1
|
Short KL, Lao J, Lam R, Moreau JLM, Ng J, Piran M, Combes AN, Cottle DL, Cole TJ. Disrupted glucocorticoid receptor cell signalling causes a ciliogenesis defect in the fetal mouse renal tubule. EMBO Rep 2025:10.1038/s44319-025-00454-0. [PMID: 40247090 DOI: 10.1038/s44319-025-00454-0] [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: 07/24/2024] [Revised: 03/14/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025] Open
Abstract
Primary cilia are cell signalling and environment sensing organelles and have important roles during embryogenesis and homeostasis. We demonstrate glucocorticoid signalling is essential for normal cilia formation in mouse and human renal tubules. RNA sequencing of E18.5 kidneys from glucocorticoid receptor (GR) null mice identified significant reductions in key ciliogenesis-related genes including Ccp110, Cep97, Cep290 and Kif3a. Confocal microscopy reveals abnormal, stunted cilia on proximal tubules, podocytes, and collecting duct cells in mice with global or conditional deletion of GR. In contrast, activation of GR signalling with dexamethasone in human kidney organoids or mouse IMCD3 cells increases cilia length, an effect blocked by the GR antagonist RU486. Analysis of GR-null kidney extracts demonstrates reduced levels of pERK and SUFU identifying potential cell pathway crosstalk with GR signalling that coordinately regulate ciliogenesis in the renal tubule. Finally, dexamethasone reduces Aurora kinase A levels, a factor driving cilia disassembly and implicated in the pathogenesis of polycystic kidney disease.
Collapse
Affiliation(s)
- Kelly L Short
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Jianshen Lao
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Rachel Lam
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Julie L M Moreau
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Judy Ng
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Mehran Piran
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Alexander N Combes
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Denny L Cottle
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Timothy J Cole
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
2
|
Itabashi T, Hosoba K, Morita T, Kimura S, Yamaoka K, Hirosawa M, Kobayashi D, Kishi H, Kume K, Itoh H, Kawakami H, Hashimoto K, Yamamoto T, Miyamoto T. Cholesterol ensures ciliary polycystin-2 localization to prevent polycystic kidney disease. Life Sci Alliance 2025; 8:e202403063. [PMID: 39900437 PMCID: PMC11791027 DOI: 10.26508/lsa.202403063] [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: 09/26/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/05/2025] Open
Abstract
The plasma membrane covering the primary cilium has a diverse accumulation of receptors and channels. To ensure the sensor function of the cilia, the ciliary membrane has higher cholesterol content than other cell membrane regions. A peroxisomal biogenesis disorder, Zellweger syndrome, characterized by polycystic kidney, is associated with a reduced level of ciliary cholesterol in cells. However, the etiological mechanism by which ciliary cholesterol lowering causes polycystic kidney disease remains unclear. Here, we demonstrated that lowering ciliary cholesterol by either pharmacological treatment or genetic depletion of peroxisomes impairs the localization of a ciliary ion channel polycystin-2. We also generated cultured renal medullary cells and mice carrying a missense variant in the cholesterol-binding site of polycystin-2 detected in the patient database of autosomal dominant polycystic kidney disease. This missense protein showed normal channel activity but decreased localization to the ciliary membrane. The homozygous mice exhibited embryonic lethality and the ciliopathy spectrum conditions of situs inversus and polycystic kidney. Our results suggest that cholesterol controls the ciliary localization of polycystin-2 to prevent polycystic kidney disease.
Collapse
Affiliation(s)
- Takeshi Itabashi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
- Division of Advanced Genome Editing Therapy, Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan
| | - Kosuke Hosoba
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
- Program of Mathematical and Life Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Tomoka Morita
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
- Division of Advanced Genome Editing Therapy, Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan
| | - Sotai Kimura
- Department of Molecular Pathology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
- Department of Anatomic Pathology, Hirosaki University Hospital, Aomori, Japan
| | - Kenji Yamaoka
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Moe Hirosawa
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
- Division of Advanced Genome Editing Therapy, Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan
| | - Daigo Kobayashi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Hiroko Kishi
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
- Department of Environmental Physiology, Faculty of Medicine, Shimane University, Shimane, Japan
| | - Kodai Kume
- Department of Molecular Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Itoh
- Department of Molecular Pathology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Hideshi Kawakami
- Department of Molecular Epidemiology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Kouichi Hashimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takashi Yamamoto
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
- Program of Mathematical and Life Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Tatsuo Miyamoto
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
- Division of Advanced Genome Editing Therapy, Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan
| |
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
|
Palicharla VR, Badgandi HB, Hwang SH, Legué E, Liem KF, Mukhopadhyay S. A defined tubby domain β-barrel surface region of TULP3 mediates ciliary trafficking of diverse cargoes. Mol Biol Cell 2025; 36:ar1. [PMID: 39565681 PMCID: PMC11742108 DOI: 10.1091/mbc.e24-09-0426] [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: 09/27/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024] Open
Abstract
The primary cilium is a paradigmatic subcellular compartment at the nexus of numerous cellular and morphogenetic pathways. The tubby family protein TULP3 acts as an adapter of the intraflagellar transport complex A in transporting integral membrane and membrane-associated lipidated proteins into cilia. However, the mechanisms by which TULP3 coordinates ciliary transport of diverse cargoes is not well understood. Here, we provide molecular insights into TULP3-mediated ciliary cargo recognition. We screened for critical TULP3 residues by proximity biotinylation-mass spectrometry, structural analysis, and testing TULP3 variants in human patients with hepatorenal fibrocystic disease and spina bifida. The TULP3 residues we identified 1) were located on one side of the β-barrel of the tubby domain away from the phosphoinositide binding site, 2) mediated ciliary trafficking of lipidated and transmembrane cargoes, and 3) determined proximity with these cargoes in vivo without affecting ciliary localization, phosphoinositide binding or hydrodynamic properties of TULP3. Overall, these findings implicate a specific region of one of the surfaces of the TULP3 β-barrel in ciliary trafficking of diverse cargoes. This region overlooks the β-strands 8-12 of the β-barrel and is away from the membrane anchoring phosphoinositide binding site. Targeting the TULP3-cargo interactions could provide therapeutics in ciliary trafficking diseases.
Collapse
Affiliation(s)
- Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Hemant B. Badgandi
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Sun-Hee Hwang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Emilie Legué
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Karel F. Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
5
|
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
|
6
|
Zhang C, Rehman M, Tian X, Pei SLC, Gu J, Bell TA, Dong K, Tham MS, Cai Y, Wei Z, Behrens F, Jetten AM, Zhao H, Lek M, Somlo S. Glis2 is an early effector of polycystin signaling and a target for therapy in polycystic kidney disease. Nat Commun 2024; 15:3698. [PMID: 38693102 PMCID: PMC11063051 DOI: 10.1038/s41467-024-48025-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 04/15/2024] [Indexed: 05/03/2024] Open
Abstract
Mouse models of autosomal dominant polycystic kidney disease (ADPKD) show that intact primary cilia are required for cyst growth following the inactivation of polycystin-1. The signaling pathways underlying this process, termed cilia-dependent cyst activation (CDCA), remain unknown. Using translating ribosome affinity purification RNASeq on mouse kidneys with polycystin-1 and cilia inactivation before cyst formation, we identify the differential 'CDCA pattern' translatome specifically dysregulated in kidney tubule cells destined to form cysts. From this, Glis2 emerges as a candidate functional effector of polycystin signaling and CDCA. In vitro changes in Glis2 expression mirror the polycystin- and cilia-dependent changes observed in kidney tissue, validating Glis2 as a cell culture-based indicator of polycystin function related to cyst formation. Inactivation of Glis2 suppresses polycystic kidney disease in mouse models of ADPKD, and pharmacological targeting of Glis2 with antisense oligonucleotides slows disease progression. Glis2 transcript and protein is a functional target of CDCA and a potential therapeutic target for treating ADPKD.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Michael Rehman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Xin Tian
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Steven Lim Cho Pei
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jianlei Gu
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT, USA
| | | | - Ke Dong
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ming Shen Tham
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Yiqiang Cai
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Zemeng Wei
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Felix Behrens
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Anton M Jetten
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Computational Biology and Bioinformatics Program, Yale University, New Haven, CT, USA
| | - Monkol Lek
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Stefan Somlo
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
7
|
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
|
8
|
Tham MS, Cottle DL, Zylberberg AK, Short KM, Jones LK, Chan P, Conduit SE, Dyson JM, Mitchell CA, Smyth IM. Deletion of Aurora kinase A prevents the development of polycystic kidney disease in mice. Nat Commun 2024; 15:371. [PMID: 38191531 PMCID: PMC10774271 DOI: 10.1038/s41467-023-44410-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 12/09/2023] [Indexed: 01/10/2024] Open
Abstract
Aurora Kinase A (AURKA) promotes cell proliferation and is overexpressed in different types of polycystic kidney disease (PKD). To understand AURKA's role in regulating renal cyst development we conditionally deleted the gene in mouse models of Autosomal Dominant PKD (ADPKD) and Joubert Syndrome, caused by Polycystin 1 (Pkd1) and Inositol polyphosphate-5-phosphatase E (Inpp5e) mutations respectively. We show that while Aurka is dispensable for collecting duct development and homeostasis, its deletion prevents cyst formation in both disease models. Cross-comparison of transcriptional changes implicated AKT signaling in cyst prevention and we show that (i) AURKA and AKT physically interact, (ii) AURKA regulates AKT activity in a kinase-independent manner and (iii) inhibition of AKT can reduce disease severity. AKT activation also regulates Aurka expression, creating a feed-forward loop driving renal cystogenesis. We find that the AURKA kinase inhibitor Alisertib stabilises the AURKA protein, agonizing its cystogenic functions. These studies identify AURKA as a master regulator of renal cyst development in different types of PKD, functioning in-part via AKT.
Collapse
Affiliation(s)
- Ming Shen Tham
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Denny L Cottle
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
| | - Allara K Zylberberg
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Kieran M Short
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Lynelle K Jones
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Perkin Chan
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Sarah E Conduit
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jennifer M Dyson
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Christina A Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Ian M Smyth
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
9
|
Van Sciver RE, Long AB, Katz HG, Gigante ED, Caspary T. Ciliary ARL13B inhibits developmental kidney cystogenesis in mouse. Dev Biol 2023; 500:1-9. [PMID: 37209936 PMCID: PMC10330881 DOI: 10.1016/j.ydbio.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
ARL13B is a small GTPase enriched in cilia. Deletion of Arl13b in mouse kidney results in renal cysts and an associated absence of primary cilia. Similarly, ablation of cilia leads to kidney cysts. To investigate whether ARL13B functions from within cilia to direct kidney development, we examined kidneys of mice expressing an engineered cilia-excluded ARL13B variant, ARL13BV358A. These mice retained renal cilia and developed cystic kidneys. Because ARL13B functions as a guanine nucleotide exchange factor (GEF) for ARL3, we examined kidneys of mice expressing an ARL13B variant that lacks ARL3 GEF activity, ARL13BR79Q. We found normal kidney development with no evidence of cysts in these mice. Taken together, our results show that ARL13B functions within cilia to inhibit renal cystogenesis during mouse development, and that this function does not depend on its role as a GEF for ARL3.
Collapse
Affiliation(s)
- Robert E Van Sciver
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA, 30322, USA.
| | - Alyssa B Long
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA, 30322, USA.
| | - Harrison G Katz
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA, 30322, USA; Department of Biology, Brown University, Providence, RI, 02912, USA.
| | - Eduardo D Gigante
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA, 30322, USA; Graduate Program in Neuroscience, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA, 30322, USA; Department of Biology, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA, 30322, USA.
| |
Collapse
|
10
|
Van Sciver RE, Long AB, Katz HG, Gigante ED, Caspary T. Ciliary ARL13B inhibits developmental kidney cystogenesis in mouse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527739. [PMID: 36798281 PMCID: PMC9934666 DOI: 10.1101/2023.02.08.527739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
ARL13B is a small GTPase enriched in cilia. Deletion of Arl13b in mouse kidney results in renal cysts and an associated absence of primary cilia. Similarly, ablation of cilia leads to kidney cysts. To investigate whether ARL13B functions from within cilia to direct kidney development, we examined kidneys of mice expressing an engineered cilia-excluded ARL13B variant, ARL13BV358A. These mice retained renal cilia and developed cystic kidneys. Because ARL13B functions as a guanine nucleotide exchange factor (GEF) for ARL3, we examined kidneys of mice expressing an ARL13B variant that lacks ARL3 GEF activity, ARL13BR79Q. We found normal kidney development with no evidence of cysts in these mice. Taken together, our results show that ARL13B functions within cilia to inhibit renal cystogenesis during mouse development, and that this function does not depend on its role as a GEF for ARL3.
Collapse
Affiliation(s)
- Robert E. Van Sciver
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA 30322, USA
| | - Alyssa B. Long
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA 30322, USA
| | - Harrison G. Katz
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA 30322, USA
- Present address: Department of Biology, Brown University, Providence, RI 02912, USA
| | - Eduardo D. Gigante
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA 30322, USA
- Graduate Program in Neuroscience, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA 30322, USA
- Present address: Department of Biology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street, Suite 301, Atlanta, GA 30322, USA
| |
Collapse
|
11
|
Jiang M, Palicharla VR, Miller D, Hwang SH, Zhu H, Hixson P, Mukhopadhyay S, Sun J. Human IFT-A complex structures provide molecular insights into ciliary transport. Cell Res 2023; 33:288-298. [PMID: 36775821 PMCID: PMC10066299 DOI: 10.1038/s41422-023-00778-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/11/2023] [Indexed: 02/14/2023] Open
Abstract
Intraflagellar transport (IFT) complexes, IFT-A and IFT-B, form bidirectional trains that move along the axonemal microtubules and are essential for assembling and maintaining cilia. Mutations in IFT subunits lead to numerous ciliopathies involving multiple tissues. However, how IFT complexes assemble and mediate cargo transport lacks mechanistic understanding due to missing high-resolution structural information of the holo-complexes. Here we report cryo-EM structures of human IFT-A complexes in the presence and absence of TULP3 at overall resolutions of 3.0-3.9 Å. IFT-A adopts a "lariat" shape with interconnected core and peripheral subunits linked by structurally vital zinc-binding domains. TULP3, the cargo adapter, interacts with IFT-A through its N-terminal region, and interface mutations disrupt cargo transport. We also determine the molecular impacts of disease mutations on complex formation and ciliary transport. Our work reveals IFT-A architecture, sheds light on ciliary transport and IFT train formation, and enables the rationalization of disease mutations in ciliopathies.
Collapse
Affiliation(s)
- Meiqin Jiang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Darcie Miller
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sun-Hee Hwang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hanwen Zhu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Patricia Hixson
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Ji Sun
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
12
|
Garaycoechea JI, Quinlan C, Luijsterburg MS. Pathological consequences of DNA damage in the kidney. Nat Rev Nephrol 2023; 19:229-243. [PMID: 36702905 DOI: 10.1038/s41581-022-00671-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 01/27/2023]
Abstract
DNA lesions that evade repair can lead to mutations that drive the development of cancer, and cellular responses to DNA damage can trigger senescence and cell death, which are associated with ageing. In the kidney, DNA damage has been implicated in both acute and chronic kidney injury, and in renal cell carcinoma. The susceptibility of the kidney to chemotherapeutic agents that damage DNA is well established, but an unexpected link between kidney ciliopathies and the DNA damage response has also been reported. In addition, human genetic deficiencies in DNA repair have highlighted DNA crosslinks, DNA breaks and transcription-blocking damage as lesions that are particularly toxic to the kidney. Genetic tools in mice, as well as advances in kidney organoid and single-cell RNA sequencing technologies, have provided important insights into how specific kidney cell types respond to DNA damage. The emerging view is that in the kidney, DNA damage affects the local microenvironment by triggering a damage response and cell proliferation to replenish injured cells, as well as inducing systemic responses aimed at reducing exposure to genotoxic stress. The pathological consequences of DNA damage are therefore key to the nephrotoxicity of DNA-damaging agents and the kidney phenotypes observed in human DNA repair-deficiency disorders.
Collapse
Affiliation(s)
- Juan I Garaycoechea
- Hubrecht Institute-KNAW, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Catherine Quinlan
- Department of Paediatrics, University of Melbourne, Parkville, Australia
- Department of Nephrology, Royal Children's Hospital, Melbourne, Australia
- Department of Kidney Regeneration, Murdoch Children's Research Institute, Melbourne, Australia
| | - Martijn S Luijsterburg
- Department of Human Genetics, Leiden University Medical Center (LUMC), Leiden, The Netherlands.
| |
Collapse
|
13
|
Mahajan D, Madugula V, Lu L. Rab8 and TNPO1 are ciliary transport adaptors for GTPase Arl13b by interacting with its RVEP motif-containing ciliary targeting sequence. J Biol Chem 2023; 299:104604. [PMID: 36907439 PMCID: PMC10124946 DOI: 10.1016/j.jbc.2023.104604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Arl13b, an ARF/Arl-family GTPase, is highly enriched in the cilium. Recent studies have established Arl13b as one of the most crucial regulators for ciliary organization, trafficking, and signaling. The ciliary localization of Arl13b is known to require the RVEP motif. However, its cognitive ciliary transport adaptor has been elusive. Here, by imaging the ciliary localization of truncation and point mutations, we defined the ciliary targeting sequence (CTS) of Arl13b as a C-terminal stretch of 17 amino acids containing the RVEP motif. We found Rab8-GDP, but not Rab8-GTP, and TNPO1 simultaneously and directly bind to the CTS of Arl13b in pull-down assays using cell lysates or purified recombinant proteins. Furthermore, Rab8-GDP substantially enhances the interaction between TNPO1 and CTS. Additionally, we determined that the RVEP motif is an essential element as its mutation abolishes the interaction of the CTS with Rab8-GDP and TNPO1 in pull-down and TurboID-based proximity ligation assays. Finally, knockdown of endogenous Rab8 or TNPO1 decreases the ciliary localization of endogenous Arl13b. Therefore, our results suggest Rab8 and TNPO1 might function together as a ciliary transport adaptor for Arl13b by interacting with its RVEP-containing CTS.
Collapse
Affiliation(s)
- Divyanshu Mahajan
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Viswanadh Madugula
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Lei Lu
- School of Biological Sciences, Nanyang Technological University, Singapore.
| |
Collapse
|
14
|
Palicharla VR, Hwang SH, Somatilaka BN, Legué E, Shimada IS, Familiari NE, Tran VM, Woodruff JB, Liem KF, Mukhopadhyay S. Interactions between TULP3 tubby domain and ARL13B amphipathic helix promote lipidated protein transport to cilia. Mol Biol Cell 2023; 34:ar18. [PMID: 36652335 PMCID: PMC10011728 DOI: 10.1091/mbc.e22-10-0473] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The primary cilium is a nexus for cell signaling and relies on specific protein trafficking for function. The tubby family protein TULP3 transports integral membrane proteins into cilia through interactions with the intraflagellar transport complex-A (IFT-A) and phosphoinositides. It was previously shown that short motifs called ciliary localization sequences (CLSs) are necessary and sufficient for TULP3-dependent ciliary trafficking of transmembrane cargoes. However, the mechanisms by which TULP3 regulates ciliary compartmentalization of nonintegral, membrane-associated proteins and whether such trafficking requires TULP3-dependent CLSs is unknown. Here we show that TULP3 is required for ciliary transport of the Joubert syndrome-linked palmitoylated GTPase ARL13B through a CLS. An N-terminal amphipathic helix, preceding the GTPase domain of ARL13B, couples with the TULP3 tubby domain for ciliary trafficking, irrespective of palmitoylation. ARL13B transport requires TULP3 binding to IFT-A but not to phosphoinositides, indicating strong membrane-proximate interactions, unlike transmembrane cargo transport requiring both properties of TULP3. TULP3-mediated trafficking of ARL13B also regulates ciliary enrichment of farnesylated and myristoylated downstream effectors of ARL13B. The lipidated cargoes show distinctive depletion kinetics from kidney epithelial cilia with relation to Tulp3 deletion-induced renal cystogenesis. Overall, these findings indicate an expanded role of the tubby domain in capturing analogous helical secondary structural motifs from diverse cargoes.
Collapse
Affiliation(s)
- Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Sun-Hee Hwang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | - Emilie Legué
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Issei S Shimada
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Nicole E Familiari
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Vanna M Tran
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jeffrey B Woodruff
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Karel F Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
15
|
Liu P, Liu Y, Zhou J. Ciliary mechanosensation - roles of polycystins and mastigonemes. J Cell Sci 2023; 136:286945. [PMID: 36752106 DOI: 10.1242/jcs.260565] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Cilia are surface-exposed organelles that provide motility and sensory functions for cells, and it is widely believed that mechanosensation can be mediated through cilia. Polycystin-1 and -2 (PC-1 and PC-2, respectively) are transmembrane proteins that can localize to cilia; however, the molecular mechanisms by which polycystins contribute to mechanosensation are still controversial. Studies detail two prevailing models for the molecular roles of polycystins on cilia; one stresses the mechanosensation capabilities and the other unveils their ligand-receptor nature. The discovery that polycystins interact with mastigonemes, the 'hair-like' protrusions of flagella, is a novel finding in identifying the interactors of polycystins in cilia. While the functions of polycystins proposed by both models may coexist in cilia, it is hoped that a precise understanding of the mechanism of action of polycystins can be achieved by uncovering their distribution and interacting factors inside cilia. This will hopefully provide a satisfying answer to the pathogenesis of autosomal dominant polycystic kidney disease (ADPKD), which is caused by mutations in PC-1 and PC-2. In this Review, we discuss the characteristics of polycystins in the context of cilia and summarize the functions of mastigonemes in unicellular ciliates. Finally, we compare flagella and molecular features of PC-2 between unicellular and multicellular organisms, with the aim of providing new insights into the ciliary roles of polycystins in general.
Collapse
Affiliation(s)
- Peiwei Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology , College of Life Sciences in Shandong Normal University, Jinan 250358, China
| | - Ying Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology , College of Life Sciences in Shandong Normal University, Jinan 250358, China
| | - Jun Zhou
- Shandong Provincial Key Laboratory of Animal Resistance Biology , College of Life Sciences in Shandong Normal University, Jinan 250358, China.,College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
16
|
Abstract
The assembly and maintenance of most cilia and eukaryotic flagella depends on intraflagellar transport (IFT), the bidirectional movement of multi-megadalton IFT trains along the axonemal microtubules. These IFT trains function as carriers, moving ciliary proteins between the cell body and the organelle. Whereas tubulin, the principal protein of cilia, binds directly to IFT particle proteins, the transport of other ciliary proteins and complexes requires adapters that link them to the trains. Large axonemal substructures, such as radial spokes, outer dynein arms and inner dynein arms, assemble in the cell body before attaching to IFT trains, using the adapters ARMC2, ODA16 and IDA3, respectively. Ciliary import of several membrane proteins involves the putative adapter tubby-like protein 3 (TULP3), whereas membrane protein export involves the BBSome, an octameric complex that co-migrates with IFT particles. Thus, cells employ a variety of adapters, each of which is substoichiometric to the core IFT machinery, to expand the cargo range of the IFT trains. This Review summarizes the individual and shared features of the known cargo adapters and discusses their possible role in regulating the transport capacity of the IFT pathway.
Collapse
Affiliation(s)
- Karl Lechtreck
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
17
|
Jafari Khamirani H, Palicharla VR, Dastgheib SA, Dianatpour M, Imanieh MH, Tabei SS, Besse W, Mukhopadhyay S, Liem KF. A pathogenic variant of TULP3 causes renal and hepatic fibrocystic disease. Front Genet 2022; 13:1021037. [PMID: 36276950 PMCID: PMC9585244 DOI: 10.3389/fgene.2022.1021037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/22/2022] [Indexed: 11/26/2022] Open
Abstract
Patient variants in Tubby Like Protein-3 (TULP3) have recently been associated with progressive fibrocystic disease in tissues and organs. TULP3 is a ciliary trafficking protein that links membrane-associated proteins to the intraflagellar transport complex A. In mice, mutations in Tulp3 drive phenotypes consistent with ciliary dysfunction which include renal cystic disease, as part of a ciliopathic spectrum. Here we report two sisters from consanguineous parents with fibrocystic renal and hepatic disease harboring a homozygous missense mutation in TULP3 (NM_003324.5: c.1144C>T, p.Arg382Trp). The R382W patient mutation resides within the C-terminal Tubby domain, a conserved domain required for TULP3 to associate with phosphoinositides. We show that inner medullary collecting duct-3 cells expressing the TULP3 R382W patient variant have a severely reduced ability to localize the membrane-associated proteins ARL13b, INPP5E, and GPR161 to the cilium, consistent with a loss of TULP3 function. These studies establish Arginine 382 as a critical residue in the Tubby domain, which is essential for TULP3-mediated protein trafficking within the cilium, and expand the phenotypic spectrum known to result from recessive deleterious mutations in TULP3.
Collapse
Affiliation(s)
| | - Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | | | - Mehdi Dianatpour
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hadi Imanieh
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Sajjad Tabei
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Whitney Besse
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT, United States
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Karel F. Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
18
|
Cilleros-Rodriguez D, Martin-Morales R, Barbeito P, Deb Roy A, Loukil A, Sierra-Rodero B, Herranz G, Pampliega O, Redrejo-Rodriguez M, Goetz SC, Izquierdo M, Inoue T, Garcia-Gonzalo FR. Multiple ciliary localization signals control INPP5E ciliary targeting. eLife 2022; 11:e78383. [PMID: 36063381 PMCID: PMC9444247 DOI: 10.7554/elife.78383] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/21/2022] [Indexed: 12/04/2022] Open
Abstract
Primary cilia are sensory membrane protrusions whose dysfunction causes ciliopathies. INPP5E is a ciliary phosphoinositide phosphatase mutated in ciliopathies like Joubert syndrome. INPP5E regulates numerous ciliary functions, but how it accumulates in cilia remains poorly understood. Herein, we show INPP5E ciliary targeting requires its folded catalytic domain and is controlled by four conserved ciliary localization signals (CLSs): LLxPIR motif (CLS1), W383 (CLS2), FDRxLYL motif (CLS3) and CaaX box (CLS4). We answer two long-standing questions in the field. First, partial CLS1-CLS4 redundancy explains why CLS4 is dispensable for ciliary targeting. Second, the essential need for CLS2 clarifies why CLS3-CLS4 are together insufficient for ciliary accumulation. Furthermore, we reveal that some Joubert syndrome mutations perturb INPP5E ciliary targeting, and clarify how each CLS works: (i) CLS4 recruits PDE6D, RPGR and ARL13B, (ii) CLS2-CLS3 regulate association to TULP3, ARL13B, and CEP164, and (iii) CLS1 and CLS4 cooperate in ATG16L1 binding. Altogether, we shed light on the mechanisms of INPP5E ciliary targeting, revealing a complexity without known parallels among ciliary cargoes.
Collapse
Affiliation(s)
- Dario Cilleros-Rodriguez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridSpain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), Consejo Superior de Investigaciones Científicas (CSIC)-UAMMadridSpain
- Instituto de Investigación del Hospital Universitario de La Paz (IdiPAZ)MadridSpain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII)MadridSpain
| | - Raquel Martin-Morales
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridSpain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), Consejo Superior de Investigaciones Científicas (CSIC)-UAMMadridSpain
- Instituto de Investigación del Hospital Universitario de La Paz (IdiPAZ)MadridSpain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII)MadridSpain
| | - Pablo Barbeito
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridSpain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), Consejo Superior de Investigaciones Científicas (CSIC)-UAMMadridSpain
- Instituto de Investigación del Hospital Universitario de La Paz (IdiPAZ)MadridSpain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII)MadridSpain
| | - Abhijit Deb Roy
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Abdelhalim Loukil
- Department of Pharmacology and Cancer Biology, Duke University School of MedicineDurhamUnited States
| | - Belen Sierra-Rodero
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridSpain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), Consejo Superior de Investigaciones Científicas (CSIC)-UAMMadridSpain
- Instituto de Investigación del Hospital Universitario de La Paz (IdiPAZ)MadridSpain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII)MadridSpain
| | - Gonzalo Herranz
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridSpain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), Consejo Superior de Investigaciones Científicas (CSIC)-UAMMadridSpain
| | - Olatz Pampliega
- Department of Neurosciences, University of the Basque Country, Achucarro Basque Center for Neuroscience-UPV/EHULeioaSpain
| | - Modesto Redrejo-Rodriguez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridSpain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), Consejo Superior de Investigaciones Científicas (CSIC)-UAMMadridSpain
| | - Sarah C Goetz
- Department of Pharmacology and Cancer Biology, Duke University School of MedicineDurhamUnited States
| | - Manuel Izquierdo
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridSpain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), Consejo Superior de Investigaciones Científicas (CSIC)-UAMMadridSpain
| | - Takanari Inoue
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Francesc R Garcia-Gonzalo
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridSpain
- Instituto de Investigaciones Biomédicas “Alberto Sols” (IIBM), Consejo Superior de Investigaciones Científicas (CSIC)-UAMMadridSpain
- Instituto de Investigación del Hospital Universitario de La Paz (IdiPAZ)MadridSpain
- CIBER de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII)MadridSpain
| |
Collapse
|
19
|
Wang W, Silva LM, Wang HH, Kavanaugh MA, Pottorf TS, Allard BA, Jacobs DT, Dong R, Cornelius JT, Chaturvedi A, Swenson-Fields KI, Fields TA, Pritchard MT, Sharma M, Slawson C, Wallace DP, Calvet JP, Tran PV. Ttc21b deficiency attenuates autosomal dominant polycystic kidney disease in a kidney tubular- and maturation-dependent manner. Kidney Int 2022; 102:577-591. [PMID: 35644283 PMCID: PMC9398994 DOI: 10.1016/j.kint.2022.04.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 01/26/2023]
Abstract
Primary cilia are sensory organelles built and maintained by intraflagellar transport (IFT) multiprotein complexes. Deletion of several IFT-B genes attenuates polycystic kidney disease (PKD) severity in juvenile and adult autosomal dominant polycystic kidney disease (ADPKD) mouse models. However, deletion of an IFT-A adaptor, Tulp3, attenuates PKD severity in adult mice only. These studies indicate that dysfunction of specific cilia components has potential therapeutic value. To broaden our understanding of cilia dysfunction and its therapeutic potential, we investigate the role of global deletion of an IFT-A gene, Ttc21b, in juvenile and adult mouse models of ADPKD. Both juvenile (postnatal day 21) and adult (six months of age) ADPKD mice exhibited kidney cysts, increased kidney weight/body weight ratios, lengthened kidney cilia, inflammation, and increased levels of the nutrient sensor, O-linked β-N-acetylglucosamine (O-GlcNAc). Deletion of Ttc21b in juvenile ADPKD mice reduced cortical collecting duct cystogenesis and kidney weight/body weight ratios, increased proximal tubular and glomerular dilations, but did not reduce cilia length, inflammation, nor O-GlcNAc levels. In contrast, Ttc21b deletion in adult ADPKD mice markedly attenuated kidney cystogenesis and reduced cilia length, inflammation, and O-GlcNAc levels. Thus, unlike IFT-B, the effect of Ttc21b deletion in mouse models of ADPKD is development-specific. Unlike an IFT-A adaptor, deleting Ttc21b in juvenile ADPKD mice is partially ameliorative. Thus, our studies suggest that different microenvironmental factors, found in distinct nephron segments and in developing versus mature stages, modify ciliary homeostasis and ADPKD pathobiology. Further, elevated levels of O-GlcNAc, which regulates cellular metabolism and ciliogenesis, may be a pathological feature of ADPKD.
Collapse
Affiliation(s)
- Wei Wang
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Luciane M Silva
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Henry H Wang
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Matthew A Kavanaugh
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Tana S Pottorf
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Bailey A Allard
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Damon T Jacobs
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Rouchen Dong
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Joseph T Cornelius
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Aakriti Chaturvedi
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Katherine I Swenson-Fields
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Timothy A Fields
- Department of Pathology and Laboratory Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Michele T Pritchard
- Pharmacology, Toxicology and Therapeutics, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Madhulika Sharma
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Chad Slawson
- Department of Biochemistry and Molecular Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Darren P Wallace
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - James P Calvet
- Department of Biochemistry and Molecular Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Pamela V Tran
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA.
| |
Collapse
|
20
|
Walker RV, Maranto A, Palicharla VR, Hwang SH, Mukhopadhyay S, Qian F. Cilia-Localized Counterregulatory Signals as Drivers of Renal Cystogenesis. Front Mol Biosci 2022; 9:936070. [PMID: 35832738 PMCID: PMC9272769 DOI: 10.3389/fmolb.2022.936070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/30/2022] [Indexed: 12/18/2022] Open
Abstract
Primary cilia play counterregulatory roles in cystogenesis-they inhibit cyst formation in the normal renal tubule but promote cyst growth when the function of polycystins is impaired. Key upstream cilia-specific signals and components involved in driving cystogenesis have remained elusive. Recent studies of the tubby family protein, Tubby-like protein 3 (TULP3), have provided new insights into the cilia-localized mechanisms that determine cyst growth. TULP3 is a key adapter of the intraflagellar transport complex A (IFT-A) in the trafficking of multiple proteins specifically into the ciliary membrane. Loss of TULP3 results in the selective exclusion of its cargoes from cilia without affecting their extraciliary pools and without disrupting cilia or IFT-A complex integrity. Epistasis analyses have indicated that TULP3 inhibits cystogenesis independently of the polycystins during kidney development but promotes cystogenesis in adults when polycystins are lacking. In this review, we discuss the current model of the cilia-dependent cyst activation (CDCA) mechanism in autosomal dominant polycystic kidney disease (ADPKD) and consider the possible roles of ciliary and extraciliary polycystins in regulating CDCA. We then describe the limitations of this model in not fully accounting for how cilia single knockouts cause significant cystic changes either in the presence or absence of polycystins. Based on available data from TULP3/IFT-A-mediated differential regulation of cystogenesis in kidneys with deletion of polycystins either during development or in adulthood, we hypothesize the existence of cilia-localized components of CDCA (cCDCA) and cilia-localized cyst inhibition (CLCI) signals. We develop the criteria for cCDCA/CLCI signals and discuss potential TULP3 cargoes as possible cilia-localized components that determine cystogenesis in kidneys during development and in adult mice.
Collapse
Affiliation(s)
- Rebecca V. Walker
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Anthony Maranto
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | | | - Sun-Hee Hwang
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Saikat Mukhopadhyay
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Feng Qian
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
21
|
Dutta P, Ray K. Ciliary membrane, localised lipid modification and cilia function. J Cell Physiol 2022; 237:2613-2631. [PMID: 35661356 DOI: 10.1002/jcp.30787] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 11/08/2022]
Abstract
Cilium, a tiny microtubule-based cellular appendage critical for cell signalling and physiology, displays a large variety of receptors. The composition and turnover of ciliary lipids and receptors determine cell behaviour. Due to the exclusion of ribosomal machinery and limited membrane area, a cilium needs adaptive logistics to actively reconstitute the lipid and receptor compositions during development and differentiation. How is this dynamicity generated? Here, we examine whether, along with the Intraflagellar-Transport, targeted changes in sector-wise lipid composition could control the receptor localisation and functions in the cilia. We discuss how an interplay between ciliary lipid composition, localised lipid modification, and receptor function could contribute to cilia growth and signalling. We argue that lipid modification at the cell-cilium interface could generate an added thrust for a selective exchange of membrane lipids and the transmembrane and membrane-associated proteins.
Collapse
Affiliation(s)
- Priya Dutta
- 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
|
22
|
Oyama Y, Miyata H, Shimada K, Larasati T, Fujihara Y, Ikawa M. TULP2 deletion mice exhibit abnormal outer dense fiber structure and male infertility. Reprod Med Biol 2022; 21:e12467. [PMID: 35619658 PMCID: PMC9126596 DOI: 10.1002/rmb2.12467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/12/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022] Open
Abstract
Purpose Tulp2 (tubby-like protein 2) is a member of the tubby protein family and expressed predominantly in mouse testis. Recently, it was reported that Tulp2 knockout (KO) mice exhibited disrupted sperm tail morphology; however, it remains to be determined how TULP2 deletion causes abnormal tail formation. Methods The authors analyzed male fertility, sperm morphology, and motility of two Tulp2 KO mouse lines that were generated using the conventional method that utilizes homologous recombination in embryonic stem (ES) cells as well as the clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) system. Furthermore, the authors observed the spermatogenesis of Tulp2 KO mice in more detail using scanning and transmission electron microscopy (SEM and TEM). Results Both mouse lines of Tulp2 KO exhibited male infertility, abnormal tail morphology, and impaired sperm motility. No overt abnormalities were found in the formation of the mitochondrial sheath in Tulp2 KO mice using the freeze-fracture method with SEM. In contrast, abnormal outer dense fiber (ODF) structure was observed in Tulp2 KO testis with TEM. Conclusions TULP2 may play roles in the correct formation and/or maintenance of ODF, which may lead to abnormal tail morphology, impaired sperm motility, and male infertility.
Collapse
Affiliation(s)
- Yuki Oyama
- Graduate School of Pharmaceutical SciencesOsaka UniversitySuitaJapan,Department of Experimental Genome ResearchResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan
| | - Haruhiko Miyata
- Department of Experimental Genome ResearchResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan
| | - Keisuke Shimada
- Department of Experimental Genome ResearchResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan
| | - Tamara Larasati
- Department of Experimental Genome ResearchResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan,Graduate School of MedicineOsaka UniversitySuitaJapan
| | - Yoshitaka Fujihara
- Department of Experimental Genome ResearchResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan,Department of Bioscience and GeneticsNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Masahito Ikawa
- Graduate School of Pharmaceutical SciencesOsaka UniversitySuitaJapan,Department of Experimental Genome ResearchResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan,The Institute of Medical ScienceThe University of TokyoTokyoJapan,Center for Infectious Disease Education and ResearchOsaka UniversityOsakaJapan
| |
Collapse
|
23
|
Devane J, Ott E, Olinger EG, Epting D, Decker E, Friedrich A, Bachmann N, Renschler G, Eisenberger T, Briem-Richter A, Grabhorn EF, Powell L, Wilson IJ, Rice SJ, Miles CG, Wood K, Trivedi P, Hirschfield G, Pietrobattista A, Wohler E, Mezina A, Sobreira N, Agolini E, Maggiore G, Dahmer-Heath M, Yilmaz A, Boerries M, Metzger P, Schell C, Grünewald I, Konrad M, König J, Schlevogt B, Sayer JA, Bergmann C. Progressive liver, kidney, and heart degeneration in children and adults affected by TULP3 mutations. Am J Hum Genet 2022; 109:928-943. [PMID: 35397207 PMCID: PMC9118107 DOI: 10.1016/j.ajhg.2022.03.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/22/2022] [Indexed: 12/31/2022] Open
Abstract
Organ fibrosis is a shared endpoint of many diseases, yet underlying mechanisms are not well understood. Several pathways governed by the primary cilium, a sensory antenna present on most vertebrate cells, have been linked with fibrosis. Ciliopathies usually start early in life and represent a considerable disease burden. We performed massively parallel sequencing by using cohorts of genetically unsolved individuals with unexplained liver and kidney failure and correlated this with clinical, imaging, and histopathological analyses. Mechanistic studies were conducted with a vertebrate model and primary cells. We detected bi-allelic deleterious variants in TULP3, encoding a critical adaptor protein for ciliary trafficking, in a total of 15 mostly adult individuals, originating from eight unrelated families, with progressive degenerative liver fibrosis, fibrocystic kidney disease, and hypertrophic cardiomyopathy with atypical fibrotic patterns on histopathology. We recapitulated the human phenotype in adult zebrafish and confirmed disruption of critical ciliary cargo composition in several primary cell lines derived from affected individuals. Further, we show interaction between TULP3 and the nuclear deacetylase SIRT1, with roles in DNA damage repair and fibrosis, and report increased DNA damage ex vivo. Transcriptomic studies demonstrated upregulation of profibrotic pathways with gene clusters for hypertrophic cardiomyopathy and WNT and TGF-β signaling. These findings identify variants in TULP3 as a monogenic cause for progressive degenerative disease of major organs in which affected individuals benefit from early detection and improved clinical management. Elucidation of mechanisms crucial for DNA damage repair and tissue maintenance will guide novel therapeutic avenues for this and similar genetic and non-genomic diseases.
Collapse
Affiliation(s)
- John Devane
- Department of Medicine IV, Faculty of Medicine, Medical Center-University of Freiburg, 79106 Freiburg, Germany
| | - Elisabeth Ott
- Department of Medicine IV, Faculty of Medicine, Medical Center-University of Freiburg, 79106 Freiburg, Germany
| | - Eric G Olinger
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Daniel Epting
- Department of Medicine IV, Faculty of Medicine, Medical Center-University of Freiburg, 79106 Freiburg, Germany
| | - Eva Decker
- Medizinische Genetik Mainz, Limbach Genetics, 55128 Mainz, Germany
| | - Anja Friedrich
- Medizinische Genetik Mainz, Limbach Genetics, 55128 Mainz, Germany
| | - Nadine Bachmann
- Medizinische Genetik Mainz, Limbach Genetics, 55128 Mainz, Germany
| | - Gina Renschler
- Medizinische Genetik Mainz, Limbach Genetics, 55128 Mainz, Germany
| | | | - Andrea Briem-Richter
- University Medical Center Hamburg-Eppendorf, Department of Pediatrics, 20251 Hamburg, Germany
| | - Enke Freya Grabhorn
- University Medical Center Hamburg-Eppendorf, Department of Pediatrics, 20251 Hamburg, Germany
| | - Laura Powell
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Ian J Wilson
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Sarah J Rice
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Colin G Miles
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Katrina Wood
- Histopathology Department, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
| | - Palak Trivedi
- NIHR Birmingham BRC, Centre for Liver and Gastrointestinal Research, University of Birmingham, Birmingham B15 2TT, UK; Liver Unit, University Hospitals Birmingham, Birmingham B15 2GW, UK; Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK; Institute of Applied Health Research, University of Birmingham, Birmingham B15 2TT, UK
| | - Gideon Hirschfield
- Toronto Centre for Liver Disease, University Health Network, Toronto, ON M6H 3M1, Canada
| | - Andrea Pietrobattista
- Hepatogastroenterology and Liver Transplant Unit and Medical Genetics Laboratory, IRCCS Bambino Gesù Children's Hospital, 00165 Rome, Italy
| | - Elizabeth Wohler
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Anya Mezina
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Nara Sobreira
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emanuele Agolini
- Translational Cytogenomics Research Unit, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Giuseppe Maggiore
- Hepatogastroenterology and Liver Transplant Unit and Medical Genetics Laboratory, IRCCS Bambino Gesù Children's Hospital, 00165 Rome, Italy
| | - Mareike Dahmer-Heath
- Department of General Pediatrics, University Hospital Münster, 48149 Münster, Germany
| | - Ali Yilmaz
- Department of Cardiology I, University Hospital Münster, 48149 Münster, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine Medical Center - University of Freiburg, Medical Faculty, University of Freiburg, 79110 Freiburg, Germany; The German Cancer Consortium, Partner Site Freiburg and Cancer Research Center, 69120 Heidelberg, Germany
| | - Patrick Metzger
- Institute of Medical Bioinformatics and Systems Medicine Medical Center - University of Freiburg, Medical Faculty, University of Freiburg, 79110 Freiburg, Germany
| | - Christoph Schell
- Institute for Pathology, Medical Center - University of Freiburg, Medical Faculty, University of Freiburg, 79002 Freiburg, Germany
| | - Inga Grünewald
- Institute for Pathology, University Hospital Münster, 48149 Münster, Germany
| | - Martin Konrad
- Department of General Pediatrics, University Hospital Münster, 48149 Münster, Germany
| | - Jens König
- Department of General Pediatrics, University Hospital Münster, 48149 Münster, Germany
| | - Bernhard Schlevogt
- Department of Internal Medicine B, Gastroenterology, University Hospital Münster, 48149 Münster, Germany
| | - John A Sayer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK; Renal Services, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK; Newcastle Biomedical Research Centre, NIHR, Newcastle upon Tyne NE4 5PL, UK.
| | - Carsten Bergmann
- Department of Medicine IV, Faculty of Medicine, Medical Center-University of Freiburg, 79106 Freiburg, Germany; Medizinische Genetik Mainz, Limbach Genetics, 55128 Mainz, Germany.
| |
Collapse
|
24
|
Shimada IS, Kato Y. Ciliary signaling in stem cells in health and disease: Hedgehog pathway and beyond. Semin Cell Dev Biol 2022; 129:115-125. [PMID: 35466055 DOI: 10.1016/j.semcdb.2022.04.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
The primary cilium is a hair-like sensory compartment that protrudes from the cellular surface. The primary cilium is enriched in a variety of signaling molecules that regulate cellular activities. Stem cells have primary cilia. They reside in a specialized environment, called the stem cell niche. This niche contains a variety of secreted factors, and some of their receptors are localized in the primary cilia of stem cells. Here, we summarize the current understanding of the function of cilia in compartmentalized signaling in stem cells. We describe how ciliary signaling regulates stem cells and progenitor cells during development, tissue homeostasis and tumorigenesis. We summarize our understanding of cilia regulated signaling -primary involving the hedgehog pathway- in stem cells in diverse settings that include neuroepithelial cells, radial glia, cerebellar granule neuron precursors, hematopoietic stem cells, hair follicle stem cells, bone marrow mesenchymal stem cells and mammary gland stem cells. Overall, our review highlights a variety of roles that ciliary signaling plays in regulating stem cells throughout life.
Collapse
Affiliation(s)
- Issei S Shimada
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Azakawasumi, Mizuzho-cho, Mizuho-ku, Nagoya, 467-8601 Aichi, Japan.
| | - Yoichi Kato
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Azakawasumi, Mizuzho-cho, Mizuho-ku, Nagoya, 467-8601 Aichi, Japan.
| |
Collapse
|
25
|
Turn RE, Hu Y, Dewees SI, Devi N, East MP, Hardin KR, Khatib T, Linnert J, Wolfrum U, Lim MJ, Casanova JE, Caspary T, Kahn RA. The ARF GAPs ELMOD1 and ELMOD3 act at the Golgi and cilia to regulate ciliogenesis and ciliary protein traffic. Mol Biol Cell 2022; 33:ar13. [PMID: 34818063 PMCID: PMC9236152 DOI: 10.1091/mbc.e21-09-0443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 11/11/2022] Open
Abstract
ELMODs are a family of three mammalian paralogues that display GTPase-activating protein (GAP) activity toward a uniquely broad array of ADP-ribosylation factor (ARF) family GTPases that includes ARF-like (ARL) proteins. ELMODs are ubiquitously expressed in mammalian tissues, highly conserved across eukaryotes, and ancient in origin, being present in the last eukaryotic common ancestor. We described functions of ELMOD2 in immortalized mouse embryonic fibroblasts (MEFs) in the regulation of cell division, microtubules, ciliogenesis, and mitochondrial fusion. Here, using similar strategies with the paralogues ELMOD1 and ELMOD3, we identify novel functions and locations of these cell regulators and compare them to those of ELMOD2, allowing the determination of functional redundancy among the family members. We found strong similarities in phenotypes resulting from deletion of either Elmod1 or Elmod3 and marked differences from those arising in Elmod2 deletion lines. Deletion of either Elmod1 or Elmod3 results in the decreased ability of cells to form primary cilia, loss of a subset of proteins from cilia, and accumulation of some ciliary proteins at the Golgi, predicted to result from compromised traffic from the Golgi to cilia. These phenotypes are reversed upon activating mutant expression of either ARL3 or ARL16, linking their roles to ELMOD1/3 actions.
Collapse
Affiliation(s)
- Rachel E. Turn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
- Biochemistry, Cell & Developmental Biology Graduate Program, Emory University, Atlanta, GA 30322
- Department of Microbiology and Immunology, Stanford University, Palo Alto, CA 94305
| | - Yihan Hu
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Skylar I. Dewees
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
- Biochemistry, Cell & Developmental Biology Graduate Program, Emory University, Atlanta, GA 30322
| | - Narra Devi
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
| | - Michael P. East
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Katherine R. Hardin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
- Biochemistry, Cell & Developmental Biology Graduate Program, Emory University, Atlanta, GA 30322
| | - Tala Khatib
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322
- Biochemistry, Cell & Developmental Biology Graduate Program, Emory University, Atlanta, GA 30322
| | - Joshua Linnert
- Institute of Molecular Physiology, Johannes Gutenberg University, Mainz 55128, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg University, Mainz 55128, Germany
| | - Michael J. Lim
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - James E. Casanova
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Richard A. Kahn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
26
|
Yu SS, Wang E, Chiang CY, Cheng PH, Yeh YS, Wu YY, Chiou YY, Jiang ST. Large deletion of Wdr19 in developing renal tubules disrupts primary ciliogenesis leading to polycystic kidney disease in mice. J Pathol 2022; 257:5-16. [PMID: 35007346 DOI: 10.1002/path.5863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/03/2021] [Accepted: 01/06/2022] [Indexed: 11/11/2022]
Abstract
WD repeat domain 19 (Wdr19) is a major component of the intraflagellar transport (IFT) machinery, which is involved in the function of primary cilia. However, the effects of Wdr19 on primary cilia formation, cystogenesis, and polycystic kidney disease (PKD) progression remain unclear. To study these effects, we generated three lines of kidney-specific conditional knockout mice: Wdr19-knockout (Wdr19-KO, Wdr19f/- ::Cdh16-CreTg/0 ), Pkd1-knockout (Pkd1-KO, Pkd1f/- ::Cdh16-CreTg/0 ), and Wdr19/Pkd1-double knockout (Wdr19&Pkd1-dKO, Wdr19f/- ;Pkd1f/- ::Cdh16-CreTg/0 ) mice. Ultrastructural analysis using transmission electron microscopy (TEM) indicated that the primary cilia were almost absent at postnatal day 10 in Wdr19-KO mice compared with Pkd1-KO and wild-type (WT) mice. However, the primary cilia appeared structurally normal even if malfunctional in Pkd1-deficient cysts. The Pkd1-KO mice had the most severe PKD progression, including the shortest lifespan (14 days) and the largest renal cysts, among the three knockout lines. Thus, the molecular mechanism of renal cystogenesis in Wdr19-KO mice (primary cilia abrogation) was different from that in Pkd1-KO mice (primary cilia malfunction). In summary, Wdr19 deficiency leads to primary cilia abrogation and renal cyst formation. Wdr19 is primarily proposed to participate in retrograde IFT and to be crucial for the construction of primary cilia, which are critical organelles for tubulogenesis in the developing kidneys. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shang-Shiuan Yu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 70457, Taiwan.,National Laboratory Animal Center, National Applied Research Laboratories, Tainan, 74147, Taiwan
| | - Ellian Wang
- Department of Physiology, National Cheng Kung University Medical College, Tainan, 70101, Taiwan
| | - Chih-Ying Chiang
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan, 74147, Taiwan
| | - Po-Hao Cheng
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan, 74147, Taiwan
| | - Yu-Shan Yeh
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan, 74147, Taiwan
| | - Ying-Ying Wu
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan, 74147, Taiwan
| | - Yuan-Yow Chiou
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan, 74147, Taiwan.,Division of Pediatric Nephrology, Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Si-Tse Jiang
- National Laboratory Animal Center, National Applied Research Laboratories, Tainan, 74147, Taiwan
| |
Collapse
|
27
|
Wang W, Jack BM, Wang HH, Kavanaugh MA, Maser RL, Tran PV. Intraflagellar Transport Proteins as Regulators of Primary Cilia Length. Front Cell Dev Biol 2021; 9:661350. [PMID: 34095126 PMCID: PMC8170031 DOI: 10.3389/fcell.2021.661350] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/06/2021] [Indexed: 12/21/2022] Open
Abstract
Primary cilia are small, antenna-like organelles that detect and transduce chemical and mechanical cues in the extracellular environment, regulating cell behavior and, in turn, tissue development and homeostasis. Primary cilia are assembled via intraflagellar transport (IFT), which traffics protein cargo bidirectionally along a microtubular axoneme. Ranging from 1 to 10 μm long, these organelles typically reach a characteristic length dependent on cell type, likely for optimum fulfillment of their specific roles. The importance of an optimal cilia length is underscored by the findings that perturbation of cilia length can be observed in a number of cilia-related diseases. Thus, elucidating mechanisms of cilia length regulation is important for understanding the pathobiology of ciliary diseases. Since cilia assembly/disassembly regulate cilia length, we review the roles of IFT in processes that affect cilia assembly/disassembly, including ciliary transport of structural and membrane proteins, ectocytosis, and tubulin posttranslational modification. Additionally, since the environment of a cell influences cilia length, we also review the various stimuli encountered by renal epithelia in healthy and diseased states that alter cilia length and IFT.
Collapse
Affiliation(s)
- Wei Wang
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Brittany M Jack
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Henry H Wang
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Matthew A Kavanaugh
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Robin L Maser
- Department of Clinical Laboratory Sciences, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Pamela V Tran
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
28
|
HTR6 and SSTR3 targeting to primary cilia. Biochem Soc Trans 2021; 49:79-91. [PMID: 33599752 DOI: 10.1042/bst20191005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 12/25/2020] [Accepted: 01/20/2021] [Indexed: 12/30/2022]
Abstract
Primary cilia are hair-like projections of the cell membrane supported by an inner microtubule scaffold, the axoneme, which polymerizes out of a membrane-docked centriole at the ciliary base. By working as specialized signaling compartments, primary cilia provide an optimal environment for many G protein-coupled receptors (GPCRs) and their effectors to efficiently transmit their signals to the rest of the cell. For this to occur, however, all necessary receptors and signal transducers must first accumulate at the ciliary membrane. Serotonin receptor 6 (HTR6) and Somatostatin receptor 3 (SSTR3) are two GPCRs whose signaling in brain neuronal cilia affects cognition and is implicated in psychiatric, neurodegenerative, and oncologic diseases. Over a decade ago, the third intracellular loops (IC3s) of HTR6 and SSTR3 were shown to contain ciliary localization sequences (CLSs) that, when grafted onto non-ciliary GPCRs, could drive their ciliary accumulation. Nevertheless, these CLSs were dispensable for ciliary targeting of HTR6 and SSTR3, suggesting the presence of additional CLSs, which we have recently identified in their C-terminal tails. Herein, we review the discovery and mapping of these CLSs, as well as the state of the art regarding how these CLSs may orchestrate ciliary accumulation of these GPCRs by controlling when and where they interact with the ciliary entry and exit machinery via adaptors such as TULP3, RABL2 and the BBSome.
Collapse
|
29
|
Kobayashi T, Ishida Y, Hirano T, Katoh Y, Nakayama K. Cooperation of the IFT-A complex with the IFT-B complex is required for ciliary retrograde protein trafficking and GPCR import. Mol Biol Cell 2021; 32:45-56. [PMID: 33175651 PMCID: PMC8098818 DOI: 10.1091/mbc.e20-08-0556] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/22/2020] [Accepted: 11/03/2020] [Indexed: 12/20/2022] Open
Abstract
Cilia sense and transduce extracellular signals via specific receptors. The intraflagellar transport (IFT) machinery mediates not only bidirectional protein trafficking within cilia but also the import/export of ciliary proteins across the ciliary gate. The IFT machinery is known to comprise two multisubunit complexes, namely, IFT-A and IFT-B; however, little is known about how the two complexes cooperate to mediate ciliary protein trafficking. We here show that IFT144-IFT122 from IFT-A and IFT88-IFT52 from IFT-B make major contributions to the interface between the two complexes. Exogenous expression of the IFT88(Δα) mutant, which has decreased binding to IFT-A, partially restores the ciliogenesis defect of IFT88-knockout (KO) cells. However, IFT88(Δα)-expressing IFT88-KO cells demonstrate a defect in IFT-A entry into cilia, aberrant accumulation of IFT-B proteins at the bulged ciliary tips, and impaired import of ciliary G protein-coupled receptors (GPCRs). Furthermore, overaccumulated IFT proteins at the bulged tips appeared to be released as extracellular vesicles. These phenotypes of IFT88(Δα)-expressing IFT88-KO cells resembled those of IFT144-KO cells. These observations together indicate that the IFT-A complex cooperates with the IFT-B complex to mediate the ciliary entry of GPCRs as well as retrograde trafficking of the IFT machinery from the ciliary tip.
Collapse
Affiliation(s)
- Takuya Kobayashi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yamato Ishida
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Tomoaki Hirano
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yohei Katoh
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Kazuhisa Nakayama
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
30
|
Kerek EM, Yoon KH, Luo SY, Chen J, Valencia R, Julien O, Waskiewicz AJ, Hubbard BP. A conserved acetylation switch enables pharmacological control of tubby-like protein stability. J Biol Chem 2020; 296:100073. [PMID: 33187986 PMCID: PMC7948452 DOI: 10.1074/jbc.ra120.015839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/30/2020] [Accepted: 11/13/2020] [Indexed: 12/29/2022] Open
Abstract
Tubby-like proteins (TULPs) are characterized by a conserved C-terminal domain that binds phosphoinositides. Collectively, mammalian TULP1-4 proteins play essential roles in intracellular transport, cell differentiation, signaling, and motility. Yet, little is known about how the function of these proteins is regulated in cells. Here, we present the protein–protein interaction network of TULP3, a protein that is responsible for the trafficking of G-protein-coupled receptors to cilia and whose aberrant expression is associated with severe developmental disorders and polycystic kidney disease. We identify several protein interaction nodes linked to TULP3 that include enzymes involved in acetylation and ubiquitination. We show that acetylation of two key lysine residues on TULP3 by p300 increases TULP3 protein abundance and that deacetylation of these sites by HDAC1 decreases protein levels. Furthermore, we show that one of these sites is ubiquitinated in the absence of acetylation and that acetylation inversely correlates with ubiquitination of TULP3. This mechanism is evidently conserved across species and is active in zebrafish during development. Finally, we identify this same regulatory module in TULP1, TULP2, and TULP4 and demonstrate that the stability of these proteins is similarly modulated by an acetylation switch. This study unveils a signaling pathway that links nuclear enzymes to ciliary membrane receptors via TULP3, describes a dynamic mechanism for the regulation of all tubby-like proteins, and explores how to exploit it pharmacologically using drugs.
Collapse
Affiliation(s)
- Evan M Kerek
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Kevin H Yoon
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Shu Y Luo
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Jerry Chen
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Robert Valencia
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Olivier Julien
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Andrew J Waskiewicz
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Basil P Hubbard
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
31
|
Hu J, Harris PC. Regulation of polycystin expression, maturation and trafficking. Cell Signal 2020; 72:109630. [PMID: 32275942 PMCID: PMC7269868 DOI: 10.1016/j.cellsig.2020.109630] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 12/26/2022]
Abstract
The major autosomal dominant polycystic kidney disease (ADPKD) genes, PKD1 and PKD2, are wildly expressed at the organ and tissue level. PKD1 encodes polycystin 1 (PC1), a large membrane associated receptor-like protein that can complex with the PKD2 product, PC2. Various cellular locations have been described for both PC1, including the plasma membrane and extracellular vesicles, and PC2, especially the endoplasmic reticulum (ER), but compelling evidence indicates that the primary cilium, a sensory organelle, is the key site for the polycystin complex to prevent PKD. As with other membrane proteins, the ER biogenesis pathway is key to appropriately folding, performing quality control, and exporting fully folded PC1 to the Golgi apparatus. There is a requirement for binding with PC2 and cleavage of PC1 at the GPS for this folding and export to occur. Six different monogenic defects in this pathway lead to cystic disease development, with PC1 apparently particularly sensitive to defects in this general protein processing pathway. Trafficking of membrane proteins, and the polycystins in particular, through the Golgi to the primary cilium have been analyzed in detail, but at this time, there is no clear consensus on a ciliary targeting sequence required to export proteins to the cilium. After transitioning though the trans-Golgi network, polycystin-bearing vesicles are likely sorted to early or recycling endosomes and then transported to the ciliary base, possibly via docking to transition fibers (TF). The membrane-bound polycystin complex then undergoes facilitated trafficking through the transition zone, the diffusion barrier at the base of the cilium, before entering the cilium. Intraflagellar transport (IFT) may be involved in moving the polycystins along the cilia, but data also indicates other mechanisms. The ciliary polycystin complex can be ubiquitinated and removed from cilia by internalization at the ciliary base and may be sent back to the plasma membrane for recycling or to lysosomes for degradation. Monogenic defects in processes regulating the protein composition of cilia are associated with syndromic disorders involving many organ systems, reflecting the pleotropic role of cilia during development and for tissue maintenance. Many of these ciliopathies have renal involvement, likely because of faulty polycystin signaling from cilia. Understanding the expression, maturation and trafficking of the polycystins helps understand PKD pathogenesis and suggests opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jinghua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA; Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.
| | - Peter C Harris
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA; Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
32
|
Legué E, Liem KF. Mutations in Ciliary Trafficking Genes affect Sonic Hedgehog-dependent Neural Tube Patterning Differentially along the Anterior-Posterior Axis. Neuroscience 2020; 450:3-14. [PMID: 32682825 DOI: 10.1016/j.neuroscience.2020.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/30/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022]
Abstract
Cell specification in the ventral spinal cord is a well-studied model system to understand how tissue pattern develops in response to a morphogen gradient. Ventral cell types including motor neurons (MNs) are induced in the neural tube in response to graded Sonic Hedgehog (Shh) signaling. We performed a forward genetic screen in the mouse that incorporated a GFP-expressing transgene to visualize MNs to identify genes regulating ventral patterning. Here we contrast the neural patterning phenotypes of two mouse lines carrying induced mutations in ciliary trafficking genes. We show that a hypomorphic mutation in the gene Tubby-like protein 3 (Tulp3) resulted in a dorsal expansion of MNs consistent with an up-regulation of Shh signaling. Interestingly, patterning defects in Tulp3 mutants were restricted to posterior regions of the spinal cord as patterning was similar to WT in the anterior spinal cord. In contrast, a mutation in the ciliary trafficking gene cytoplasmic dynein 2 heavy chain 1 (Dync2h1), led to a complete loss of MNs in anterior regions of the spinal cord, indicating a strong down-regulation of Shh signaling. However, this severe phenotype was restricted to the cervical region as MNs developed posteriorly. Mutations in cilia trafficking genes affect Shh-dependent signaling in the neural tube differentially along the anterior-posterior (A-P) axis in a process that is not understood.
Collapse
Affiliation(s)
- Emilie Legué
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520, United States
| | - Karel F Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520, United States.
| |
Collapse
|
33
|
Cilia and polycystic kidney disease. Semin Cell Dev Biol 2020; 110:139-148. [PMID: 32475690 DOI: 10.1016/j.semcdb.2020.05.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 05/03/2020] [Accepted: 05/03/2020] [Indexed: 11/20/2022]
Abstract
Polycystic kidney disease (PKD), comprising autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD), is characterized by incessant cyst formation in the kidney and liver. ADPKD and ARPKD represent the leading genetic causes of renal disease in adults and children, respectively. ADPKD is caused by mutations in PKD1 encoding polycystin1 (PC1) and PKD2 encoding polycystin 2 (PC2). PC1/2 are multi-pass transmembrane proteins that form a complex localized in the primary cilium. Predominant ARPKD cases are caused by mutations in polycystic kidney and hepatic disease 1 (PKHD1) gene that encodes the Fibrocystin/Polyductin (FPC) protein, whereas a small subset of cases are caused by mutations in DAZ interacting zinc finger protein 1 like (DZIP1L) gene. FPC is a type I transmembrane protein, localizing to the cilium and basal body, in addition to other compartments, and DZIP1L encodes a transition zone/basal body protein. Apparently, PC1/2 and FPC are signaling molecules, while the mechanism that cilia employ to govern renal tubule morphology and prevent cyst formation is unclear. Nonetheless, recent genetic and biochemical studies offer a glimpse of putative physiological malfunctions and the pathomechanisms underlying both disease entities. In this review, I summarize the results of genetic studies that deduced the function of PC1/2 on cilia and of cilia themselves in cyst formation in ADPKD, and I discuss studies regarding regulation of polycystin biogenesis and cilia trafficking. I also summarize the synergistic genetic interactions between Pkd1 and Pkhd1, and the unique tissue patterning event controlled by FPC, but not PC1. Interestingly, while DZIP1L mutations generate compromised PC1/2 cilia expression, FPC deficiency does not affect PC1/2 biogenesis and ciliary localization, indicating that divergent mechanisms could lead to cyst formation in ARPKD. I conclude by outlining promising areas for future PKD research and highlight rationales for potential therapeutic interventions for PKD treatment.
Collapse
|
34
|
Distinct roles of stereociliary links in the nonlinear sound processing and noise resistance of cochlear outer hair cells. Proc Natl Acad Sci U S A 2020; 117:11109-11117. [PMID: 32358189 DOI: 10.1073/pnas.1920229117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Outer hair cells (OHCs) play an essential role in hearing by acting as a nonlinear amplifier which helps the cochlea detect sounds with high sensitivity and accuracy. This nonlinear sound processing generates distortion products, which can be measured as distortion-product otoacoustic emissions (DPOAEs). The OHC stereocilia that respond to sound vibrations are connected by three kinds of extracellular links: tip links that connect the taller stereocilia to shorter ones and convey force to the mechanoelectrical transduction channels, tectorial membrane-attachment crowns (TM-ACs) that connect the tallest stereocilia to one another and to the overlying TM, and horizontal top connectors (HTCs) that link adjacent stereocilia. While the tip links have been extensively studied, the roles that the other two types of links play in hearing are much less clear, largely because of a lack of suitable animal models. Here, while analyzing genetic combinations of tubby mice, we encountered models missing both HTCs and TM-ACs or HTCs alone. We found that the tubby mutation causes loss of both HTCs and TM-ACs due to a mislocalization of stereocilin, which results in OHC dysfunction leading to severe hearing loss. Intriguingly, the addition of the modifier allele modifier of tubby hearing 1 in tubby mice selectively rescues the TM-ACs but not the HTCs. Hearing is significantly rescued in these mice with robust DPOAE production, indicating an essential role of the TM-ACs but not the HTCs in normal OHC function. In contrast, the HTCs are required for the resistance of hearing to damage caused by noise stress.
Collapse
|
35
|
Polycystins as components of large multiprotein complexes of polycystin interactors. Cell Signal 2020; 72:109640. [PMID: 32305669 DOI: 10.1016/j.cellsig.2020.109640] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/27/2022]
Abstract
Naturally occurring mutations in two separate genes, PKD1 and PKD2, are responsible for the vast majority of all cases of autosomal dominant polycystic kidney disease (ADPKD), one of the most common genetic diseases affecting 1 in 1000 Americans. The hallmark of ADPKD is the development of epithelial cysts in the kidney, liver, and pancreas. PKD1 encodes a large plasma membrane protein (PKD1, PC1, or Polycystin-1) with a long extracellular domain and has been speculated to function as an atypical G protein coupled receptor. PKD2 encodes an ion channel of the Transient Receptor Potential superfamily (TRPP2, PKD2, PC2, or Polycystin-2). Despite the identification of these genes more than 20 years ago, the molecular function of their encoded proteins and the mechanism(s) by which mutations in PKD1 and PKD2 cause ADPKD remain elusive. Genetic, biochemical, and functional evidence suggests they form a multiprotein complex present in multiple locations in the cell, including the plasma membrane, endoplasmic reticulum, and the primary cilium. Over the years, numerous interacting proteins have been identified using directed and unbiased approaches, and shown to modulate function, cellular localization, and protein stability and turnover of Polycystins. Delineation of the molecular composition of the Polycystin complex can have a significant impact on understanding their cellular function in health and disease states and on the identification of more specific and effective therapeutic targets.
Collapse
|
36
|
Hilgendorf KI, Johnson CT, Mezger A, Rice SL, Norris AM, Demeter J, Greenleaf WJ, Reiter JF, Kopinke D, Jackson PK. Omega-3 Fatty Acids Activate Ciliary FFAR4 to Control Adipogenesis. Cell 2019; 179:1289-1305.e21. [PMID: 31761534 DOI: 10.1016/j.cell.2019.11.005] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 09/23/2019] [Accepted: 10/31/2019] [Indexed: 10/25/2022]
Abstract
Adult mesenchymal stem cells, including preadipocytes, possess a cellular sensory organelle called the primary cilium. Ciliated preadipocytes abundantly populate perivascular compartments in fat and are activated by a high-fat diet. Here, we sought to understand whether preadipocytes use their cilia to sense and respond to external cues to remodel white adipose tissue. Abolishing preadipocyte cilia in mice severely impairs white adipose tissue expansion. We discover that TULP3-dependent ciliary localization of the omega-3 fatty acid receptor FFAR4/GPR120 promotes adipogenesis. FFAR4 agonists and ω-3 fatty acids, but not saturated fatty acids, trigger mitosis and adipogenesis by rapidly activating cAMP production inside cilia. Ciliary cAMP activates EPAC signaling, CTCF-dependent chromatin remodeling, and transcriptional activation of PPARγ and CEBPα to initiate adipogenesis. We propose that dietary ω-3 fatty acids selectively drive expansion of adipocyte numbers to produce new fat cells and store saturated fatty acids, enabling homeostasis of healthy fat tissue.
Collapse
Affiliation(s)
- Keren I Hilgendorf
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Carl T Johnson
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stem Cell and Regenerative Medicine Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anja Mezger
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Selena L Rice
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Alessandra M Norris
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Janos Demeter
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| | - Daniel Kopinke
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA.
| | - Peter K Jackson
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
37
|
DiTirro D, Philbrook A, Rubino K, Sengupta P. The Caenorhabditis elegans Tubby homolog dynamically modulates olfactory cilia membrane morphogenesis and phospholipid composition. eLife 2019; 8:48789. [PMID: 31259686 PMCID: PMC6624019 DOI: 10.7554/elife.48789] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 06/21/2019] [Indexed: 12/13/2022] Open
Abstract
Plasticity in sensory signaling is partly mediated via regulated trafficking of signaling molecules to and from primary cilia. Tubby-related proteins regulate ciliary protein transport; however, their roles in remodeling cilia properties are not fully understood. We find that the C. elegans TUB-1 Tubby homolog regulates membrane morphogenesis and signaling protein transport in specialized sensory cilia. In particular, TUB-1 is essential for sensory signaling-dependent reshaping of olfactory cilia morphology. We show that compromised sensory signaling alters cilia membrane phosphoinositide composition via TUB-1-dependent trafficking of a PIP5 kinase. TUB-1 regulates localization of this lipid kinase at the cilia base in part via localization of the AP-2 adaptor complex subunit DPY-23. Our results describe new functions for Tubby proteins in the dynamic regulation of cilia membrane lipid composition, morphology, and signaling protein content, and suggest that this conserved family of proteins plays a critical role in mediating cilia structural and functional plasticity.
Collapse
Affiliation(s)
- Danielle DiTirro
- Department of Biology, Brandeis University, Waltham, United States
| | - Alison Philbrook
- Department of Biology, Brandeis University, Waltham, United States
| | - Kendrick Rubino
- Department of Biology, Brandeis University, Waltham, United States
| | - Piali Sengupta
- Department of Biology, Brandeis University, Waltham, United States
| |
Collapse
|
38
|
Legué E, Liem KF. Tulp3 Is a Ciliary Trafficking Gene that Regulates Polycystic Kidney Disease. Curr Biol 2019; 29:803-812.e5. [PMID: 30799240 DOI: 10.1016/j.cub.2019.01.054] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/12/2018] [Accepted: 01/21/2019] [Indexed: 12/17/2022]
Abstract
The primary cilium is an organelle essential for cell signaling pathways. One of the most common human genetic diseases is autosomal dominant polycystic kidney disease (ADPKD), which is caused by mutations in the PKD1 or PKD2 genes that encode Polycystin 1 and 2 (PC1/2), transmembrane proteins that translocate to the cilium. Mutations in genes that disrupt ciliogenesis also cause kidney cysts as part of a "ciliopathic" disease spectrum. The molecular mechanisms that link cilia function with renal cystic diseases are not well understood, and the mechanistic relationship between ADPKD and ciliopathic PKD is not known. Here we identify the gene Tubby-like protein-3 (Tulp3) as a key regulator of renal cystic disease from a forward genetic screen in the mouse. Mice homozygous for a hypomorphic missense mutation within the conserved Tubby domain of Tulp3 develop cysts at late embryonic stages, leading to severe postnatal loss of kidney function. In contrast to other ciliopathic disease models, Tulp3 mutations do not affect ciliogenesis. Instead, we demonstrate that Tulp3 is essential for the trafficking of the Joubert syndrome-associated small GTPase Arl13b into kidney cilia. We show that reduction of Pkd1 dosage promotes cystogenesis in the Tulp3 conditional ciliopathic PKD model. However, in an adult model of ADPKD utilizing inducible conditional Pkd1 deletion, concomitant removal of Tulp3 surprisingly ameliorates cystic disease. Therefore, Tulp3 controls distinct ciliary pathways that positively or negatively regulate cystogenesis depending on the cellular context.
Collapse
Affiliation(s)
- Emilie Legué
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Karel F Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|