1
|
Jaroenlak P, McCarty KL, Xia B, Lam C, Zwack EE, Almasri NL, Sudar J, Aubry M, Yanai I, Bhabha G, Ekiert DC. scRNA-seq uncovers the transcriptional dynamics of Encephalitozoon intestinalis parasites in human macrophages. Nat Commun 2025; 16:3269. [PMID: 40188181 PMCID: PMC11972355 DOI: 10.1038/s41467-025-57837-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 03/05/2025] [Indexed: 04/07/2025] Open
Abstract
Microsporidia are single-celled intracellular parasites that cause opportunistic diseases in humans. Encephalitozoon intestinalis is a prevalent human-infecting species that invades the small intestine. Macrophages are potential reservoirs of infection, and dissemination to other organ systems is also observed. The macrophage response to infection and the developmental trajectory of the parasite are not well studied. Here we use single cell RNA sequencing to investigate transcriptional changes in both the parasite and the host during E. intestinalis infection of human macrophages in vitro. The parasite undergoes large transcriptional changes throughout the life cycle, providing a blueprint for parasite development. While a small population of infected macrophages mount a response, most remain transcriptionally unchanged, suggesting that the majority of parasites may avoid host detection. The stealthy microsporidian lifestyle likely allows these parasites to harness macrophages for replication. Together, our data provide insights into the host response in primary human macrophages and the E. intestinalis developmental program.
Collapse
Affiliation(s)
- Pattana Jaroenlak
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Cell Biology, New York University Grossman School of Medicine, New York, 10016, USA
| | - Kacie L McCarty
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Vilcek Institute of Graduate Biomedical Sciences, NYU School of Medicine, New York, NY, 10016, USA
| | - Bo Xia
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Cherry Lam
- Department of Cell Biology, New York University Grossman School of Medicine, New York, 10016, USA
| | - Erin E Zwack
- Department of Microbiology, New York University Grossman School of Medicine, New York, 10016, USA
| | - Nadia L Almasri
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Joseph Sudar
- Vilcek Institute of Graduate Biomedical Sciences, NYU School of Medicine, New York, NY, 10016, USA
| | - Maelle Aubry
- Department of Cell Biology, New York University Grossman School of Medicine, New York, 10016, USA
| | - Itai Yanai
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Gira Bhabha
- Department of Cell Biology, New York University Grossman School of Medicine, New York, 10016, USA.
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA.
| | - Damian C Ekiert
- Department of Cell Biology, New York University Grossman School of Medicine, New York, 10016, USA.
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Microbiology, New York University Grossman School of Medicine, New York, 10016, USA.
| |
Collapse
|
2
|
Xie L, Kang F, Qin T, Kang Y, Liang T, Xie H, Froese CD, Xie H, Au A, Yip CM, Trimble WS, Gaisano HY. Septin5 deletion enhances β-cell exocytosis by releasing microtubule-tethered insulin granules onto plasma membrane. Nat Commun 2025; 16:2725. [PMID: 40108136 PMCID: PMC11923188 DOI: 10.1038/s41467-025-57421-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Septin5 interacts with SNARE proteins to regulate exocytosis in neurons, but its role in pancreatic β-cells is unknown. Here, we report that Septin5 is abundant in rodent and human β-cells, deletion of which dramatically enhances biphasic glucose-stimulated insulin secretion, including in type 2 diabetes (T2D). Super-resolution imaging shows that Septin5 is preferentially assembled in microtubule-plasma membrane contact sites in a microtubule-dependent manner, which provides discrete harbor for secretory granule anchoring. By decreasing the stability of the cortical microtubule meshwork, Septin5 depletion increases insulin granule dynamics and access to the plasma membrane. Analysis of spatiotemporal coupling of fusion events and localized Ca2+ influx through L-type Ca2+ channels show that Septin5 depletion increases releasable granule pool clustering on Ca2+ channels, previously shown to be impaired in T2D, thus rectifying this T2D defect. Hence, inhibition of Septin5 can improve insulin secretion.
Collapse
Affiliation(s)
- Li Xie
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
| | - Fei Kang
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
| | - Tairan Qin
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Youhou Kang
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Tao Liang
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Huanli Xie
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Carol D Froese
- The program in Cell Biology, Department of Biochemistry, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Hong Xie
- The program in Cell Biology, Department of Biochemistry, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Aaron Au
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Christopher M Yip
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - William S Trimble
- The program in Cell Biology, Department of Biochemistry, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Herbert Y Gaisano
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
3
|
Lacey SE, Pigino G. The intraflagellar transport cycle. Nat Rev Mol Cell Biol 2025; 26:175-192. [PMID: 39537792 DOI: 10.1038/s41580-024-00797-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
Primary and motile cilia are eukaryotic organelles that perform crucial roles in cellular signalling and motility. Intraflagellar transport (IFT) contributes to the formation of the highly specialized ciliary proteome by active and selective transport of soluble and membrane proteins into and out of cilia. IFT is performed by the IFT-A and IFT-B protein complexes, which together link cargoes to the microtubule motors kinesin and dynein. In this Review, we discuss recent structural and mechanistic insights on how the IFT complexes are first recruited to the base of the cilium, how they polymerize into an anterograde IFT train, and how this complex imports cargoes from the cytoplasm. We will describe insights into how kinesin-driven anterograde trains are carried to the ciliary tip, where they are remodelled into dynein-driven retrograde trains for cargo export. We will also present how the interplay between IFT-A and IFT-B complexes, motor proteins and cargo adaptors is regulated for bidirectional ciliary transport.
Collapse
|
4
|
Li X, Qin Y, Kong Y, Karunarathna SC, Liang Y, Xu J. Optimization of Protoplast Preparation Conditions in Lyophyllum decastes and Transcriptomic Analysis Throughout the Process. J Fungi (Basel) 2024; 10:886. [PMID: 39728382 DOI: 10.3390/jof10120886] [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: 11/25/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
Protoplasts are essential tools for genetic manipulation and functional genomics research in fungi. This study systematically optimized protoplast preparation conditions and examined transcriptional changes throughout the preparation and regeneration processes to elucidate the molecular mechanisms underlying the formation and regeneration of protoplasts in Lyophyllum decastes. The results indicated an optimal protoplast yield of 5.475 × 106 cells/mL under conditions of fungal age at 10 days, digestion time of 2.25 h, enzyme concentration of 2%, and digestion temperature of 28 °C. The Z5 medium supplemented with L. decastes mycelial extract achieved a high regeneration rate of 2.86. RNA-seq analysis revealed 2432 differentially expressed genes (DEGs) during protoplast formation and 5825 DEGs during regeneration. Casein kinase I, cytochrome P450 (CYP52), and redox-regulated input receptor (PEX5) were significantly upregulated during the protoplast stage, while β-1,3-glucan synthase (SKN1), chitin synthase (CHS2), hydrophobin-1, and hydrophobin-2 showed significant upregulation during the protoplast regeneration phase. These findings provide a reference for the efficient preparation and regeneration of protoplasts and offer new insights into the molecular mechanisms of protoplast formation and cell wall regeneration in fungi.
Collapse
Affiliation(s)
- Xiaobin Li
- College of Agriculture, Yanbian University, Yanji 133002, China
- College of Agriculture, Jilin Agricultural Science and Technology University, Jilin 132000, China
| | - Ying Qin
- College of Agriculture, Jilin Agricultural Science and Technology University, Jilin 132000, China
- College of Forestry, Beihua University, Jilin 132000, China
| | - Yufei Kong
- College of Agriculture, Yanbian University, Yanji 133002, China
- College of Agriculture, Jilin Agricultural Science and Technology University, Jilin 132000, China
| | | | - Yunjiang Liang
- College of Agriculture, Yanbian University, Yanji 133002, China
| | - Jize Xu
- College of Agriculture, Yanbian University, Yanji 133002, China
- College of Agriculture, Jilin Agricultural Science and Technology University, Jilin 132000, China
| |
Collapse
|
5
|
Schampera JN, Schwan C. Septin dynamics and organization in mammalian cells. Curr Opin Cell Biol 2024; 91:102442. [PMID: 39509956 DOI: 10.1016/j.ceb.2024.102442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024]
Abstract
Septins are involved in many important cellular processes, and septin dysfunction has been implicated in various pathologies, such as cancer. Like other components of the cytoskeleton -F-actin, microtubules, and intermediate filaments-septins can self-assemble into filaments and higher-order structures. These non-polar filaments are assembled from complex and variable multimeric building blocks. Septins exhibit a distinct preference for interacting with actin and microtubule structures, particularly at the interface with cellular membrane. Although they are crucial for many vital cellular functions and are frequently observed at prominent cellular structures like stress fibers, cilia, and neuronal processes, our understanding of the regulation of septin filament dynamics and the organized assembly of higher-order structures remains limited. However, recent insights into the architecture of septin filaments, the structure of crucial septin domains, and their interactions with other cellular components (F-actin, microtubules, membranes) and regulatory proteins may now pave the way for rapid progress.
Collapse
Affiliation(s)
- Janik N Schampera
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Carsten Schwan
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
6
|
Cavini IA, Fontes MG, Zeraik AE, Lopes JLS, Araujo APU. Novel lipid-interaction motifs within the C-terminal domain of Septin10 from Schistosoma mansoni. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184371. [PMID: 39025256 DOI: 10.1016/j.bbamem.2024.184371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/05/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Septins are cytoskeletal proteins and their interaction with membranes is crucial for their role in various cellular processes. Septins have polybasic regions (PB1 and PB2) which are important for lipid interaction. Earlier, we and others have highlighted the role of the septin C-terminal domain (CTD) to membrane interaction. However, detailed information on residues/group of residues important for such feature is lacking. In this study, we investigate the lipid-binding profile of Schistosoma mansoni Septin10 (SmSEPT10) using PIP strip and Langmuir monolayer adsorption assays. Our findings highlight the CTD as the primary domain responsible for lipid interaction in SmSEPT10, showing binding to phosphatidylinositol phosphates. SmSEPT10 CTD contains a conserved polybasic region (PB3) present in both animals and fungi septins, and a Lys (K367) within its putative amphipathic helix (AH) that we demonstrate as important for lipid binding. PB3 deletion or mutation of this Lys (K367A) strongly impairs lipid interaction. Remarkably, we observe that the AH within a construct lacking the final 43 amino acid residues is insufficient for lipid binding. Furthermore, we investigate the homocomplex formed by SmSEPT10 CTD in solution by cross-linking experiments, CD spectroscopy, SEC-MALS and SEC-SAXS. Taken together, our studies define the lipid-binding region in SmSEPT10 and offer insights into the molecular basis of septin-membrane binding. This information is particularly relevant for less-studied non-human septins, such as SmSEPT10.
Collapse
Affiliation(s)
- Italo A Cavini
- São Carlos Institute of Physics, University of São Paulo, São Carlos, SP 13560-970, Brazil; School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-903, Brazil
| | - Marina G Fontes
- São Carlos Institute of Physics, University of São Paulo, São Carlos, SP 13560-970, Brazil; Department of Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Ana Eliza Zeraik
- Laboratory of Chemistry and Function of Proteins and Peptides, Center for Biosciences and Biotechnology, North Fluminense State University Darcy Ribeiro, Campos dos Goytacazes, RJ 28013-602, Brazil
| | - Jose L S Lopes
- Laboratory of Molecular Biophysics, Department of Physics, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-901, Brazil
| | - Ana Paula U Araujo
- São Carlos Institute of Physics, University of São Paulo, São Carlos, SP 13560-970, Brazil.
| |
Collapse
|
7
|
Ewerling A, May-Simera HL. Evolutionary trajectory for nuclear functions of ciliary transport complex proteins. Microbiol Mol Biol Rev 2024; 88:e0000624. [PMID: 38995044 PMCID: PMC11426024 DOI: 10.1128/mmbr.00006-24] [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] [Indexed: 07/13/2024] Open
Abstract
SUMMARYCilia and the nucleus were two defining features of the last eukaryotic common ancestor. In early eukaryotic evolution, these structures evolved through the diversification of a common membrane-coating ancestor, the protocoatomer. While in cilia, the descendants of this protein complex evolved into parts of the intraflagellar transport complexes and BBSome, the nucleus gained its selectivity by recruiting protocoatomer-like proteins to the nuclear envelope to form the selective nuclear pore complexes. Recent studies show a growing number of proteins shared between the proteomes of the respective organelles, and it is currently unknown how ciliary transport proteins could acquire nuclear functions and vice versa. The nuclear functions of ciliary proteins are still observable today and remain relevant for the understanding of the disease mechanisms behind ciliopathies. In this work, we review the evolutionary history of cilia and nucleus and their respective defining proteins and integrate current knowledge into theories for early eukaryotic evolution. We postulate a scenario where both compartments co-evolved and that fits current models of eukaryotic evolution, explaining how ciliary proteins and nucleoporins acquired their dual functions.
Collapse
Affiliation(s)
- Alexander Ewerling
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| | - Helen Louise May-Simera
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
8
|
Varela Salgado M, Piatti S. Septin Organization and Dynamics for Budding Yeast Cytokinesis. J Fungi (Basel) 2024; 10:642. [PMID: 39330402 PMCID: PMC11433133 DOI: 10.3390/jof10090642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/28/2024] Open
Abstract
Cytokinesis, the process by which the cytoplasm divides to generate two daughter cells after mitosis, is a crucial stage of the cell cycle. Successful cytokinesis must be coordinated with chromosome segregation and requires the fine orchestration of several processes, such as constriction of the actomyosin ring, membrane reorganization, and, in fungi, cell wall deposition. In Saccharomyces cerevisiae, commonly known as budding yeast, septins play a pivotal role in the control of cytokinesis by assisting the assembly of the cytokinetic machinery at the division site and controlling its activity. Yeast septins form a collar at the division site that undergoes major dynamic transitions during the cell cycle. This review discusses the functions of septins in yeast cytokinesis, their regulation and the implications of their dynamic remodelling for cell division.
Collapse
Affiliation(s)
- Maritzaida Varela Salgado
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293 Montpellier, France
| | - Simonetta Piatti
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293 Montpellier, France
| |
Collapse
|
9
|
Mendonça DC, Morais STB, Ciol H, Pinto APA, Leonardo DA, Pereira HD, Valadares NF, Portugal RV, Klaholz BP, Garratt RC, Araujo APU. Structural Insights into Ciona intestinalis Septins: Complexes Suggest a Mechanism for Nucleotide-dependent Interfacial Cross-talk. J Mol Biol 2024; 436:168693. [PMID: 38960133 DOI: 10.1016/j.jmb.2024.168693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
Septins are filamentous nucleotide-binding proteins which can associate with membranes in a curvature-dependent manner leading to structural remodelling and barrier formation. Ciona intestinalis, a model for exploring the development and evolution of the chordate lineage, has only four septin-coding genes within its genome. These represent orthologues of the four classical mammalian subgroups, making it a minimalist non-redundant model for studying the modular assembly of septins into linear oligomers and thereby filamentous polymers. Here, we show that C. intestinalis septins present a similar biochemistry to their human orthologues and also provide the cryo-EM structures of an octamer, a hexamer and a tetrameric sub-complex. The octamer, which has the canonical arrangement (2-6-7-9-9-7-6-2) clearly shows an exposed NC-interface at its termini enabling copolymerization with hexamers into mixed filaments. Indeed, only combinations of septins which had CiSEPT2 occupying the terminal position were able to assemble into filaments via NC-interface association. The CiSEPT7-CiSEPT9 tetramer is the smallest septin particle to be solved by Cryo-EM to date and its good resolution (2.7 Å) provides a well-defined view of the central NC-interface. On the other hand, the CiSEPT7-CiSEPT9 G-interface shows signs of fragility permitting toggling between hexamers and octamers, similar to that seen in human septins but not in yeast. The new structures provide insights concerning the molecular mechanism for cross-talk between adjacent interfaces. This indicates that C. intestinalis may represent a valuable tool for future studies, fulfilling the requirements of a complete but simpler system to understand the mechanisms behind the assembly and dynamics of septin filaments.
Collapse
Affiliation(s)
| | | | - Heloísa Ciol
- São Carlos Institute of Physics, USP, São Carlos, SP, Brazil
| | | | | | | | | | - Rodrigo V Portugal
- Brazilian Nanotechnology National Laboratory, CNPEM, Campinas, SP, Brazil; Biotechnosciency Program, Federal University of ABC, Santo André, SP, Brazil
| | - Bruno P Klaholz
- Centre for Integrative Biology (CBI), Department of Integrated Structural Biology, IGBMC (Institute of Genetics and of Molecular and Cellular Biology), 67404 Illkirch, France; Centre National de la Recherche Scientifique (CNRS) UMR 7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U964, 67404 Illkirch, France; Université de Strasbourg, 67081 Strasbourg, France
| | | | - Ana P U Araujo
- São Carlos Institute of Physics, USP, São Carlos, SP, Brazil.
| |
Collapse
|
10
|
Macarelli V, Harding EC, Gershlick DC, Merkle FT. A Short Sequence Targets Transmembrane Proteins to Primary Cilia. Cells 2024; 13:1156. [PMID: 38995007 PMCID: PMC11240719 DOI: 10.3390/cells13131156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
Primary cilia are finger-like sensory organelles that extend from the bodies of most cell types and have a distinct lipid and protein composition from the plasma membrane. This partitioning is maintained by a diffusion barrier that restricts the entry of non-ciliary proteins, and allows the selective entry of proteins harboring a ciliary targeting sequence (CTS). However, CTSs are not stereotyped and previously reported sequences are insufficient to drive efficient ciliary localisation across diverse cell types. Here, we describe a short peptide sequence that efficiently targets transmembrane proteins to primary cilia in all tested cell types, including human neurons. We generate human-induced pluripotent stem cell (hiPSC) lines stably expressing a transmembrane construct bearing an extracellular HaloTag and intracellular fluorescent protein, which enables the bright, specific labeling of primary cilia in neurons and other cell types to facilitate studies of cilia in health and disease. We demonstrate the utility of this resource by developing an image analysis pipeline for the automated measurement of primary cilia to detect changes in their length associated with altered signaling or disease state.
Collapse
Affiliation(s)
- Viviana Macarelli
- Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK; (V.M.); (E.C.H.)
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Edward C. Harding
- Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK; (V.M.); (E.C.H.)
| | - David C. Gershlick
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK;
| | - Florian T. Merkle
- Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK; (V.M.); (E.C.H.)
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| |
Collapse
|
11
|
Reddy Palicharla V, Mukhopadhyay S. Molecular and structural perspectives on protein trafficking to the primary cilium membrane. Biochem Soc Trans 2024; 52:1473-1487. [PMID: 38864436 PMCID: PMC11346432 DOI: 10.1042/bst20231403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/13/2024]
Abstract
The primary cilium is a dynamic subcellular compartment templated from the mother centriole or basal body. Cilia are solitary and tiny, but remarkably consequential in cellular pathways regulating proliferation, differentiation, and maintenance. Multiple transmembrane proteins such as G-protein-coupled receptors, channels, enzymes, and membrane-associated lipidated proteins are enriched in the ciliary membrane. The precise regulation of ciliary membrane content is essential for effective signal transduction and maintenance of tissue homeostasis. Surprisingly, a few conserved molecular factors, intraflagellar transport complex A and the tubby family adapter protein TULP3, mediate the transport of most membrane cargoes into cilia. Recent advances in cryogenic electron microscopy provide fundamental insights into these molecular players. Here, we review the molecular players mediating cargo delivery into the ciliary membrane through the lens of structural biology. These mechanistic insights into ciliary transport provide a framework for understanding of disease variants in ciliopathies, enable precise manipulation of cilia-mediated pathways, and provide a platform for the development of targeted therapeutics.
Collapse
Affiliation(s)
- Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, U.S.A
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, U.S.A
| |
Collapse
|
12
|
Houston BJ, Merriner DJ, Stathatos GG, Nguyen JH, O'Connor AE, Lopes AM, Conrad DF, Baker M, Dunleavy JE, O'Bryan MK. Genetic mutation of Cep76 results in male infertility due to abnormal sperm tail composition. Life Sci Alliance 2024; 7:e202302452. [PMID: 38570187 PMCID: PMC10992998 DOI: 10.26508/lsa.202302452] [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: 10/22/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
The transition zone is a specialised gate at the base of cilia/flagella, which separates the ciliary compartment from the cytoplasm and strictly regulates protein entry. We identified a potential new regulator of the male germ cell transition zone, CEP76. We demonstrated that CEP76 was involved in the selective entry and incorporation of key proteins required for sperm function and fertility into the ciliary compartment and ultimately the sperm tail. In the mutant, sperm tails were shorter and immotile as a consequence of deficits in essential sperm motility proteins including DNAH2 and AKAP4, which accumulated at the sperm neck in the mutant. Severe annulus, fibrous sheath, and outer dense fibre abnormalities were also detected in sperm lacking CEP76. Finally, we identified that CEP76 dictates annulus positioning and structure. This study suggests CEP76 as a male germ cell transition zone protein and adds further evidence to the hypothesis that the spermatid transition zone and annulus are part of the same functional structure.
Collapse
Affiliation(s)
- Brendan J Houston
- School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - D Jo Merriner
- School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - G Gemma Stathatos
- School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Joseph H Nguyen
- School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Anne E O'Connor
- School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Alexandra M Lopes
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology & Immunology, University of Porto, Porto, Portugal
| | - Donald F Conrad
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Mark Baker
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, Australia
| | - Jessica Em Dunleavy
- School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| | - Moira K O'Bryan
- School of BioSciences and Bio21 Molecular Sciences and Biotechnology Institute, The University of Melbourne, Parkville, Australia
| |
Collapse
|
13
|
Mercey O, Mukherjee S, Guichard P, Hamel V. The molecular architecture of the ciliary transition zones. Curr Opin Cell Biol 2024; 88:102361. [PMID: 38648677 DOI: 10.1016/j.ceb.2024.102361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/25/2024]
Abstract
Cilia and flagella are specialized eukaryotic organelles projecting from the surface of eukaryotic cells that play a central role in various physiological processes, including cell motility, sensory perception, and signal transduction. At the base of these structures lies the ciliary transition zone, a pivotal region that functions as a gatekeeper and communication hub for ciliary activities. Despite its crucial role, the intricacies of its architecture remain poorly understood, especially given the variations in its organization across different cell types and species. In this review, we explore the molecular architecture of the ciliary transition zone, with a particular focus on recent findings obtained using cryotomography and super-resolution imaging techniques.
Collapse
Affiliation(s)
- Olivier Mercey
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Souradip Mukherjee
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Paul Guichard
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland.
| | - Virginie Hamel
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
14
|
Zhang Y, Lin C. Lipid osmosis, membrane tension, and other mechanochemical driving forces of lipid flow. Curr Opin Cell Biol 2024; 88:102377. [PMID: 38823338 PMCID: PMC11193448 DOI: 10.1016/j.ceb.2024.102377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/03/2024]
Abstract
Nonvesicular lipid transport among different membranes or membrane domains plays crucial roles in lipid homeostasis and organelle biogenesis. However, the forces that drive such lipid transport are not well understood. We propose that lipids tend to flow towards the membrane area with a higher membrane protein density in a process termed lipid osmosis. This process lowers the membrane tension in the area, resulting in a membrane tension difference called osmotic membrane tension. We examine the thermodynamic basis and experimental evidence of lipid osmosis and osmotic membrane tension. We predict that lipid osmosis can drive bulk lipid flows between different membrane regions through lipid transfer proteins, scramblases, or similar barriers that selectively pass lipids but not membrane proteins. We also speculate on the biological functions of lipid osmosis. Finally, we explore other driving forces for lipid transfer and describe potential methods and systems to further test our theory.
Collapse
Affiliation(s)
- Yongli Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA.
| | - Chenxiang Lin
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Nanobiology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
15
|
Jaroenlak P, McCarty KL, Xia B, Lam C, Zwack EE, Yanai I, Bhabha G, Ekiert DC. scRNA-seq reveals transcriptional dynamics of Encephalitozoon intestinalis parasites in human macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596468. [PMID: 38853846 PMCID: PMC11160751 DOI: 10.1101/2024.05.30.596468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Microsporidia are single-celled intracellular parasites that cause opportunistic diseases in humans. Encephalitozoon intestinalis is a prevalent human-infecting species that invades the small intestine. Dissemination to other organ systems is also observed, and is potentially facilitated by macrophages. The macrophage response to infection and the developmental trajectory of the parasite are not well studied. Here we use single cell RNA sequencing to investigate transcriptional changes in both the host and parasite during infection. While a small population of infected macrophages mount a response, most remain transcriptionally unchanged, suggesting that the majority of parasites may avoid host detection. The parasite transcriptome reveals large transcriptional changes throughout the life cycle, providing a blueprint for parasite development. The stealthy microsporidian lifestyle likely allows these parasites to harness macrophages for replication and dissemination. Together, our data provide insights into the host response in primary human macrophages and the E. intestinalis developmental program.
Collapse
Affiliation(s)
- Pattana Jaroenlak
- Department of Cell Biology, New York University Grossman School of Medicine, New York 10016, USA
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kacie L. McCarty
- Department of Cell Biology, New York University Grossman School of Medicine, New York 10016, USA
- Department of Microbiology, New York University Grossman School of Medicine, New York 10016, USA
| | - Bo Xia
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Cherry Lam
- Department of Cell Biology, New York University Grossman School of Medicine, New York 10016, USA
| | - Erin E. Zwack
- Department of Microbiology, New York University Grossman School of Medicine, New York 10016, USA
| | - Itai Yanai
- Institute for Computational Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Gira Bhabha
- Department of Cell Biology, New York University Grossman School of Medicine, New York 10016, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Damian C. Ekiert
- Department of Cell Biology, New York University Grossman School of Medicine, New York 10016, USA
- Department of Microbiology, New York University Grossman School of Medicine, New York 10016, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
16
|
Zhang Y, Lin C. Lipid osmosis, membrane tension, and other mechanochemical driving forces of lipid flow. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574656. [PMID: 38260424 PMCID: PMC10802412 DOI: 10.1101/2024.01.08.574656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Nonvesicular lipid transport among different membranes or membrane domains plays crucial roles in lipid homeostasis and organelle biogenesis. However, the forces that drive such lipid transport are not well understood. We propose that lipids tend to flow towards the membrane area with a higher membrane protein density in a process termed lipid osmosis. This process lowers the membrane tension in the area, resulting in a membrane tension difference called osmotic membrane tension. We examine the thermodynamic basis and experimental evidence of lipid osmosis and osmotic membrane tension. We predict that lipid osmosis can drive bulk lipid flows between different membrane regions through lipid transfer proteins, scramblases, or other similar barriers that selectively pass lipids but not membrane proteins. We also speculate on the biological functions of lipid osmosis. Finally, we explore other driving forces for lipid transfer and describe potential methods and systems to further test our theory.
Collapse
Affiliation(s)
- Yongli Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Chenxiang Lin
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Nanobiology Institute, Yale University, West Haven, CT 06516, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
17
|
Varela Salgado M, Adriaans IE, Touati SA, Ibanes S, Lai-Kee-Him J, Ancelin A, Cipelletti L, Picas L, Piatti S. Phosphorylation of the F-BAR protein Hof1 drives septin ring splitting in budding yeast. Nat Commun 2024; 15:3383. [PMID: 38649354 PMCID: PMC11035697 DOI: 10.1038/s41467-024-47709-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
A double septin ring accompanies cytokinesis in yeasts and mammalian cells. In budding yeast, reorganisation of the septin collar at the bud neck into a dynamic double ring is essential for actomyosin ring constriction and cytokinesis. Septin reorganisation requires the Mitotic Exit Network (MEN), a kinase cascade essential for cytokinesis. However, the effectors of MEN in this process are unknown. Here we identify the F-BAR protein Hof1 as a critical target of MEN in septin remodelling. Phospho-mimicking HOF1 mutant alleles overcome the inability of MEN mutants to undergo septin reorganisation by decreasing Hof1 binding to septins and facilitating its translocation to the actomyosin ring. Hof1-mediated septin rearrangement requires its F-BAR domain, suggesting that it may involve a local membrane remodelling that leads to septin reorganisation. In vitro Hof1 can induce the formation of intertwined septin bundles, while a phosphomimetic Hof1 protein has impaired septin-bundling activity. Altogether, our data indicate that Hof1 modulates septin architecture in distinct ways depending on its phosphorylation status.
Collapse
Affiliation(s)
- Maritzaida Varela Salgado
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France
| | - Ingrid E Adriaans
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France
| | - Sandra A Touati
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013, Paris, France
| | - Sandy Ibanes
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France
| | - Joséphine Lai-Kee-Him
- CBS (Centre de Biologie Structurale), University of Montpellier, CNRS UMR 5048, INSERM U 1054, 34090, Montpellier, France
| | - Aurélie Ancelin
- CBS (Centre de Biologie Structurale), University of Montpellier, CNRS UMR 5048, INSERM U 1054, 34090, Montpellier, France
| | - Luca Cipelletti
- L2C (Laboratoire Charles Coulomb), University of Montpellier, CNRS 34095, Montpellier, France
- IUF (Institut Universitaire de France, 75231, Paris, France
| | - Laura Picas
- IRIM (Institut de Recherche en Infectiologie de Montpellier), University of Montpellier, CNRS UMR 9004, 34293, Montpellier, France
| | - Simonetta Piatti
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), University of Montpellier, CNRS UMR 5237, 34293, Montpellier, France.
| |
Collapse
|
18
|
Pazour GJ. Cilia Structure and Function in Human Disease. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2024; 34:100509. [PMID: 38836197 PMCID: PMC11147146 DOI: 10.1016/j.coemr.2024.100509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Ciliary dysfunction causes a large group of developmental and degenerative human diseases known as ciliopathies. These diseases reflect the critical roles that cilia play in sensing the environment and in force generation for motility. Sensory functions include our senses of vision and olfaction. In addition, primary and motile cilia throughout our body monitor the environment allowing cells to coordinate their biology with the cells around them. This coordination is critical to organ development and maintenance, and ciliary dysfunction causes diverse structural birth defects and degenerative diseases. Defects in motility cause lung disease due to the failure of mucociliary clearance, male infertility due to the failure of sperm motility and the ability of sperm to move through the efferent ducts, and disturbances of the left-right axis due to a failure of nodal cilia to establish proper left-right cues.
Collapse
Affiliation(s)
- Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Biotech II, Worcester, Massachusetts, USA
| |
Collapse
|
19
|
Moran AL, Louzao-Martinez L, Norris DP, Peters DJM, Blacque OE. Transport and barrier mechanisms that regulate ciliary compartmentalization and ciliopathies. Nat Rev Nephrol 2024; 20:83-100. [PMID: 37872350 DOI: 10.1038/s41581-023-00773-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2023] [Indexed: 10/25/2023]
Abstract
Primary cilia act as cell surface antennae, coordinating cellular responses to sensory inputs and signalling molecules that regulate developmental and homeostatic pathways. Cilia are therefore critical to physiological processes, and defects in ciliary components are associated with a large group of inherited pleiotropic disorders - known collectively as ciliopathies - that have a broad spectrum of phenotypes and affect many or most tissues, including the kidney. A central feature of the cilium is its compartmentalized structure, which imparts its unique molecular composition and signalling environment despite its membrane and cytosol being contiguous with those of the cell. Such compartmentalization is achieved via active transport pathways that bring protein cargoes to and from the cilium, as well as gating pathways at the ciliary base that establish diffusion barriers to protein exchange into and out of the organelle. Many ciliopathy-linked proteins, including those involved in kidney development and homeostasis, are components of the compartmentalizing machinery. New insights into the major compartmentalizing pathways at the cilium, namely, ciliary gating, intraflagellar transport, lipidated protein flagellar transport and ciliary extracellular vesicle release pathways, have improved our understanding of the mechanisms that underpin ciliary disease and associated renal disorders.
Collapse
Affiliation(s)
- Ailis L Moran
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Laura Louzao-Martinez
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| | - Oliver E Blacque
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
20
|
Chatzifrangkeskou M, Kouis P, Skourides PA. JNK regulates ciliogenesis through the interflagellar transport complex and actin networks. J Cell Biol 2023; 222:e202303052. [PMID: 37851005 PMCID: PMC10585068 DOI: 10.1083/jcb.202303052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/16/2023] [Accepted: 08/29/2023] [Indexed: 10/19/2023] Open
Abstract
The c-Jun N-terminal kinase (JNK) regulates various important physiological processes. Although the JNK pathway has been under intense investigation for over 20 yr, its complexity is still perplexing, with multiple protein partners underlying the diversity of its activity. We show that JNK is associated with the basal bodies in both primary and motile cilia. Loss of JNK disrupts basal body migration and docking and leads to severe ciliogenesis defects. JNK's involvement in ciliogenesis stems from a dual role in the regulation of the actin networks of multiciliated cells (MCCs) and the establishment of the intraflagellar transport-B core complex. JNK signaling is also critical for the maintenance of the actin networks and ciliary function in mature MCCs. JNK is implicated in the development of diabetes, neurodegeneration, and liver disease, all of which have been linked to ciliary dysfunction. Our work uncovers a novel role of JNK in ciliogenesis and ciliary function that could have important implications for JNK's role in the disease.
Collapse
Affiliation(s)
| | - Panayiotis Kouis
- Respiratory Physiology Laboratory, Medical School, University of Cyprus, Nicosia, Cyprus
| | - Paris A. Skourides
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
21
|
Lian F, Li H, Ma Y, Zhou R, Wu W. Recent advances in primary cilia in bone metabolism. Front Endocrinol (Lausanne) 2023; 14:1259650. [PMID: 37886641 PMCID: PMC10598340 DOI: 10.3389/fendo.2023.1259650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/12/2023] [Indexed: 10/28/2023] Open
Abstract
Primary cilia are microtubule-based organelles that are widespread on the cell surface and play a key role in tissue development and homeostasis by sensing and transducing various signaling pathways. The process of intraflagellar transport (IFT), which is propelled by kinesin and dynein motors, plays a crucial role in the formation and functionality of cilia. Abnormalities in the cilia or ciliary transport system often cause a range of clinical conditions collectively known as ciliopathies, which include polydactyly, short ribs, scoliosis, thoracic stenosis and many abnormalities in the bones and cartilage. In this review, we summarize recent findings on the role of primary cilia and ciliary transport systems in bone development, we describe the role of cilia in bone formation, cartilage development and bone resorption, and we summarize advances in the study of primary cilia in fracture healing. In addition, the recent discovery of crosstalk between integrins and primary cilia provides new insights into how primary cilia affect bone.
Collapse
Affiliation(s)
- Fenfen Lian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Hui Li
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Yuwei Ma
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Rui Zhou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Wei Wu
- School of Athletic Performance, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
22
|
Masek M, Bachmann-Gagescu R. Control of protein and lipid composition of photoreceptor outer segments-Implications for retinal disease. Curr Top Dev Biol 2023; 155:165-225. [PMID: 38043951 DOI: 10.1016/bs.ctdb.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Vision is arguably our most important sense, and its loss brings substantial limitations to daily life for affected individuals. Light is perceived in retinal photoreceptors (PRs), which are highly specialized neurons subdivided into several compartments with distinct functions. The outer segments (OSs) of photoreceptors represent highly specialized primary ciliary compartments hosting the phototransduction cascade, which transforms incoming light into a neuronal signal. Retinal disease can result from various pathomechanisms originating in distinct subcompartments of the PR cell, or in the retinal pigment epithelium which supports the PRs. Dysfunction of primary cilia causes human disorders known as "ciliopathies", in which retinal disease is a common feature. This chapter focuses on PR OSs, discussing the mechanisms controlling their complex structure and composition. A sequence of tightly regulated sorting and trafficking events, both upstream of and within this ciliary compartment, ensures the establishment and maintenance of the adequate proteome and lipidome required for signaling in response to light. We discuss in particular our current understanding of the role of ciliopathy proteins involved in multi-protein complexes at the ciliary transition zone (CC2D2A) or BBSome (BBS1) and how their dysfunction causes retinal disease. While the loss of CC2D2A prevents the fusion of vesicles and delivery of the photopigment rhodopsin to the ciliary base, leading to early OS ultrastructural defects, BBS1 deficiency results in precocious accumulation of cholesterol in mutant OSs and decreased visual function preceding morphological changes. These distinct pathomechanisms underscore the central role of ciliary proteins involved in multiple processes controlling OS protein and lipid composition.
Collapse
Affiliation(s)
- Markus Masek
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Ruxandra Bachmann-Gagescu
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland; University Research Priority Program AdaBD, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
23
|
Cavini IA, Winter AJ, D’Muniz Pereira H, Woolfson DN, Crump MP, Garratt RC. X-ray structure of the metastable SEPT14-SEPT7 coiled coil reveals a hendecad region crucial for heterodimerization. Acta Crystallogr D Struct Biol 2023; 79:881-894. [PMID: 37712436 PMCID: PMC10565730 DOI: 10.1107/s2059798323006514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/27/2023] [Indexed: 09/16/2023] Open
Abstract
Septins are membrane-associated, GTP-binding proteins that are present in most eukaryotes. They polymerize to play important roles as scaffolds and/or diffusion barriers as part of the cytoskeleton. α-Helical coiled-coil domains are believed to contribute to septin assembly, and those observed in both human SEPT6 and SEPT8 form antiparallel homodimers. These are not compatible with their parallel heterodimeric organization expected from the current model for protofilament assembly, but they could explain the interfilament cross-bridges observed by microscopy. Here, the first structure of a heterodimeric septin coiled coil is presented, that between SEPT14 and SEPT7; the former is a SEPT6/SEPT8 homolog. This new structure is parallel, with two long helices that are axially shifted by a full helical turn with reference to their sequence alignment. The structure also has unusual knobs-into-holes packing of side chains. Both standard seven-residue (heptad) and the less common 11-residue (hendecad) repeats are present, creating two distinct regions with opposite supercoiling, which gives rise to an overall straight coiled coil. Part of the hendecad region is required for heterodimerization and therefore may be crucial for selective septin recognition. These unconventional sequences and structural features produce a metastable heterocomplex that nonetheless has enough specificity to promote correct protofilament assembly. For instance, the lack of supercoiling may facilitate unzipping and transitioning to the antiparallel homodimeric state.
Collapse
Affiliation(s)
- Italo A. Cavini
- São Carlos Institute of Physics, University of São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-120, Brazil
| | - Ashley J. Winter
- School of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, United Kingdom
| | - Humberto D’Muniz Pereira
- São Carlos Institute of Physics, University of São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-120, Brazil
| | - Derek N. Woolfson
- School of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, United Kingdom
- School of Biochemistry, University of Bristol, Medical Sciences Building, University Walk, Bristol BS8 1TD, United Kingdom
- BrisSynBio, University of Bristol, School of Chemistry, Bristol BS8 1TS, United Kingdom
| | - Matthew P. Crump
- School of Chemistry, University of Bristol, Cantock’s Close, Bristol BS8 1TS, United Kingdom
- BrisSynBio, University of Bristol, School of Chemistry, Bristol BS8 1TS, United Kingdom
| | - Richard C. Garratt
- São Carlos Institute of Physics, University of São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-120, Brazil
| |
Collapse
|
24
|
Nakazawa K, Chauvin B, Mangenot S, Bertin A. Reconstituted in vitro systems to reveal the roles and functions of septins. J Cell Sci 2023; 136:jcs259448. [PMID: 37815088 DOI: 10.1242/jcs.259448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023] Open
Abstract
Septins are essential cytoskeletal proteins involved in key cellular processes and have also been implicated in diseases from cancers to neurodegenerative pathologies. However, they have not been as thoroughly studied as other cytoskeletal proteins. In vivo, septins interact with other cytoskeletal proteins and with the inner plasma membrane. Hence, bottom-up in vitro cell-free assays are well suited to dissect the roles and behavior of septins in a controlled environment. Specifically, in vitro studies have been invaluable in describing the self-assembly of septins into a large diversity of ultrastructures. Given that septins interact specifically with membrane, the details of these septin-membrane interactions have been analyzed using reconstituted lipid systems. In particular, at a membrane, septins are often localized at curvatures of micrometer scale. In that context, in vitro assays have been performed with substrates of varying curvatures (spheres, cylinders or undulated substrates) to probe the sensitivity of septins to membrane curvature. This Review will first present the structural properties of septins in solution and describe the interplay of septins with cytoskeletal partners. We will then discuss how septins interact with biomimetic membranes and induce their reshaping. Finally, we will highlight the curvature sensitivity of septins and how they alter the mechanical properties of membranes.
Collapse
Affiliation(s)
- Koyomi Nakazawa
- Physico Chimie Curie , Institut Curie, CNRS UMR 168, Sorbonne Université, 11 Rue Pierre et Paris Curie, 75005 Paris, France
| | - Brieuc Chauvin
- Physico Chimie Curie , Institut Curie, CNRS UMR 168, Sorbonne Université, 11 Rue Pierre et Paris Curie, 75005 Paris, France
| | - Stéphanie Mangenot
- Laboratoire Matière et Systèmes Complexes , Université de Paris Cité, CNRS UMR 7057, 45 Rue des Saint Pères, 75006 Paris, France
| | - Aurélie Bertin
- Physico Chimie Curie , Institut Curie, CNRS UMR 168, Sorbonne Université, 11 Rue Pierre et Paris Curie, 75005 Paris, France
| |
Collapse
|
25
|
Saito M, Otsu W, Miyadera K, Nishimura Y. Recent advances in the understanding of cilia mechanisms and their applications as therapeutic targets. Front Mol Biosci 2023; 10:1232188. [PMID: 37780208 PMCID: PMC10538646 DOI: 10.3389/fmolb.2023.1232188] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/24/2023] [Indexed: 10/03/2023] Open
Abstract
The primary cilium is a single immotile microtubule-based organelle that protrudes into the extracellular space. Malformations and dysfunctions of the cilia have been associated with various forms of syndromic and non-syndromic diseases, termed ciliopathies. The primary cilium is therefore gaining attention due to its potential as a therapeutic target. In this review, we examine ciliary receptors, ciliogenesis, and ciliary trafficking as possible therapeutic targets. We first discuss the mechanisms of selective distribution, signal transduction, and physiological roles of ciliary receptors. Next, pathways that regulate ciliogenesis, specifically the Aurora A kinase, mammalian target of rapamycin, and ubiquitin-proteasome pathways are examined as therapeutic targets to regulate ciliogenesis. Then, in the photoreceptors, the mechanism of ciliary trafficking which takes place at the transition zone involving the ciliary membrane proteins is reviewed. Finally, some of the current therapeutic advancements highlighting the role of large animal models of photoreceptor ciliopathy are discussed.
Collapse
Affiliation(s)
- Masaki Saito
- Department of Molecular Physiology and Pathology, School of Pharma-Sciences, Teikyo University, Tokyo, Japan
| | - Wataru Otsu
- Department of Biomedical Research Laboratory, Gifu Pharmaceutical University, Gifu, Japan
| | - Keiko Miyadera
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
- Mie University Research Center for Cilia and Diseases, Tsu, Mie, Japan
| |
Collapse
|
26
|
Truong HM, Cruz-Colón KO, Martínez-Márquez JY, Willer JR, Travis AM, Biswas SK, Lo WK, Bolz HJ, Pearring JN. The tectonic complex regulates membrane protein composition in the photoreceptor cilium. Nat Commun 2023; 14:5671. [PMID: 37704658 PMCID: PMC10500017 DOI: 10.1038/s41467-023-41450-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 08/30/2023] [Indexed: 09/15/2023] Open
Abstract
The primary cilium is a signaling organelle with a unique membrane composition maintained by a diffusional barrier residing at the transition zone. Many transition zone proteins, such as the tectonic complex, are linked to preserving ciliary composition but the mechanism remains unknown. To understand tectonic's role, we generate a photoreceptor-specific Tctn1 knockout mouse. Loss of Tctn1 results in the absence of the entire tectonic complex and associated MKS proteins yet has minimal effects on the transition zone structure of rod photoreceptors. We find that the protein composition of the photoreceptor cilium is disrupted as non-resident membrane proteins accumulate in the cilium over time, ultimately resulting in photoreceptor degeneration. We further show that fluorescent rhodopsin moves faster through the transition zone in photoreceptors lacking tectonic, which suggests that the tectonic complex acts as a physical barrier to slow down membrane protein diffusion in the photoreceptor transition zone to ensure proper removal of non-resident membrane proteins.
Collapse
Affiliation(s)
- Hanh M Truong
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Kevin O Cruz-Colón
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | | | - Jason R Willer
- Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, MI, USA
| | - Amanda M Travis
- Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, MI, USA
| | - Sondip K Biswas
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Woo-Kuen Lo
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Hanno J Bolz
- Senckenberg Centre for Human Genetics, Frankfurt am Main, Germany
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany
| | - Jillian N Pearring
- Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
27
|
Yanda MK, Ciobanu C, Guggino WB, Cebotaru L. CFTR and PC2, partners in the primary cilia in autosomal dominant polycystic kidney disease. Am J Physiol Cell Physiol 2023; 325:C682-C693. [PMID: 37519231 PMCID: PMC10635646 DOI: 10.1152/ajpcell.00197.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
Defects in the primary cilium are associated with autosomal dominant polycystic kidney disease (ADPKD). We used a combination of animal models, Western blotting, and confocal microscopy and discovered that CFTR and polycystin 2 (PC2) are both colocalized to the cilium in normal kidneys, with the levels of both being decreased in cystic epithelia. Cilia were longer in CFTR-null mice and in cystic cells in our ADPKD animal models. We examined septin 2, known to play a role in cilia length, to act as a diffusion barrier and to serve as an enhancer of proliferation. We found that septin 2 protein levels were upregulated and colocalized strongly with CFTR in cystic cells. Application of VX-809, the CFTR corrector, restored CFTR and PC2 toward normal in the cilia, decreased the protein levels of septin 2, and drastically reduced septin 2 colocalization with CFTR. Our data suggest that CFTR is present in the cilia and plays a role there, perhaps through its conductance of Cl-. We also postulate that septin 2 is important for localizing CFTR to the apical membrane in cystic epithelia.NEW & NOTEWORTHY CFTR is present in the primary cilia together with polycystin 2 (PC2). Ablation of CFTR makes cilia longer suggesting that CFTR plays a role there, perhaps through its conductance of Cl.
Collapse
Affiliation(s)
- Murali K Yanda
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Cristian Ciobanu
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - William B Guggino
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Liudmila Cebotaru
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
28
|
de Freitas Fernandes A, Leonardo DA, Cavini IA, Rosa HVD, Vargas JA, D'Muniz Pereira H, Nascimento AS, Garratt RC. Conservation and divergence of the G-interfaces of Drosophila melanogaster septins. Cytoskeleton (Hoboken) 2023; 80:153-168. [PMID: 36576069 DOI: 10.1002/cm.21740] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022]
Abstract
Septins possess a conserved guanine nucleotide-binding (G) domain that participates in the stabilization of organized hetero-oligomeric complexes which assemble into filaments, rings and network-like structures. The fruit fly, Drosophila melanogaster, has five such septin genes encoding Sep1, Sep2, Sep4, Sep5 and Pnut. Here, we report the crystal structure of the heterodimer formed between the G-domains of Sep1 and Sep2, the first from an insect to be described to date. A G-interface stabilizes the dimer (in agreement with the expected arrangement for the Drosophila hexameric particle) and this bears significant resemblance to its human counterparts, even down to the level of individual amino acid interactions. On the other hand, a model for the G-interface formed between the two copies of Pnut which occupy the centre of the hexamer, shows important structural differences, including the loss of a highly favourable bifurcated salt-bridge network. Whereas wild-type Pnut purifies as a monomer, the reintroduction of the salt-bridge network results in stabilizing the dimeric interface in solution as shown by size exclusion chromatography and thermal stability measurements. Adaptive steered molecular dynamics reveals an unzipping mechanism for dimer dissociation which initiates at a point of electrostatic repulsion within the switch II region. Overall, the data contribute to a better understanding of the molecular interactions involved in septin assembly/disassembly.
Collapse
Affiliation(s)
| | | | | | | | - Jhon Antoni Vargas
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| | | | | | | |
Collapse
|
29
|
Polino AJ, Sviben S, Melena I, Piston DW, Hughes JW. Scanning electron microscopy of human islet cilia. Proc Natl Acad Sci U S A 2023; 120:e2302624120. [PMID: 37205712 PMCID: PMC10235940 DOI: 10.1073/pnas.2302624120] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/12/2023] [Indexed: 05/21/2023] Open
Abstract
Human islet primary cilia are vital glucose-regulating organelles whose structure remains uncharacterized. Scanning electron microscopy (SEM) is a useful technique for studying the surface morphology of membrane projections like cilia, but conventional sample preparation does not reveal the submembrane axonemal structure, which holds key implications for ciliary function. To overcome this challenge, we combined SEM with membrane-extraction techniques to examine primary cilia in native human islets. Our data show well-preserved cilia subdomains which demonstrate both expected and unexpected ultrastructural motifs. Morphometric features were quantified when possible, including axonemal length and diameter, microtubule conformations, and chirality. We further describe a ciliary ring, a structure that may be a specialization in human islets. Key findings are correlated with fluorescence microscopy and interpreted in the context of cilia function as a cellular sensor and communications locus in pancreatic islets.
Collapse
Affiliation(s)
- Alexander J. Polino
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO63110
| | - Sanja Sviben
- Washington University Center for Cellular Imaging, Washington University School of Medicine, Saint Louis, MO63110
| | - Isabella Melena
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO63110
| | - David W. Piston
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO63110
| | - Jing W. Hughes
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO63110
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO63110
| |
Collapse
|
30
|
Safavian D, Kim MS, Xie H, El-Zeiry M, Palander O, Dai L, Collins RF, Froese C, Shannon R, Nagata KI, Trimble WS. Septin-mediated RhoA activation engages the exocyst complex to recruit the cilium transition zone. J Cell Biol 2023; 222:e201911062. [PMID: 36912772 PMCID: PMC10039714 DOI: 10.1083/jcb.201911062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/25/2022] [Accepted: 01/05/2023] [Indexed: 03/14/2023] Open
Abstract
Septins are filamentous GTPases that play important but poorly characterized roles in ciliogenesis. Here, we show that SEPTIN9 regulates RhoA signaling at the base of cilia by binding and activating the RhoA guanine nucleotide exchange factor, ARHGEF18. GTP-RhoA is known to activate the membrane targeting exocyst complex, and suppression of SEPTIN9 causes disruption of ciliogenesis and mislocalization of an exocyst subunit, SEC8. Using basal body-targeted proteins, we show that upregulating RhoA signaling at the cilium can rescue ciliary defects and mislocalization of SEC8 caused by global SEPTIN9 depletion. Moreover, we demonstrate that the transition zone components, RPGRIP1L and TCTN2, fail to accumulate at the transition zone in cells lacking SEPTIN9 or depleted of the exocyst complex. Thus, SEPTIN9 regulates the recruitment of transition zone proteins on Golgi-derived vesicles by activating the exocyst via RhoA to allow the formation of primary cilia.
Collapse
Affiliation(s)
- Darya Safavian
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Moshe S. Kim
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Hong Xie
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maha El-Zeiry
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Oliva Palander
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Lu Dai
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Richard F. Collins
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Carol Froese
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rachel Shannon
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Koh-ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi, Japan
| | - William S. Trimble
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Bigge BM, Rosenthal NE, Avasthi P. Initial ciliary assembly in Chlamydomonas requires Arp2/3 complex-dependent endocytosis. Mol Biol Cell 2023; 34:ar24. [PMID: 36753382 PMCID: PMC10092647 DOI: 10.1091/mbc.e22-09-0443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Ciliary assembly, trafficking, and regulation are dependent on microtubules, but the mechanisms of ciliary assembly also require the actin cytoskeleton. Here, we dissect subcellular roles of actin in ciliogenesis by focusing on actin networks nucleated by the Arp2/3 complex in the powerful ciliary model, Chlamydomonas. We find that the Arp2/3 complex is required for the initial stages of ciliary assembly when protein and membrane are in high demand but cannot yet be supplied from the Golgi complex. We provide evidence for Arp2/3 complex-dependent endocytosis of ciliary proteins, an increase in endocytic activity upon induction of ciliary growth, and relocalization of plasma membrane proteins to newly formed cilia.
Collapse
Affiliation(s)
- Brae M Bigge
- Biochemistry and Cell Biology Department, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755; Anatomy and Cell Biology Department, University of Kansas Medical Center, Kansas City, KS 66103
| | - Nicholas E Rosenthal
- Biochemistry and Cell Biology Department, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755; Anatomy and Cell Biology Department, University of Kansas Medical Center, Kansas City, KS 66103
| | - Prachee Avasthi
- Biochemistry and Cell Biology Department, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755; Anatomy and Cell Biology Department, University of Kansas Medical Center, Kansas City, KS 66103
| |
Collapse
|
32
|
Devlin L, Dhondurao Sudhindar P, Sayer JA. Renal ciliopathies: promising drug targets and prospects for clinical trials. Expert Opin Ther Targets 2023; 27:325-346. [PMID: 37243567 DOI: 10.1080/14728222.2023.2218616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/12/2023] [Accepted: 05/23/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Renal ciliopathies represent a collection of genetic disorders characterized by deficiencies in the biogenesis, maintenance, or functioning of the ciliary complex. These disorders, which encompass autosomal dominant polycystic kidney disease (ADPKD), autosomal recessive polycystic kidney disease (ARPKD), and nephronophthisis (NPHP), typically result in cystic kidney disease, renal fibrosis, and a gradual deterioration of kidney function, culminating in kidney failure. AREAS COVERED Here we review the advances in basic science and clinical research into renal ciliopathies which have yielded promising small compounds and drug targets, within both preclinical studies and clinical trials. EXPERT OPINION Tolvaptan is currently the sole approved treatment option available for ADPKD patients, while no approved treatment alternatives exist for ARPKD or NPHP patients. Clinical trials are presently underway to evaluate additional medications in ADPKD and ARPKD patients. Based on preclinical models, other potential therapeutic targets for ADPKD, ARPKD, and NPHP look promising. These include molecules targeting fluid transport, cellular metabolism, ciliary signaling and cell-cycle regulation. There is a real and urgent clinical need for translational research to bring novel treatments to clinical use for all forms of renal ciliopathies to reduce kidney disease progression and prevent kidney failure.
Collapse
Affiliation(s)
- Laura Devlin
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Praveen Dhondurao Sudhindar
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - John A Sayer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
- Renal Services, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle Upon Tyne, UK
| |
Collapse
|
33
|
Polino AJ, Sviben S, Melena I, Piston DW, Hughes J. Scanning electron microscopy of human islet cilia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528685. [PMID: 36824775 PMCID: PMC9949088 DOI: 10.1101/2023.02.15.528685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Human islet primary cilia are vital glucose-regulating organelles whose structure remains uncharacterized. Scanning electron microscopy (SEM) is a useful technique for studying the surface morphology of membrane projections like primary cilia, but conventional sample preparation does not reveal the sub-membrane axonemal structure which holds key implications for cilia function. To overcome this challenge, we combined SEM with membrane-extraction techniques to examine cilia in native human islets. Our data show well-preserved cilia subdomains which demonstrate both expected and unexpected ultrastructural motifs. Morphometric features were quantified when possible, including axonemal length and diameter, microtubule conformations and chirality. We further describe a novel ciliary ring, a structure that may be a specialization in human islets. Key findings are correlated with fluorescence microscopy and interpreted in the context of cilia function as a cellular sensor and communications locus in pancreatic islets.
Collapse
|
34
|
Zheng NX, Miao YT, Zhang X, Huang MZ, Jahangir M, Luo S, Lang B. Primary cilia-associated protein IFT172 in ciliopathies. Front Cell Dev Biol 2023; 11:1074880. [PMID: 36733456 PMCID: PMC9887189 DOI: 10.3389/fcell.2023.1074880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023] Open
Abstract
Cilium is a highly conserved antenna-like structure protruding from the surface of the cell membrane, which is widely distributed on most mammalian cells. Two types of cilia have been described so far which include motile cilia and immotile cilia and the latter are also known as primary cilia. Dysfunctional primary cilia are commonly associated with a variety of congenital diseases called ciliopathies with multifaceted presentations such as retinopathy, congenital kidney disease, intellectual disability, cancer, polycystic kidney, obesity, Bardet Biedl syndrome (BBS), etc. Intraflagellar transport (IFT) is a bi-directional transportation process that helps maintain a balanced flow of proteins or signaling molecules essential for the communication between cilia and cytoplasm. Disrupted IFT contributes to the abnormal structure or function of cilia and frequently promotes the occurrence of ciliopathies. Intraflagellar transport 172 (IFT172) is a newly identified member of IFT proteins closely involved in some rare ciliopathies such as Mainzer-Saldino syndrome (MZSDS) and BBS, though the underpinning causal mechanisms remain largely elusive. In this review, we summarize the key findings on the genetic and protein characteristic of IFT172, as well as its function in intraflagellar transport, to provide comprehensive insights to understand IFT172-related ciliopathies.
Collapse
Affiliation(s)
- Nan-Xi Zheng
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ya-Ting Miao
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Mu-Zhi Huang
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Muhammad Jahangir
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shilin Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China,Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China,*Correspondence: Shilin Luo, ; Bing Lang,
| | - Bing Lang
- Department of Psychiatry, National Clinical Research Centre for Mental Health, The Second Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Shilin Luo, ; Bing Lang,
| |
Collapse
|
35
|
Benoit B, Poüs C, Baillet A. Septins as membrane influencers: direct play or in association with other cytoskeleton partners. Front Cell Dev Biol 2023; 11:1112319. [PMID: 36875762 PMCID: PMC9982393 DOI: 10.3389/fcell.2023.1112319] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/23/2023] [Indexed: 02/19/2023] Open
Abstract
The cytoskeleton comprises three polymerizing structures that have been studied for a long time, actin microfilaments, microtubules and intermediate filaments, plus more recently investigated dynamic assemblies like septins or the endocytic-sorting complex required for transport (ESCRT) complex. These filament-forming proteins control several cell functions through crosstalks with each other and with membranes. In this review, we report recent works that address how septins bind to membranes, and influence their shaping, organization, properties and functions, either by binding to them directly or indirectly through other cytoskeleton elements.
Collapse
Affiliation(s)
- Béatrice Benoit
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
| | - Christian Poüs
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France.,Laboratoire de Biochimie-Hormonologie, Hôpital Antoine Béclère, AP-HP, Hôpitaux Universitaires Paris-Saclay, Clamart, France
| | - Anita Baillet
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
| |
Collapse
|
36
|
Park K, Leroux MR. Composition, organization and mechanisms of the transition zone, a gate for the cilium. EMBO Rep 2022; 23:e55420. [PMID: 36408840 PMCID: PMC9724682 DOI: 10.15252/embr.202255420] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/08/2022] [Accepted: 10/31/2022] [Indexed: 11/22/2022] Open
Abstract
The cilium evolved to provide the ancestral eukaryote with the ability to move and sense its environment. Acquiring these functions required the compartmentalization of a dynein-based motility apparatus and signaling proteins within a discrete subcellular organelle contiguous with the cytosol. Here, we explore the potential molecular mechanisms for how the proximal-most region of the cilium, termed transition zone (TZ), acts as a diffusion barrier for both membrane and soluble proteins and helps to ensure ciliary autonomy and homeostasis. These include a unique complement and spatial organization of proteins that span from the microtubule-based axoneme to the ciliary membrane; a protein picket fence; a specialized lipid microdomain; differential membrane curvature and thickness; and lastly, a size-selective molecular sieve. In addition, the TZ must be permissive for, and functionally integrates with, ciliary trafficking systems (including intraflagellar transport) that cross the barrier and make the ciliary compartment dynamic. The quest to understand the TZ continues and promises to not only illuminate essential aspects of human cell signaling, physiology, and development, but also to unravel how TZ dysfunction contributes to ciliopathies that affect multiple organ systems, including eyes, kidney, and brain.
Collapse
Affiliation(s)
- Kwangjin Park
- Department of Molecular Biology and BiochemistrySimon Fraser UniversityBurnabyBCCanada
- Centre for Cell Biology, Development, and DiseaseSimon Fraser UniversityBurnabyBCCanada
- Present address:
Terry Fox LaboratoryBC CancerVancouverBCCanada
- Present address:
Department of Medical GeneticsUniversity of British ColumbiaVancouverBCCanada
| | - Michel R Leroux
- Department of Molecular Biology and BiochemistrySimon Fraser UniversityBurnabyBCCanada
- Centre for Cell Biology, Development, and DiseaseSimon Fraser UniversityBurnabyBCCanada
| |
Collapse
|
37
|
Wang G, Zhu X, Gao Y, Lv M, Li K, Tang D, Wu H, Xu C, Geng H, Shen Q, Zha X, Duan Z, Zhang J, Hua R, Tao F, Zhou P, Wei Z, Cao Y, Guo R, He X. Biallelic loss-of-function mutations in SEPTIN4 (C17ORF47), encoding a conserved annulus protein, cause thin midpiece spermatozoa and male infertility in humans. Hum Mutat 2022; 43:2079-2090. [PMID: 36135717 DOI: 10.1002/humu.24475] [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] [Received: 07/18/2022] [Revised: 08/29/2022] [Accepted: 09/14/2022] [Indexed: 01/25/2023]
Abstract
Asthenoteratozoospermia is the primary cause of infertility in humans. However, the genetic etiology remains largely unknown for those suffering from severe asthenoteratozoospermia caused by thin midpiece defects. In this study, we identified two biallelic loss-of-function variants of SEPTIN4 (previously SEPT4) (Patient 1: c.A721T, p.R241* and Patient 2: c.C205T, p.R69*) in two unrelated individuals from two consanguineous Chinese families. SEPT4 is a conserved annulus protein that is critical for male fertility and the structural integrity of the sperm midpiece in mice. SEPT4 mutations disrupted the formation of SEPT-based annulus and localization of SEPTIN subunits in sperms from patients. The ultrastructural analysis demonstrated striking thin midpiece spermatozoa defects owing to annulus loss and disorganized mitochondrial sheath. Immunofluorescence and immunoblotting analyses of the mitochondrial sheath proteins TOMM20 and HSP60 further indicated that the distribution and abundance of mitochondria were impaired in men harboring biallelic SEPT4 variants. Additionally, we found that the precise localization of SLC26A8, a testis-specific anion transporter that colocalizes with SEPT4 at the sperm annulus, was missing without SEPT4. Moreover, the patient achieved a good pregnancy outcome following intracytoplasmic sperm injection. Overall, our study demonstrated for the first time that SEPT4 variants that induced thin midpiece spermatozoa defects were directly associated with human asthenoteratozoospermia.
Collapse
Affiliation(s)
- Guanxiong Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Xiaoyu Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Yang Gao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Mingrong Lv
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Kuokuo Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Dongdong Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Huan Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Chuan Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Hao Geng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Qunshan Shen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Anhui Provincial Human Sperm Bank, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaomin Zha
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Zongliu Duan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Jingjing Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Rong Hua
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Fangbiao Tao
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Ping Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Zhaolian Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Rui Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Xiaojin He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, Hefei, Anhui, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China.,Anhui Provincial Engineering Research Center of Biopreservation and Artificial Organs, Hefei, Anhui, China.,Anhui Provincial Human Sperm Bank, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
38
|
Xu K, Ou G. Cilia regeneration requires an RNA splicing factor from the ciliary base. CELL REGENERATION (LONDON, ENGLAND) 2022; 11:29. [PMID: 36180752 PMCID: PMC9525525 DOI: 10.1186/s13619-022-00130-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/19/2022] [Indexed: 11/10/2022]
Abstract
Cilia are microtubule-based organelles projected from most eukaryotic cell surfaces performing cell motility and signaling. Several previously recognized non-ciliary proteins play crucial roles in cilium formation and function. Here, we provide additional evidence that the Caenorhabditis elegans RNA splicing factor PRP-8/PRPF8 regulates ciliogenesis and regeneration from the ciliary base. Live imaging of GFP knock-in animals reveals that the endogenous PRP-8 localizes in the nuclei and the ciliary base. A weak loss-of-function allele of prp-8 affects ciliary structure but with little impact on RNA splicing. Conditional degradation of PRP-8 within ciliated sensory neurons showed its direct and specific roles in cilium formation. Notably, the penetrance of ciliary defects correlates with the reduction of PRP-8 at the ciliary base but not nuclei, and sensory neurons regenerated cilia accompanying PRP-8 recovery from the ciliary base rather than the nuclei. We suggest that PRP-8 at the ciliary base contributes to cilium formation and regeneration.
Collapse
Affiliation(s)
- Kaiming Xu
- grid.12527.330000 0001 0662 3178Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| | - Guangshuo Ou
- grid.12527.330000 0001 0662 3178Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, McGovern Institute for Brain Research, School of Life Sciences and MOE Key Laboratory for Protein Science, Tsinghua University, Beijing, China
| |
Collapse
|
39
|
van den Hoek H, Klena N, Jordan MA, Alvarez Viar G, Righetto RD, Schaffer M, Erdmann PS, Wan W, Geimer S, Plitzko JM, Baumeister W, Pigino G, Hamel V, Guichard P, Engel BD. In situ architecture of the ciliary base reveals the stepwise assembly of intraflagellar transport trains. Science 2022; 377:543-548. [PMID: 35901159 DOI: 10.1126/science.abm6704] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cilium is an antenna-like organelle that performs numerous cellular functions, including motility, sensing, and signaling. The base of the cilium contains a selective barrier that regulates the entry of large intraflagellar transport (IFT) trains, which carry cargo proteins required for ciliary assembly and maintenance. However, the native architecture of the ciliary base and the process of IFT train assembly remain unresolved. In this work, we used in situ cryo-electron tomography to reveal native structures of the transition zone region and assembling IFT trains at the ciliary base in Chlamydomonas. We combined this direct cellular visualization with ultrastructure expansion microscopy to describe the front-to-back stepwise assembly of IFT trains: IFT-B forms the backbone, onto which bind IFT-A, dynein-1b, and finally kinesin-2 before entry into the cilium.
Collapse
Affiliation(s)
- Hugo van den Hoek
- Biozentrum, University of Basel, 4056 Basel, Switzerland.,Helmholtz Pioneer Campus, Helmholtz Munich, 85764 Neuherberg, Germany.,Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Nikolai Klena
- Department of Molecular and Cellular Biology, Section of Biology, University of Geneva, 1211 Geneva, Switzerland.,Human Technopole, 20157 Milan, Italy
| | - Mareike A Jordan
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Gonzalo Alvarez Viar
- Human Technopole, 20157 Milan, Italy.,Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Ricardo D Righetto
- Biozentrum, University of Basel, 4056 Basel, Switzerland.,Helmholtz Pioneer Campus, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Miroslava Schaffer
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | | | - William Wan
- Department of Biochemistry and Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Stefan Geimer
- Cell Biology and Electron Microscopy, University of Bayreuth, 95447 Bayreuth, Germany
| | - Jürgen M Plitzko
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Wolfgang Baumeister
- Department of Molecular Structural Biology, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Gaia Pigino
- Human Technopole, 20157 Milan, Italy.,Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Virginie Hamel
- Department of Molecular and Cellular Biology, Section of Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Paul Guichard
- Department of Molecular and Cellular Biology, Section of Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Benjamin D Engel
- Biozentrum, University of Basel, 4056 Basel, Switzerland.,Helmholtz Pioneer Campus, Helmholtz Munich, 85764 Neuherberg, Germany
| |
Collapse
|
40
|
Keppner A, Correia M, Santambrogio S, Koay TW, Maric D, Osterhof C, Winter DV, Clerc A, Stumpe M, Chalmel F, Dewilde S, Odermatt A, Kressler D, Hankeln T, Wenger RH, Hoogewijs D. Androglobin, a chimeric mammalian globin, is required for male fertility. eLife 2022; 11:72374. [PMID: 35700329 PMCID: PMC9249397 DOI: 10.7554/elife.72374] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Spermatogenesis is a highly specialized differentiation process driven by a dynamic gene expression program and ending with the production of mature spermatozoa. Whereas hundreds of genes are known to be essential for male germline proliferation and differentiation, the contribution of several genes remains uncharacterized. The predominant expression of the latest globin family member, androglobin (Adgb), in mammalian testis tissue prompted us to assess its physiological function in spermatogenesis. Adgb knockout mice display male infertility, reduced testis weight, impaired maturation of elongating spermatids, abnormal sperm shape, and ultrastructural defects in microtubule and mitochondrial organization. Epididymal sperm from Adgb knockout animals display multiple flagellar malformations including coiled, bifid or shortened flagella, and erratic acrosomal development. Following immunoprecipitation and mass spectrometry, we could identify septin 10 (Sept10) as interactor of Adgb. The Sept10-Adgb interaction was confirmed both in vivo using testis lysates and in vitro by reciprocal co-immunoprecipitation experiments. Furthermore, the absence of Adgb leads to mislocalization of Sept10 in sperm, indicating defective manchette and sperm annulus formation. Finally, in vitro data suggest that Adgb contributes to Sept10 proteolysis in a calmodulin-dependent manner. Collectively, our results provide evidence that Adgb is essential for murine spermatogenesis and further suggest that Adgb is required for sperm head shaping via the manchette and proper flagellum formation.
Collapse
Affiliation(s)
- Anna Keppner
- Department of Endocrinology, Metabolism and Cardiovascular system, University of Fribourg, Fribourg, Switzerland
| | - Miguel Correia
- Department of Endocrinology, Metabolism and Cardiovascular system, University of Fribourg, Fribourg, Switzerland
| | | | - Teng Wei Koay
- Department of Endocrinology, Metabolism and Cardiovascular system, University of Fribourg, Fribourg, Switzerland
| | - Darko Maric
- Department of Endocrinology, Metabolism and Cardiovascular system, University of Fribourg, Fribourg, Switzerland
| | - Carina Osterhof
- Institute for Organismic and Molecular Evolutionary Biology, University of Mainz, Mainz, Germany
| | - Denise V Winter
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Angèle Clerc
- Department of Endocrinology, Metabolism and Cardiovascular system, University of Fribourg, Fribourg, Switzerland
| | - Michael Stumpe
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Sylvia Dewilde
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Alex Odermatt
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Dieter Kressler
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Thomas Hankeln
- Institute for Organismic and Molecular Evolutionary Biology, University of Mainz, Mainz, Germany
| | - Roland H Wenger
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - David Hoogewijs
- Department of Endocrinology, Metabolism and Cardiovascular system, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
41
|
Kumari A, Ghosh A, Kolay S, Raghu P. Septins tune lipid kinase activity and PI(4,5)P 2 turnover during G-protein–coupled PLC signalling in vivo. Life Sci Alliance 2022; 5:5/6/e202101293. [PMID: 35277468 PMCID: PMC8921834 DOI: 10.26508/lsa.202101293] [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] [Received: 11/09/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 11/24/2022] Open
Abstract
Phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] hydrolysis by phospholipase C (PLC) is a conserved mechanism of signalling. Given the low abundance of PI(4,5)P2, its hydrolysis needs to be coupled to resynthesis to ensure continued PLC activity; however, the mechanism by which depletion is coupled to resynthesis remains unknown. PI(4,5)P2 synthesis is catalyzed by the phosphorylation of phosphatidylinositol 4 phosphate (PI4P) by phosphatidylinositol 4 phosphate 5 kinase (PIP5K). In Drosophila photoreceptors, photon absorption is transduced into PLC activity and during this process, PI(4,5)P2 is resynthesized by a PIP5K. However, the mechanism by which PIP5K activity is coupled to PI(4,5)P2 hydrolysis is unknown. In this study, we identify a unique isoform dPIP5KL, that is both necessary and sufficient to mediate PI(4,5)P2 synthesis during phototransduction. Depletion of PNUT, a non-redundant subunit of the septin family, enhances dPIP5KL activity in vitro and PI(4,5)P2 resynthesis in vivo; co-depletion of dPIP5KL reverses the enhanced rate of PI(4,5)P2 resynthesis in vivo. Thus, our work defines a septin-mediated mechanism through which PIP5K activity is coupled to PLC-mediated PI(4,5)P2 hydrolysis.
Collapse
Affiliation(s)
- Aastha Kumari
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bengaluru, India
| | - Avishek Ghosh
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bengaluru, India
- Department of Surgery, Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sourav Kolay
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bengaluru, India
- UT Southwestern Medical Center, Dallas, TX, USA
| | - Padinjat Raghu
- National Centre for Biological Sciences, TIFR-GKVK Campus, Bengaluru, India
| |
Collapse
|
42
|
Shi H, Wang H, Zhang C, Lu Y, Yao J, Chen Z, Xing G, Wei Q, Cao X. Mutations in OSBPL2 cause hearing loss associated with primary cilia defects via Sonic Hedgehog signaling. JCI Insight 2022; 7:149626. [PMID: 35041619 PMCID: PMC8876550 DOI: 10.1172/jci.insight.149626] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 01/12/2022] [Indexed: 11/17/2022] Open
Abstract
Defective primary cilia cause a range of diseases called ciliopathies, which include hearing loss (HL). Variants in the human oxysterol-binding protein like 2 (OSBPL2/ORP2) are responsible for autosomal dominant nonsyndromic HL (DFNA67). However, the pathogenesis of OSBPL2 deficiency has not been fully elucidated. In this study, we show that the Osbpl2-KO mice exhibited progressive HL and abnormal cochlear development with defective cilia. Further research revealed that OSBPL2 was located at the base of the kinocilia in hair cells (HCs) and primary cilia in supporting cells (SCs) and functioned in the maintenance of ciliogenesis by regulating the homeostasis of PI(4,5)P2 (phosphatidylinositol 4,5-bisphosphate) on the cilia membrane. OSBPL2 deficiency led to a significant increase of PI(4,5)P2 on the cilia membrane, which could be partially rescued by the overexpression of INPP5E. In addition, smoothened and GL13, the key molecules in the Sonic Hedgehog (Shh) signaling pathway, were detected to be downregulated in Osbpl2-KO HEI-OC1 cells. Our findings revealed that OSBPL2 deficiency resulted in ciliary defects and abnormal Shh signaling transduction in auditory cells, which helped to elucidate the underlying mechanism of OSBPL2 deficiency in HL.
Collapse
Affiliation(s)
- Hairong Shi
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Hongshun Wang
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Cheng Zhang
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Yajie Lu
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Jun Yao
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Zhibin Chen
- Department of Otolaryngology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guangqian Xing
- Department of Otolaryngology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qinjun Wei
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| | - Xin Cao
- Department of Medical Genetics, Nanjing Medical Univeristy, Nanjing, China
| |
Collapse
|
43
|
Kanamaru T, Neuner A, Kurtulmus B, Pereira G. Balancing the length of the distal tip by septins is key for stability and signalling function of primary cilia. EMBO J 2022; 41:e108843. [PMID: 34981518 PMCID: PMC8724769 DOI: 10.15252/embj.2021108843] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 01/08/2023] Open
Abstract
Primary cilia are antenna-like organelles required for signalling transduction. How cilia structure is mechanistically maintained at steady-state to promote signalling is largely unknown. Here, we define that mammalian primary cilia axonemes are formed by proximal segment (PS) and distal segment (DS) delineated by tubulin polyglutamylation-rich and -poor regions, respectively. The analysis of proximal/distal segmentation indicated that perturbations leading to cilia over-elongation influenced PS or DS length with a different impact on cilia behaviour. We identified septins as novel repressors of DS growth. We show that septins control the localisation of MKS3 and CEP290 required for a functional transition zone (TZ), and the cilia tip accumulation of the microtubule-capping kinesin KIF7, a cilia-growth inhibitor. Live-cell imaging and analysis of sonic-hedgehog (SHH) signalling activation established that DS over-extension increased cilia ectocytosis events and decreased SHH activation. Our data underlines the importance of understanding cilia segmentation for length control and cilia-dependent signalling.
Collapse
Affiliation(s)
- Taishi Kanamaru
- Centre for Organismal Studies (COS)University of HeidelbergHeidelbergGermany
- German Cancer Research Centre (DKFZ)DKFZ‐ZMBH AllianceHeidelbergGermany
- Centre for Molecular Biology (ZMBH)University of HeidelbergHeidelbergGermany
| | - Annett Neuner
- Centre for Molecular Biology (ZMBH)University of HeidelbergHeidelbergGermany
| | - Bahtiyar Kurtulmus
- Centre for Organismal Studies (COS)University of HeidelbergHeidelbergGermany
- German Cancer Research Centre (DKFZ)DKFZ‐ZMBH AllianceHeidelbergGermany
- Centre for Molecular Biology (ZMBH)University of HeidelbergHeidelbergGermany
| | - Gislene Pereira
- Centre for Organismal Studies (COS)University of HeidelbergHeidelbergGermany
- German Cancer Research Centre (DKFZ)DKFZ‐ZMBH AllianceHeidelbergGermany
- Centre for Molecular Biology (ZMBH)University of HeidelbergHeidelbergGermany
| |
Collapse
|
44
|
Cavini IA, Leonardo DA, Rosa HVD, Castro DKSV, D'Muniz Pereira H, Valadares NF, Araujo APU, Garratt RC. The Structural Biology of Septins and Their Filaments: An Update. Front Cell Dev Biol 2021; 9:765085. [PMID: 34869357 PMCID: PMC8640212 DOI: 10.3389/fcell.2021.765085] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/27/2021] [Indexed: 01/22/2023] Open
Abstract
In order to fully understand any complex biochemical system from a mechanistic point of view, it is necessary to have access to the three-dimensional structures of the molecular components involved. Septins and their oligomers, filaments and higher-order complexes are no exception. Indeed, the spontaneous recruitment of different septin monomers to specific positions along a filament represents a fascinating example of subtle molecular recognition. Over the last few years, the amount of structural information available about these important cytoskeletal proteins has increased dramatically. This has allowed for a more detailed description of their individual domains and the different interfaces formed between them, which are the basis for stabilizing higher-order structures such as hexamers, octamers and fully formed filaments. The flexibility of these structures and the plasticity of the individual interfaces have also begun to be understood. Furthermore, recently, light has been shed on how filaments may bundle into higher-order structures by the formation of antiparallel coiled coils involving the C-terminal domains. Nevertheless, even with these advances, there is still some way to go before we fully understand how the structure and dynamics of septin assemblies are related to their physiological roles, including their interactions with biological membranes and other cytoskeletal components. In this review, we aim to bring together the various strands of structural evidence currently available into a more coherent picture. Although it would be an exaggeration to say that this is complete, recent progress seems to suggest that headway is being made in that direction.
Collapse
Affiliation(s)
- Italo A Cavini
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| | - Diego A Leonardo
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| | - Higor V D Rosa
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| | - Danielle K S V Castro
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil.,São Carlos Institute of Chemistry, University of São Paulo, São Carlos, Brazil
| | | | | | - Ana P U Araujo
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| | - Richard C Garratt
- São Carlos Institute of Physics, University of São Paulo, São Carlos, Brazil
| |
Collapse
|
45
|
Stochasticity and positive feedback enable enzyme kinetics at the membrane to sense reaction size. Proc Natl Acad Sci U S A 2021; 118:2103626118. [PMID: 34789575 PMCID: PMC8617498 DOI: 10.1073/pnas.2103626118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2021] [Indexed: 12/27/2022] Open
Abstract
Cellular membranes span a wide range of spatial dimensions, from the plasma membrane with a scale of microns to vesicles on the nanometer scale. The work presented here identifies a molecular mechanism, based on common features of cellular signaling enzymes, that causes the average enzymatic catalytic rate to exhibit reaction size dependency. This effect stems from stochastic variation, but the final results can be essentially deterministic. In competitive enzymatic reaction cycles, the final product can depend on the size of the reaction system. The simplicity of the mechanism suggests that size-dependent reaction rates may be widespread among signaling enzymes and thus enable reaction size to be an important factor in signal regulation at the membrane. Here, we present detailed kinetic analyses of a panel of soluble lipid kinases and phosphatases, as well as Ras activating proteins, acting on their respective membrane surface substrates. The results reveal that the mean catalytic rate of such interfacial enzymes can exhibit a strong dependence on the size of the reaction system—in this case membrane area. Experimental measurements and kinetic modeling reveal how stochastic effects stemming from low molecular copy numbers of the enzymes alter reaction kinetics based on mechanistic characteristics of the enzyme, such as positive feedback. For the competitive enzymatic cycles studied here, the final product—consisting of a specific lipid composition or Ras activity state—depends on the size of the reaction system. Furthermore, we demonstrate how these reaction size dependencies can be controlled by engineering feedback mechanisms into the enzymes.
Collapse
|
46
|
Comparison of Ciliary Targeting of Two Rhodopsin-Like GPCRs: Role of C-Terminal Localization Sequences in Relation to Cilium Type. J Neurosci 2021; 41:7514-7531. [PMID: 34301828 PMCID: PMC8425976 DOI: 10.1523/jneurosci.0357-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/20/2021] [Accepted: 06/16/2021] [Indexed: 11/21/2022] Open
Abstract
Primary cilia exhibit a distinct complement of proteins, including G-protein-coupled receptors (GPCRs) that mediate sensory and developmental signals. The localization of GPCRs to the ciliary membrane involves ciliary localization sequences (CLSs), but it is not known how CLSs might relate to cilium type. Here, we studied the localization of two rhodopsin (RHO)-like GPCRs, somatostatin receptor (SSTR3) and RHO, in three types of cilia, from inner medullary collecting duct (IMCD3) cells, hTERT-RPE1 cells (possessing pocket cilia), and rod photoreceptors (whose cilia grow into elaborate phototransductive outer segments). SSTR3 was localized specifically to all three types of cilia, whereas RHO showed more selectivity for the photoreceptor cilium. Focusing on C-terminal CLSs, we characterized a novel CLS in the SSTR3 C terminus, which was required for the robust ciliary localization of SSTR3. Replacing the C terminus of RHO with this SSTR3 CLS-enhanced ciliary localization, compared with full-length RHO in IMCD3 and hTERT-RPE1 cells. Addition of the SSTR3 CLS to the single transmembrane protein CD8A enabled ciliary localization. In hTERT-RPE1 cells, a partial SSTR3 CLS added to CD8A effected specific localization to the periciliary (pocket) membrane, demonstrating C-terminal localization sequence targeting to this domain. Using retinas from mice, including both sexes, we show that deletion of the C terminus of RHO reduced the rod outer segment localization and that addition of the SSTR3 C-terminal CLS to the truncated RHO partly rescued this mislocalization. Overall, the study details elements of the different C termini of SSTR3 and RHO that are major effectors in determining specificity of cilium (or pericilium) localization among different types of cilia.SIGNIFICANCE STATEMENT The localization of G-protein-coupled receptors to primary cilia is key to many types of signal transduction. After characterizing a novel C-terminal CLS in SSTR3, we investigated how SSTR3 and RHO localization to the cilium relates to C-terminal CLSs and to cilium type. We found that the SSTR3 C-terminal CLS was effective in three different types of cilia, but the RHO C terminus showed a clear localization preference for the highly elaborate photoreceptor cilium. When added to CD8A, part of the SSTR3 CLS promoted specific periciliary membrane localization in hTERT-RPE1 cells, demonstrating an effective CLS for this domain. Thus, we demonstrate that elements of the C termini of SSTR3 and RHO determine different localization patterns among different types of cilia.
Collapse
|
47
|
Vial A, Taveneau C, Costa L, Chauvin B, Nasrallah H, Godefroy C, Dosset P, Isambert H, Ngo KX, Mangenot S, Levy D, Bertin A, Milhiet PE. Correlative AFM and fluorescence imaging demonstrate nanoscale membrane remodeling and ring-like and tubular structure formation by septins. NANOSCALE 2021; 13:12484-12493. [PMID: 34225356 DOI: 10.1039/d1nr01978c] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Septins are ubiquitous cytoskeletal filaments that interact with the inner plasma membrane and are essential for cell division in eukaryotes. In cellular contexts, septins are often localized at micrometric Gaussian curvatures, where they assemble onto ring-like structures. The behavior of budding yeast septins depends on their specific interaction with inositol phospholipids, enriched at the inner leaflet of the plasma membrane. Septin filaments are built from the non-polar self-assembly of short rods into filaments. However, the molecular mechanisms regulating the interplay with the inner plasma membrane and the resulting interaction with specific curvatures are not fully understood. In this report, we have imaged dynamical molecular assemblies of budding yeast septins on PIP2-containing supported lipid bilayers using a combination of high-speed AFM and correlative AFM-fluorescence microscopy. Our results clearly demonstrate that septins are able to bind to flat supported lipid bilayers and thereafter induce the remodeling of membranes. Short septin rods (octamers subunits) can indeed destabilize supported lipid bilayers and reshape the membrane to form 3D structures such as rings and tubes, demonstrating that long filaments are not necessary for septin-induced membrane buckling.
Collapse
Affiliation(s)
- Anthony Vial
- Centre de Biochimie Structurale (CBS), Université de Montpellier, CNRS, INSERM, 34090 Montpellier, France.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Gould NR, Torre OM, Leser JM, Stains JP. The cytoskeleton and connected elements in bone cell mechano-transduction. Bone 2021; 149:115971. [PMID: 33892173 PMCID: PMC8217329 DOI: 10.1016/j.bone.2021.115971] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/30/2021] [Accepted: 04/17/2021] [Indexed: 02/07/2023]
Abstract
Bone is a mechano-responsive tissue that adapts to changes in its mechanical environment. Increases in strain lead to increased bone mass acquisition, whereas decreases in strain lead to a loss of bone mass. Given that mechanical stress is a regulator of bone mass and quality, it is important to understand how bone cells sense and transduce these mechanical cues into biological changes to identify druggable targets that can be exploited to restore bone cell mechano-sensitivity or to mimic mechanical load. Many studies have identified individual cytoskeletal components - microtubules, actin, and intermediate filaments - as mechano-sensors in bone. However, given the high interconnectedness and interaction between individual cytoskeletal components, and that they can assemble into multiple discreet cellular structures, it is likely that the cytoskeleton as a whole, rather than one specific component, is necessary for proper bone cell mechano-transduction. This review will examine the role of each cytoskeletal element in bone cell mechano-transduction and will present a unified view of how these elements interact and work together to create a mechano-sensor that is necessary to control bone formation following mechanical stress.
Collapse
Affiliation(s)
- Nicole R Gould
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Olivia M Torre
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jenna M Leser
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joseph P Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA..
| |
Collapse
|
49
|
Jana SC, Dutta P, Jain A, Singh A, Adusumilli L, Girotra M, Kumari D, Shirolikar S, Ray K. Kinesin-2 transports Orco into the olfactory cilium of Drosophila melanogaster at specific developmental stages. PLoS Genet 2021; 17:e1009752. [PMID: 34411092 PMCID: PMC8407544 DOI: 10.1371/journal.pgen.1009752] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 08/31/2021] [Accepted: 07/29/2021] [Indexed: 12/21/2022] Open
Abstract
The cilium, the sensing centre for the cell, displays an extensive repertoire of receptors for various cell signalling processes. The dynamic nature of ciliary signalling indicates that the ciliary entry of receptors and associated proteins must be regulated and conditional. To understand this process, we studied the ciliary localisation of the odour-receptor coreceptor (Orco), a seven-pass transmembrane protein essential for insect olfaction. Little is known about when and how Orco gets into the cilia. Here, using Drosophila melanogaster, we show that the bulk of Orco selectively enters the cilia on adult olfactory sensory neurons in two discrete, one-hour intervals after eclosion. A conditional loss of heterotrimeric kinesin-2 during this period reduces the electrophysiological response to odours and affects olfactory behaviour. We further show that Orco binds to the C-terminal tail fragments of the heterotrimeric kinesin-2 motor, which is required to transfer Orco from the ciliary base to the outer segment and maintain within an approximately four-micron stretch at the distal portion of the ciliary outer-segment. The Orco transport was not affected by the loss of critical intraflagellar transport components, IFT172/Oseg2 and IFT88/NompB, respectively, during the adult stage. These results highlight a novel developmental regulation of seven-pass transmembrane receptor transport into the cilia and indicate that ciliary signalling is both developmentally and temporally regulated.
Collapse
Affiliation(s)
- Swadhin Chandra Jana
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- Instituto Gulbenkian de Ciência (IGC), Oeiras, Portugal
| | - Priya Dutta
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Akanksha Jain
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Anjusha Singh
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Lavanya Adusumilli
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Mukul Girotra
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Diksha Kumari
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Seema Shirolikar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Krishanu Ray
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
50
|
Focșa IO, Budișteanu M, Bălgrădean M. Clinical and genetic heterogeneity of primary ciliopathies (Review). Int J Mol Med 2021; 48:176. [PMID: 34278440 PMCID: PMC8354309 DOI: 10.3892/ijmm.2021.5009] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/28/2021] [Indexed: 01/11/2023] Open
Abstract
Ciliopathies comprise a group of complex disorders, with involvement of the majority of organs and systems. In total, >180 causal genes have been identified and, in addition to Mendelian inheritance, oligogenicity, genetic modifications, epistatic interactions and retrotransposon insertions have all been described when defining the ciliopathic phenotype. It is remarkable how the structural and functional impairment of a single, minuscule organelle may lead to the pathogenesis of highly pleiotropic diseases. Thus, combined efforts have been made to identify the genetic substratum and to determine the pathophysiological mechanism underlying the clinical presentation, in order to diagnose and classify ciliopathies. Yet, predicting the phenotype, given the intricacy of the genetic cause and overlapping clinical characteristics, represents a major challenge. In the future, advances in proteomics, cell biology and model organisms may provide new insights that could remodel the field of ciliopathies.
Collapse
Affiliation(s)
- Ina Ofelia Focșa
- Department of Medical Genetics, University of Medicine and Pharmacy 'Carol Davila', 021901 Bucharest, Romania
| | - Magdalena Budișteanu
- Department of Pediatric Neurology, 'Prof. Dr. Alexandru Obregia' Clinical Hospital of Psychiatry, 041914 Bucharest, Romania
| | - Mihaela Bălgrădean
- Department of Pediatrics and Pediatric Nephrology, Emergency Clinical Hospital for Children 'Maria Skłodowska Curie', 077120 Bucharest, Romania
| |
Collapse
|