Microbiology, Rutgers University, 76 Lipman Dr, New Brunswick, NJ 08873, USA
| | - Yana Bromberg
- Department of Biochemistry and Microbiology, Rutgers University, 76 Lipman Dr, New Brunswick, NJ 08873, USA
| |
Collapse
|
50
|
Atoh1 Controls Primary Cilia Formation to Allow for SHH-Triggered Granule Neuron Progenitor Proliferation. Dev Cell 2019; 48:184-199.e5. [PMID: 30695697 DOI: 10.1016/j.devcel.2018.12.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 10/11/2018] [Accepted: 12/19/2018] [Indexed: 11/23/2022]
Abstract
During cerebellar development, granule neuron progenitors (GNPs) proliferate by transducing Sonic Hedgehog (SHH) signaling via the primary cilium. Precise regulation of ciliogenesis, thus, ensures proper GNP pool expansion. Here, we report that Atoh1, a transcription factor required for GNPs formation, controls the presence of primary cilia, maintaining GNPs responsiveness to SHH. Loss of primary cilia abolishes the ability of Atoh1 to keep GNPs in a proliferative state. Mechanistically, Atoh1 promotes ciliogenesis by transcriptionally regulating Cep131, which facilitates centriolar satellite (CS) clustering to the basal body. Importantly, ectopic expression of Cep131 counteracts the effects of Atoh1 loss in GNPs by restoring proper localization of CS and ciliogenesis. This Atoh1-CS-primary cilium-SHH pro-proliferative pathway is also conserved in SHH-type medulloblastoma, a pediatric brain tumor arising from the GNPs. Together, our data reveal how Atoh1 modulates the primary cilium to regulate GNPs development.
Collapse
|