1
|
Han S, Hu Y, Jia D, Lv Y, Liu M, Wang D, Chao J, Xia X, Wang Q, Liu P, Cai Y, Ren X. IFT27 regulates the long-term maintenance of photoreceptor outer segments in zebrafish. Gene 2024; 905:148237. [PMID: 38310983 DOI: 10.1016/j.gene.2024.148237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/06/2024]
Abstract
Approximately a quarter of Retinitis Pigmentosa (RP) is caused by mutations in transport-related genes in cilia. IFT27 (Intraflagellar Transport 27), a core component of the ciliary intraflagellar transport (IFT) system, has been implicated as a significant pathogenic gene in RP. The pathogenic mechanisms and subsequent pathology related to IFT27 mutations in RP are largely obscure. Here, we utilized TALEN technology to create an ift27 knockout (ift27-/-) zebrafish model. Electroretinography (ERG) detection showed impaired vision in this model. Histopathological examinations disclosed that ift27 mutations cause progressive degeneration of photoreceptors in zebrafish, and this degeneration was late-onset. Immunofluorescence labeling of outer segments showed that rods degenerated before cones, aligning with the conventional characterization of RP. In cultured human retinal pigment epithelial cells, we found that IFT27 was involved in maintaining ciliary morphology. Furthermore, decreased IFT27 expression resulted in the inhibition of the Hedgehog (Hh) signaling pathway, including decreased expression of key factors in the Hh pathway and abnormal localization of the ciliary mediator Gli2. In summary, we generated an ift27-/- zebrafish line with retinal degeneration which mimicked the symptoms of RP patients, highlighting IFT27's integral role in the long-term maintenance of cilia via the Hh signaling pathway. This work may furnish new insights into the treatment or delay of RP caused by IFT27 mutations.
Collapse
Affiliation(s)
- Shanshan Han
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China.
| | - Yue Hu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Danna Jia
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Yuexia Lv
- Prenatal Diagnosis Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Decheng Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Jin Chao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Xuan Xia
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Qiong Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Pei Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Yu Cai
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Xiang Ren
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
2
|
Sun K, Liu L, Jiang X, Wang H, Wang L, Yang Y, Liu W, Zhang L, Zhao X, Zhu X. The endoplasmic reticulum membrane protein complex subunit Emc6 is essential for rhodopsin localization and photoreceptor cell survival. Genes Dis 2024; 11:1035-1049. [PMID: 37692493 PMCID: PMC10492031 DOI: 10.1016/j.gendis.2023.03.033] [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: 09/21/2022] [Revised: 02/17/2023] [Accepted: 03/29/2023] [Indexed: 09/12/2023] Open
Abstract
The endoplasmic reticulum (ER) membrane protein complex (EMC) is responsible for monitoring the biogenesis and synthetic quality of membrane proteins with tail-anchored or multiple transmembrane domains. The EMC subunit EMC6 is one of the core members of EMC and forms an enclosed hydrophilic vestibule in cooperation with EMC3. Despite studies demonstrating that deletion of EMC3 led to rhodopsin mislocalization in rod photoreceptors of mice, the precise mechanism leading to the failure of rhodopsin trafficking remains unclear. Here, we generated the first rod photoreceptor-specific knockout of Emc6 (RKO) and cone photoreceptor-specific knockout of Emc6 (CKO) mouse models. Deficiency of Emc6 in rod photoreceptors led to progressive shortening of outer segments (OS), impaired visual function, mislocalization and reduced expression of rhodopsin, and increased gliosis in rod photoreceptors. In addition, CKO mice displayed the progressive death of cone photoreceptors and abnormal localization of cone opsin protein. Subsequently, proteomics analysis of the RKO mouse retina illustrated that several cilium-related proteins, particularly anoctamin-2 (ANO2) and transmembrane protein 67 (TMEM67), were significantly down-regulated prior to OS degeneration. Detrimental rod photoreceptor cilia and mislocalized membrane disc proteins were evident in RKO mice. Our data revealed that in addition to monitoring the synthesis of rhodopsin-dominated membrane disc proteins, EMC6 also impacted rod photoreceptors' ciliogenesis by regulating the synthesis of membrane proteins associated with cilia, contributing to the mislocalization of membrane disc proteins.
Collapse
Affiliation(s)
- Kuanxiang Sun
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining, Qinghai 810008, China
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China
| | - Lu Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Xiaoyan Jiang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Heting Wang
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Lin Wang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Yeming Yang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Wenjing Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Lin Zhang
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Xiaohui Zhao
- Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining, Qinghai 810008, China
| | - Xianjun Zhu
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining, Qinghai 810008, China
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China
- Department of Ophthalmology, The First People's Hospital of Shangqiu, Shangqiu, Henan 476000, China
| |
Collapse
|
3
|
Haggerty KN, Eshelman SC, Sexton LA, Frimpong E, Rogers LM, Agosto MA, Robichaux MA. Super-resolution mapping in rod photoreceptors identifies rhodopsin trafficking through the inner segment plasma membrane as an essential subcellular pathway. PLoS Biol 2024; 22:e3002467. [PMID: 38190419 PMCID: PMC10773939 DOI: 10.1371/journal.pbio.3002467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 12/10/2023] [Indexed: 01/10/2024] Open
Abstract
Photoreceptor cells in the vertebrate retina have a highly compartmentalized morphology for efficient phototransduction and vision. Rhodopsin, the visual pigment in rod photoreceptors, is densely packaged into the rod outer segment sensory cilium and continuously renewed through essential synthesis and trafficking pathways housed in the rod inner segment. Despite the importance of this region for rod health and maintenance, the subcellular organization of rhodopsin and its trafficking regulators in the mammalian rod inner segment remain undefined. We used super-resolution fluorescence microscopy with optimized retinal immunolabeling techniques to perform a single molecule localization analysis of rhodopsin in the inner segments of mouse rods. We found that a significant fraction of rhodopsin molecules was localized at the plasma membrane, at the surface, in an even distribution along the entire length of the inner segment, where markers of transport vesicles also colocalized. Thus, our results collectively establish a model of rhodopsin trafficking through the inner segment plasma membrane as an essential subcellular pathway in mouse rod photoreceptors.
Collapse
Affiliation(s)
- Kristen N. Haggerty
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - Shannon C. Eshelman
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - Lauren A. Sexton
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - Emmanuel Frimpong
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - Leah M. Rogers
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - Melina A. Agosto
- Retina and Optic Nerve Research Laboratory, Department of Physiology and Biophysics, and Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Michael A. Robichaux
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| |
Collapse
|
4
|
Gilmore WB, Hultgren NW, Chadha A, Barocio SB, Zhang J, Kutsyr O, Flores-Bellver M, Canto-Soler MV, Williams DS. Expression of two major isoforms of MYO7A in the retina: Considerations for gene therapy of Usher syndrome type 1B. Vision Res 2023; 212:108311. [PMID: 37586294 PMCID: PMC10984346 DOI: 10.1016/j.visres.2023.108311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/18/2023]
Abstract
Usher syndrome type 1B (USH1B) is a deaf-blindness disorder, caused by mutations in the MYO7A gene, which encodes the heavy chain of an unconventional actin-based motor protein. Here, we examined the two retinal isoforms of MYO7A, IF1 and IF2. We compared 3D models of the two isoforms and noted that the 38-amino acid region that is present in IF1 but absent from IF2 affects the C lobe of the FERM1 domain and the opening of a cleft in this potentially important protein binding domain. Expression of each of the two isoforms of human MYO7A and pig and mouse Myo7a was detected in the RPE and neural retina. Quantification by qPCR showed that the expression of IF2 was typically ∼ 7-fold greater than that of IF1. We discuss the implications of these findings for any USH1B gene therapy strategy. Given the current incomplete knowledge of the functions of each isoform, both isoforms should be considered for targeting both the RPE and the neural retina in gene augmentation therapies.
Collapse
Affiliation(s)
- W Blake Gilmore
- Department of Ophthalmology and Stein Eye Institute, Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Nan W Hultgren
- Department of Ophthalmology and Stein Eye Institute, Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Abhishek Chadha
- Department of Ophthalmology and Stein Eye Institute, Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sonia B Barocio
- Department of Ophthalmology and Stein Eye Institute, Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Joyce Zhang
- Department of Ophthalmology and Stein Eye Institute, Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Oksana Kutsyr
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, School of Medicine, Aurora, CO, USA
| | - Miguel Flores-Bellver
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, School of Medicine, Aurora, CO, USA
| | - M Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, School of Medicine, Aurora, CO, USA
| | - David S Williams
- Department of Ophthalmology and Stein Eye Institute, Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Masek M, Bachmann-Gagescu R. Control of protein and lipid composition of photoreceptor outer segments-Implications for retinal disease. Curr Top Dev Biol 2023; 155:165-225. [PMID: 38043951 DOI: 10.1016/bs.ctdb.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Vision is arguably our most important sense, and its loss brings substantial limitations to daily life for affected individuals. Light is perceived in retinal photoreceptors (PRs), which are highly specialized neurons subdivided into several compartments with distinct functions. The outer segments (OSs) of photoreceptors represent highly specialized primary ciliary compartments hosting the phototransduction cascade, which transforms incoming light into a neuronal signal. Retinal disease can result from various pathomechanisms originating in distinct subcompartments of the PR cell, or in the retinal pigment epithelium which supports the PRs. Dysfunction of primary cilia causes human disorders known as "ciliopathies", in which retinal disease is a common feature. This chapter focuses on PR OSs, discussing the mechanisms controlling their complex structure and composition. A sequence of tightly regulated sorting and trafficking events, both upstream of and within this ciliary compartment, ensures the establishment and maintenance of the adequate proteome and lipidome required for signaling in response to light. We discuss in particular our current understanding of the role of ciliopathy proteins involved in multi-protein complexes at the ciliary transition zone (CC2D2A) or BBSome (BBS1) and how their dysfunction causes retinal disease. While the loss of CC2D2A prevents the fusion of vesicles and delivery of the photopigment rhodopsin to the ciliary base, leading to early OS ultrastructural defects, BBS1 deficiency results in precocious accumulation of cholesterol in mutant OSs and decreased visual function preceding morphological changes. These distinct pathomechanisms underscore the central role of ciliary proteins involved in multiple processes controlling OS protein and lipid composition.
Collapse
Affiliation(s)
- Markus Masek
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Ruxandra Bachmann-Gagescu
- Institute of Medical Genetics, University of Zurich, Zurich, Switzerland; Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland; University Research Priority Program AdaBD, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
6
|
Haggerty KN, Eshelman SC, Sexton LA, Frimpong E, Rogers LM, Agosto MA, Robichaux MA. Mapping rhodopsin trafficking in rod photoreceptors with quantitative super-resolution microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.20.537413. [PMID: 37131638 PMCID: PMC10153271 DOI: 10.1101/2023.04.20.537413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Photoreceptor cells in the vertebrate retina have a highly compartmentalized morphology for efficient long-term phototransduction. Rhodopsin, the visual pigment in rod photoreceptors, is densely packaged into the rod outer segment sensory cilium and continuously renewed through essential synthesis and trafficking pathways housed in the rod inner segment. Despite the importance of this region for rod health and maintenance, the subcellular organization of rhodopsin and its trafficking regulators in the mammalian rod inner segment remain undefined. We used super-resolution fluorescence microscopy with optimized retinal immunolabeling techniques to perform a single molecule localization analysis of rhodopsin in the inner segments of mouse rods. We found that a significant fraction of rhodopsin molecules was localized at the plasma membrane in an even distribution along the entire length of the inner segment, where markers of transport vesicles also colocalized. Thus, our results collectively establish a model of rhodopsin trafficking through the inner segment plasma membrane as an essential subcellular pathway in mouse rod photoreceptors.
Collapse
Affiliation(s)
- Kristen N. Haggerty
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV, 26506
| | - Shannon C. Eshelman
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV, 26506
| | - Lauren A. Sexton
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV, 26506
| | - Emmanuel Frimpong
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV, 26506
| | - Leah M. Rogers
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV, 26506
| | - Melina A. Agosto
- Retina and Optic Nerve Research Laboratory, Department of Physiology and Biophysics, and Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Michael A. Robichaux
- Department of Ophthalmology & Visual Sciences and Department of Biochemistry & Molecular Medicine, West Virginia University, Morgantown, WV, 26506
| |
Collapse
|
7
|
Hofmann KP, Lamb TD. Rhodopsin, light-sensor of vision. Prog Retin Eye Res 2023; 93:101116. [PMID: 36273969 DOI: 10.1016/j.preteyeres.2022.101116] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/06/2022]
Abstract
The light sensor of vertebrate scotopic (low-light) vision, rhodopsin, is a G-protein-coupled receptor comprising a polypeptide chain with bound chromophore, 11-cis-retinal, that exhibits remarkable physicochemical properties. This photopigment is extremely stable in the dark, yet its chromophore isomerises upon photon absorption with 70% efficiency, enabling the activation of its G-protein, transducin, with high efficiency. Rhodopsin's photochemical and biochemical activities occur over very different time-scales: the energy of retinaldehyde's excited state is stored in <1 ps in retinal-protein interactions, but it takes milliseconds for the catalytically active state to form, and many tens of minutes for the resting state to be restored. In this review, we describe the properties of rhodopsin and its role in rod phototransduction. We first introduce rhodopsin's gross structural features, its evolution, and the basic mechanisms of its activation. We then discuss light absorption and spectral sensitivity, photoreceptor electrical responses that result from the activity of individual rhodopsin molecules, and recovery of rhodopsin and the visual system from intense bleaching exposures. We then provide a detailed examination of rhodopsin's molecular structure and function, first in its dark state, and then in the active Meta states that govern its interactions with transducin, rhodopsin kinase and arrestin. While it is clear that rhodopsin's molecular properties are exquisitely honed for phototransduction, from starlight to dawn/dusk intensity levels, our understanding of how its molecular interactions determine the properties of scotopic vision remains incomplete. We describe potential future directions of research, and outline several major problems that remain to be solved.
Collapse
Affiliation(s)
- Klaus Peter Hofmann
- Institut für Medizinische Physik und Biophysik (CC2), Charité, and, Zentrum für Biophysik und Bioinformatik, Humboldt-Unversität zu Berlin, Berlin, 10117, Germany.
| | - Trevor D Lamb
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia.
| |
Collapse
|
8
|
Nagel-Wolfrum K, Fadl BR, Becker MM, Wunderlich KA, Schäfer J, Sturm D, Fritze J, Gür B, Kaplan L, Andreani T, Goldmann T, Brooks M, Starostik MR, Lokhande A, Apel M, Fath KR, Stingl K, Kohl S, DeAngelis MM, Schlötzer-Schrehardt U, Kim IK, Owen LA, Vetter JM, Pfeiffer N, Andrade-Navarro MA, Grosche A, Swaroop A, Wolfrum U. Expression and subcellular localization of USH1C/harmonin in human retina provides insights into pathomechanisms and therapy. Hum Mol Genet 2023; 32:431-449. [PMID: 35997788 PMCID: PMC9851744 DOI: 10.1093/hmg/ddac211] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 01/24/2023] Open
Abstract
Usher syndrome (USH) is the most common form of hereditary deaf-blindness in humans. USH is a complex genetic disorder, assigned to three clinical subtypes differing in onset, course and severity, with USH1 being the most severe. Rodent USH1 models do not reflect the ocular phenotype observed in human patients to date; hence, little is known about the pathophysiology of USH1 in the human eye. One of the USH1 genes, USH1C, exhibits extensive alternative splicing and encodes numerous harmonin protein isoforms that function as scaffolds for organizing the USH interactome. RNA-seq analysis of human retinae uncovered harmonin_a1 as the most abundant transcript of USH1C. Bulk RNA-seq analysis and immunoblotting showed abundant expression of harmonin in Müller glia cells (MGCs) and retinal neurons. Furthermore, harmonin was localized in the terminal endfeet and apical microvilli of MGCs, presynaptic region (pedicle) of cones and outer segments (OS) of rods as well as at adhesive junctions between MGCs and photoreceptor cells (PRCs) in the outer limiting membrane (OLM). Our data provide evidence for the interaction of harmonin with OLM molecules in PRCs and MGCs and rhodopsin in PRCs. Subcellular expression and colocalization of harmonin correlate with the clinical phenotype observed in USH1C patients. We also demonstrate that primary cilia defects in USH1C patient-derived fibroblasts could be reverted by the delivery of harmonin_a1 transcript isoform. Our studies thus provide novel insights into PRC cell biology, USH1C pathophysiology and development of gene therapy treatment(s).
Collapse
Affiliation(s)
- Kerstin Nagel-Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Benjamin R Fadl
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mirjana M Becker
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Kirsten A Wunderlich
- Department of Physiological Genomics, BioMedical Center, Ludwig-Maximilian University Munich, 82152 Planegg-Martinsried, Germany
| | - Jessica Schäfer
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Daniel Sturm
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Jacques Fritze
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Burcu Gür
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Lew Kaplan
- Department of Physiological Genomics, BioMedical Center, Ludwig-Maximilian University Munich, 82152 Planegg-Martinsried, Germany
| | - Tommaso Andreani
- Computational Biology and Data Mining, Institute of Organismic & Molecular Evolution Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Tobias Goldmann
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Matthew Brooks
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Margaret R Starostik
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anagha Lokhande
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Melissa Apel
- Department of Ophthalmology, University Medical Centre Mainz, 55131 Mainz, Germany
| | - Karl R Fath
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
- Department of Biology, Queens College of CUNY, Kissena Blvd, Flushing, NY 11367, USA
| | - Katarina Stingl
- University Eye Hospital, Centre for Ophthalmology, University of Tubingen, 72076 Tubingen, Germany
| | - Susanne Kohl
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tubingen, 72076 Tubingen, Germany
| | - Margaret M DeAngelis
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, NY 14209, USA
| | | | - Ivana K Kim
- Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Leah A Owen
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT 84132, USA
| | - Jan M Vetter
- Department of Ophthalmology, University Medical Centre Mainz, 55131 Mainz, Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Centre Mainz, 55131 Mainz, Germany
| | - Miguel A Andrade-Navarro
- Computational Biology and Data Mining, Institute of Organismic & Molecular Evolution Biology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Antje Grosche
- Department of Physiological Genomics, BioMedical Center, Ludwig-Maximilian University Munich, 82152 Planegg-Martinsried, Germany
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| |
Collapse
|
9
|
The Adhesion G-Protein-Coupled Receptor GPR115/ADGRF4 Regulates Epidermal Differentiation and Associates with Cytoskeletal KRT1. Cells 2022; 11:cells11193151. [PMID: 36231117 PMCID: PMC9563031 DOI: 10.3390/cells11193151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/23/2022] [Accepted: 10/03/2022] [Indexed: 11/27/2022] Open
Abstract
Among the 33 human adhesion G-protein-coupled receptors (aGPCRs), a unique subfamily of GPCRs, only ADGRF4, encoding GPR115, shows an obvious skin-dominated transcriptomic profile, but its expression and function in skin is largely unknown. Here, we report that GPR115 is present in a small subset of basal and in most suprabasal, noncornified keratinocytes of the stratified epidermis, supporting epidermal transcriptomic data. In psoriatic skin, characterized by hyperproliferation and delayed differentiation, the expression of GPR115 and KRT1/10, the fundamental suprabasal keratin dimer, is delayed. The deletion of ADGRF4 in HaCaT keratinocytes grown in an organotypic mode abrogates KRT1 and reduces keratinocyte stratification, indicating a role of GPR115 in epidermal differentiation. Unexpectedly, endogenous GPR115, which is not glycosylated and is likely not proteolytically processed, localizes intracellularly along KRT1/10-positive keratin filaments in a regular pattern. Our data demonstrate a hitherto unknown function of GPR115 in the regulation of epidermal differentiation and KRT1.
Collapse
|
10
|
Lewandowski D, Sander CL, Tworak A, Gao F, Xu Q, Skowronska-Krawczyk D. Dynamic lipid turnover in photoreceptors and retinal pigment epithelium throughout life. Prog Retin Eye Res 2022; 89:101037. [PMID: 34971765 PMCID: PMC10361839 DOI: 10.1016/j.preteyeres.2021.101037] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022]
Abstract
The retinal pigment epithelium-photoreceptor interphase is renewed each day in a stunning display of cellular interdependence. While photoreceptors use photosensitive pigments to convert light into electrical signals, the RPE supports photoreceptors in their function by phagocytizing shed photoreceptor tips, regulating the blood retina barrier, and modulating inflammatory responses, as well as regenerating the 11-cis-retinal chromophore via the classical visual cycle. These processes involve multiple protein complexes, tightly regulated ligand-receptors interactions, and a plethora of lipids and protein-lipids interactions. The role of lipids in maintaining a healthy interplay between the RPE and photoreceptors has not been fully delineated. In recent years, novel technologies have resulted in major advancements in understanding several facets of this interplay, including the involvement of lipids in phagocytosis and phagolysosome function, nutrient recycling, and the metabolic dependence between the two cell types. In this review, we aim to integrate the complex role of lipids in photoreceptor and RPE function, emphasizing the dynamic exchange between the cells as well as discuss how these processes are affected in aging and retinal diseases.
Collapse
Affiliation(s)
- Dominik Lewandowski
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Christopher L Sander
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA; Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Aleksander Tworak
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Fangyuan Gao
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Qianlan Xu
- Department of Physiology and Biophysics, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Dorota Skowronska-Krawczyk
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA.
| |
Collapse
|
11
|
Tmem138 is localized to the connecting cilium essential for rhodopsin localization and outer segment biogenesis. Proc Natl Acad Sci U S A 2022; 119:e2109934119. [PMID: 35394880 PMCID: PMC9169668 DOI: 10.1073/pnas.2109934119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The connecting cilium (CC) of the photoreceptor provides the only route for the trafficking of the outer segment (OS) proteins. Failure of OS protein transport causes degenerative photoreceptor diseases, including retinitis pigmentosa. We demonstrate that Tmem138, a protein linked to ciliopathy, is localized to the photoreceptor CC. Germline deletion of Tmem138 abolished OS morphogenesis, followed by rapid photoreceptor degeneration. Tmem138 interacts with rhodopsin and two additional CC compartment proteins, Ahi1 and Tmem231, likely forming a membrane complex to facilitate trafficking of rhodopsin and other OS-bound proteins across the CC. The study thus implicates a new line of regulation on the delivery of OS proteins through interactions with CC membrane complex(es) and provides insights into photoreceptor ciliopathy diseases. Photoreceptor connecting cilium (CC) is structurally analogous to the transition zone (TZ) of primary cilia and gates the molecular trafficking between the inner and the outer segment (OS). Retinal dystrophies with underlying CC defects are manifested in a broad array of syndromic conditions known as ciliopathies as well as nonsyndromic retinal degenerations. Despite extensive studies, many questions remain in the mechanism of protein trafficking across the photoreceptor CC. Here, we genetically inactivated mouse Tmem138, a gene encoding a putative transmembrane protein localized to the ciliary TZ and linked to ciliopathies. Germline deletion of Tmem138 abolished OS morphogenesis, followed by rapid photoreceptor degeneration. Tmem138 was found localized to the photoreceptor CC and was required for localization of Ahi1 to the distal subdomain of the CC. Among the examined set of OS proteins, rhodopsin was mislocalized throughout the mutant cell body prior to OS morphogenesis. Ablation of Tmem138 in mature rods recapitulated the molecular changes in the germline mutants, causing failure of disc renewal and disintegration of the OS. Furthermore, Tmem138 interacts reciprocally with rhodopsin and a related protein Tmem231, and the ciliary localization of the latter was also altered in the mutant photoreceptors. Taken together, these results suggest a crucial role of Tmem138 in the functional organization of the CC, which is essential for rhodopsin localization and OS biogenesis.
Collapse
|
12
|
Grotz S, Schäfer J, Wunderlich KA, Ellederova Z, Auch H, Bähr A, Runa-Vochozkova P, Fadl J, Arnold V, Ardan T, Veith M, Santamaria G, Dhom G, Hitzl W, Kessler B, Eckardt C, Klein J, Brymova A, Linnert J, Kurome M, Zakharchenko V, Fischer A, Blutke A, Döring A, Suchankova S, Popelar J, Rodríguez-Bocanegra E, Dlugaiczyk J, Straka H, May-Simera H, Wang W, Laugwitz KL, Vandenberghe LH, Wolf E, Nagel-Wolfrum K, Peters T, Motlik J, Fischer MD, Wolfrum U, Klymiuk N. Early disruption of photoreceptor cell architecture and loss of vision in a humanized pig model of usher syndromes. EMBO Mol Med 2022; 14:e14817. [PMID: 35254721 PMCID: PMC8988205 DOI: 10.15252/emmm.202114817] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 02/04/2022] [Accepted: 02/09/2022] [Indexed: 01/17/2023] Open
Abstract
Usher syndrome (USH) is the most common form of monogenic deaf-blindness. Loss of vision is untreatable and there are no suitable animal models for testing therapeutic strategies of the ocular constituent of USH, so far. By introducing a human mutation into the harmonin-encoding USH1C gene in pigs, we generated the first translational animal model for USH type 1 with characteristic hearing defect, vestibular dysfunction, and visual impairment. Changes in photoreceptor architecture, quantitative motion analysis, and electroretinography were characteristics of the reduced retinal virtue in USH1C pigs. Fibroblasts from USH1C pigs or USH1C patients showed significantly elongated primary cilia, confirming USH as a true and general ciliopathy. Primary cells also proved their capacity for assessing the therapeutic potential of CRISPR/Cas-mediated gene repair or gene therapy in vitro. AAV-based delivery of harmonin into the eye of USH1C pigs indicated therapeutic efficacy in vivo.
Collapse
Affiliation(s)
- Sophia Grotz
- Chair of Molecular Animal Breeding and Biotechnology, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, LMU Munich, Munich, Germany
| | - Jessica Schäfer
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg University (JGU), Mainz, Germany
| | - Kirsten A Wunderlich
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg University (JGU), Mainz, Germany
| | - Zdenka Ellederova
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic
| | - Hannah Auch
- Chair of Molecular Animal Breeding and Biotechnology, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, LMU Munich, Munich, Germany
| | - Andrea Bähr
- Center for Innovative Medical Models, LMU Munich, Munich, Germany.,Large Animal Models in Cardiovascular Research, Internal Medical Department I, TU Munich, Munich, Germany
| | - Petra Runa-Vochozkova
- Large Animal Models in Cardiovascular Research, Internal Medical Department I, TU Munich, Munich, Germany
| | - Janet Fadl
- Chair of Molecular Animal Breeding and Biotechnology, LMU Munich, Munich, Germany.,Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg University (JGU), Mainz, Germany
| | - Vanessa Arnold
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg University (JGU), Mainz, Germany
| | - Taras Ardan
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic
| | - Miroslav Veith
- Ophthalmology Clinic, University Hospital Kralovske Vinohrady, Praha, Czech Republic
| | - Gianluca Santamaria
- Large Animal Models in Cardiovascular Research, Internal Medical Department I, TU Munich, Munich, Germany
| | - Georg Dhom
- Chair of Molecular Animal Breeding and Biotechnology, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, LMU Munich, Munich, Germany
| | - Wolfgang Hitzl
- Biostatistics and Data Science, Paracelsus Medical University, Salzburg, Austria
| | - Barbara Kessler
- Chair of Molecular Animal Breeding and Biotechnology, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, LMU Munich, Munich, Germany
| | - Christian Eckardt
- Center for Innovative Medical Models, LMU Munich, Munich, Germany.,Large Animal Models in Cardiovascular Research, Internal Medical Department I, TU Munich, Munich, Germany
| | - Joshua Klein
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg University (JGU), Mainz, Germany
| | - Anna Brymova
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic
| | - Joshua Linnert
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg University (JGU), Mainz, Germany
| | - Mayuko Kurome
- Chair of Molecular Animal Breeding and Biotechnology, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, LMU Munich, Munich, Germany
| | - Valeri Zakharchenko
- Chair of Molecular Animal Breeding and Biotechnology, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, LMU Munich, Munich, Germany
| | - Andrea Fischer
- Veterinary Faculty, Small Animal Clinics, LMU Munich, Munich, Germany
| | - Andreas Blutke
- Institute of Experimental Genetics, Helmholtz Center Munich, Neuherberg, Germany
| | - Anna Döring
- Veterinary Faculty, Small Animal Clinics, LMU Munich, Munich, Germany
| | - Stepanka Suchankova
- Institute of Experimental Medicine, Czech Academy of Science, Prague, Czech Republic
| | - Jiri Popelar
- Institute of Experimental Medicine, Czech Academy of Science, Prague, Czech Republic
| | - Eduardo Rodríguez-Bocanegra
- Centre for Ophthalmology, University Eye Hospital, University Hospital Tübingen, Tübingen, Germany.,Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| | - Julia Dlugaiczyk
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich (USZ), University of Zurich, Zurich, Switzerland
| | - Hans Straka
- Faculty of Biology, LMU Munich, Planegg, Germany
| | - Helen May-Simera
- Institute of Molecular Physiology, Cilia Biology, JGU Mainz, Mainz, Germany
| | - Weiwei Wang
- Grousbeck Gene Therapy Center, Mass Eye and Ear and Harvard Medical School, Boston, MA, USA
| | - Karl-Ludwig Laugwitz
- Large Animal Models in Cardiovascular Research, Internal Medical Department I, TU Munich, Munich, Germany
| | - Luk H Vandenberghe
- Grousbeck Gene Therapy Center, Mass Eye and Ear and Harvard Medical School, Boston, MA, USA
| | - Eckhard Wolf
- Chair of Molecular Animal Breeding and Biotechnology, LMU Munich, Munich, Germany.,Center for Innovative Medical Models, LMU Munich, Munich, Germany
| | - Kerstin Nagel-Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg University (JGU), Mainz, Germany.,Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University (JGU), Mainz, Germany
| | - Tobias Peters
- Centre for Ophthalmology, University Eye Hospital, University Hospital Tübingen, Tübingen, Germany.,Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, Tübingen, Germany
| | - Jan Motlik
- Institute of Animal Physiology and Genetics, Czech Academy of Science, Libechov, Czech Republic
| | - M Dominik Fischer
- Oxford Eye Hospital, Oxford University NHS Foundation Trust, Oxford, UK.,Nuffield Laboratory of Ophthalmology, NDCN, University of Oxford, Oxford, UK
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg University (JGU), Mainz, Germany
| | - Nikolai Klymiuk
- Center for Innovative Medical Models, LMU Munich, Munich, Germany.,Large Animal Models in Cardiovascular Research, Internal Medical Department I, TU Munich, Munich, Germany
| |
Collapse
|
13
|
Wheatley DN. Primary cilia: turning points in establishing their ubiquity, sensory role and the pathological consequences of dysfunction. J Cell Commun Signal 2021; 15:291-297. [PMID: 33970456 PMCID: PMC8222448 DOI: 10.1007/s12079-021-00615-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
For over 20 years it has finally become accepted that primary cilia are without doubt important cellular organelles, involved in signalling both intrinsically and extrinsically. The consequences of their agenesis, incorrect assembly and dysfunction only began to be fully appreciated after 2000, although this had been demonstrable over the previous two decades. Before 1980, biologists at large thought the organelle rudimentary or vestigial; how a well-developed cilium could be so slated beggars belief. Many pathological conditions have implicated the primary cilium as either a major or contributing factor, ranging from kidney malfunction (e.g. polycystic kidney disease) to mental aberrations. However, the questions of how the recognition of their prevalence, their sensory function, and their pathological involvement finally emerged as substantiated and verifiable facts needs to be addressed because what happened before the 1980s, and then notably between 1980 and 2000, can help guide research towards answering further questions on these issues. Here the intention is to focus on the salient findings (the turning points) that brought about changes in our knowledge of primary cilia. The literature on them is growing fast, with the total moving towards 20,000 reports, of which > 60% have been published in the last decade. PubMed indicates that nearly 1000 papers were published in 2020 alone. We also have to appreciate that the primary cilium can assume many different forms, each of which means that there must be many genes responsible for their development and final structure. This also suggests that there are many more functions than are currently known in both their sensory reception and signalling properties, probably for many highly specialised purposes. Malfunctioning in any of these roles will undoubtedly uncover further pathological conditions.
Collapse
|
14
|
Fuster-García C, García-Bohórquez B, Rodríguez-Muñoz A, Aller E, Jaijo T, Millán JM, García-García G. Usher Syndrome: Genetics of a Human Ciliopathy. Int J Mol Sci 2021; 22:6723. [PMID: 34201633 PMCID: PMC8268283 DOI: 10.3390/ijms22136723] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/21/2022] Open
Abstract
Usher syndrome (USH) is an autosomal recessive syndromic ciliopathy characterized by sensorineural hearing loss, retinitis pigmentosa and, sometimes, vestibular dysfunction. There are three clinical types depending on the severity and age of onset of the symptoms; in addition, ten genes are reported to be causative of USH, and six more related to the disease. These genes encode proteins of a diverse nature, which interact and form a dynamic protein network called the "Usher interactome". In the organ of Corti, the USH proteins are essential for the correct development and maintenance of the structure and cohesion of the stereocilia. In the retina, the USH protein network is principally located in the periciliary region of the photoreceptors, and plays an important role in the maintenance of the periciliary structure and the trafficking of molecules between the inner and the outer segments of photoreceptors. Even though some genes are clearly involved in the syndrome, others are controversial. Moreover, expression of some USH genes has been detected in other tissues, which could explain their involvement in additional mild comorbidities. In this paper, we review the genetics of Usher syndrome and the spectrum of mutations in USH genes. The aim is to identify possible mutation associations with the disease and provide an updated genotype-phenotype correlation.
Collapse
Affiliation(s)
- Carla Fuster-García
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Belén García-Bohórquez
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
| | - Ana Rodríguez-Muñoz
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
| | - Elena Aller
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Teresa Jaijo
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - José M. Millán
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| | - Gema García-García
- Molecular, Cellular and Genomics Biomedicine Research Group, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain; (C.F.-G.); (B.G.-B.); (A.R.-M.); (E.A.); (T.J.); (G.G.-G.)
- Unidad Mixta de Enfermedades Raras IIS La Fe-Centro de Investigación Príncipe Felipe, 46026 Valencia, Spain
- Biomedical Research Network for Rare Diseases, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain
| |
Collapse
|
15
|
Functional compartmentalization of photoreceptor neurons. Pflugers Arch 2021; 473:1493-1516. [PMID: 33880652 DOI: 10.1007/s00424-021-02558-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022]
Abstract
Retinal photoreceptors are neurons that convert dynamically changing patterns of light into electrical signals that are processed by retinal interneurons and ultimately transmitted to vision centers in the brain. They represent the essential first step in seeing without which the remainder of the visual system is rendered moot. To support this role, the major functions of photoreceptors are segregated into three main specialized compartments-the outer segment, the inner segment, and the pre-synaptic terminal. This compartmentalization is crucial for photoreceptor function-disruption leads to devastating blinding diseases for which therapies remain elusive. In this review, we examine the current understanding of the molecular and physical mechanisms underlying photoreceptor functional compartmentalization and highlight areas where significant knowledge gaps remain.
Collapse
|
16
|
Sánchez-Bellver L, Toulis V, Marfany G. On the Wrong Track: Alterations of Ciliary Transport in Inherited Retinal Dystrophies. Front Cell Dev Biol 2021; 9:623734. [PMID: 33748110 PMCID: PMC7973215 DOI: 10.3389/fcell.2021.623734] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/09/2021] [Indexed: 01/14/2023] Open
Abstract
Ciliopathies are a group of heterogeneous inherited disorders associated with dysfunction of the cilium, a ubiquitous microtubule-based organelle involved in a broad range of cellular functions. Most ciliopathies are syndromic, since several organs whose cells produce a cilium, such as the retina, cochlea or kidney, are affected by mutations in ciliary-related genes. In the retina, photoreceptor cells present a highly specialized neurosensory cilium, the outer segment, stacked with membranous disks where photoreception and phototransduction occurs. The daily renewal of the more distal disks is a unique characteristic of photoreceptor outer segments, resulting in an elevated protein demand. All components necessary for outer segment formation, maintenance and function have to be transported from the photoreceptor inner segment, where synthesis occurs, to the cilium. Therefore, efficient transport of selected proteins is critical for photoreceptor ciliogenesis and function, and any alteration in either cargo delivery to the cilium or intraciliary trafficking compromises photoreceptor survival and leads to retinal degeneration. To date, mutations in more than 100 ciliary genes have been associated with retinal dystrophies, accounting for almost 25% of these inherited rare diseases. Interestingly, not all mutations in ciliary genes that cause retinal degeneration are also involved in pleiotropic pathologies in other ciliated organs. Depending on the mutation, the same gene can cause syndromic or non-syndromic retinopathies, thus emphasizing the highly refined specialization of the photoreceptor neurosensory cilia, and raising the possibility of photoreceptor-specific molecular mechanisms underlying common ciliary functions such as ciliary transport. In this review, we will focus on ciliary transport in photoreceptor cells and discuss the molecular complexity underpinning retinal ciliopathies, with a special emphasis on ciliary genes that, when mutated, cause either syndromic or non-syndromic retinal ciliopathies.
Collapse
Affiliation(s)
- Laura Sánchez-Bellver
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine (IBUB-IRSJD), Universitat de Barcelona, Barcelona, Spain
| | - Vasileios Toulis
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Barcelona, Spain
- CIBERER, ISCIII, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Marfany
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine (IBUB-IRSJD), Universitat de Barcelona, Barcelona, Spain
- CIBERER, ISCIII, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
17
|
Spectrum of MYO7A Mutations in an Indigenous South African Population Further Elucidates the Nonsyndromic Autosomal Recessive Phenotype of DFNB2 to Include Both Homozygous and Compound Heterozygous Mutations. Genes (Basel) 2021; 12:genes12020274. [PMID: 33671976 PMCID: PMC7919343 DOI: 10.3390/genes12020274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 11/17/2022] Open
Abstract
MYO7A gene encodes unconventional myosin VIIA, which, when mutated, causes a phenotypic spectrum ranging from recessive hearing loss DFNB2 to deaf-blindness, Usher Type 1B (USH1B). MYO7A mutations are reported in nine DFNB2 families to date, none from sub-Saharan Africa.In DNA, from a cohort of 94 individuals representing 92 families from the Limpopo province of South Africa, eight MYO7A variations were detected among 10 individuals. Family studies identified homozygous and compound heterozygous mutations in 17 individuals out of 32 available family members. Four mutations were novel, p.Gly329Asp, p.Arg373His, p.Tyr1780Ser, and p.Pro2126Leufs*5. Two variations, p.Ser617Pro and p.Thr381Met, previously listed as of uncertain significance (ClinVar), were confirmed to be pathogenic. The identified mutations are predicted to interfere with the conformational properties of myosin VIIA through interruption or abrogation of multiple interactions between the mutant and neighbouring residues. Specifically, p.Pro2126Leufs*5, is predicted to abolish the critical site for the interactions between the tail and the motor domain essential for the autoregulation, leaving a non-functional, unregulated protein that causes hearing loss. We have identified MYO7A as a possible key deafness gene among indigenous sub-Saharan Africans. The spectrum of MYO7A mutations in this South African population points to DFNB2 as a specific entity that may occur in a homozygous or in a compound heterozygous state.
Collapse
|
18
|
Barnes CL, Malhotra H, Calvert PD. Compartmentalization of Photoreceptor Sensory Cilia. Front Cell Dev Biol 2021; 9:636737. [PMID: 33614665 PMCID: PMC7889997 DOI: 10.3389/fcell.2021.636737] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Functional compartmentalization of cells is a universal strategy for segregating processes that require specific components, undergo regulation by modulating concentrations of those components, or that would be detrimental to other processes. Primary cilia are hair-like organelles that project from the apical plasma membranes of epithelial cells where they serve as exclusive compartments for sensing physical and chemical signals in the environment. As such, molecules involved in signal transduction are enriched within cilia and regulating their ciliary concentrations allows adaptation to the environmental stimuli. The highly efficient organization of primary cilia has been co-opted by major sensory neurons, olfactory cells and the photoreceptor neurons that underlie vision. The mechanisms underlying compartmentalization of cilia are an area of intense current research. Recent findings have revealed similarities and differences in molecular mechanisms of ciliary protein enrichment and its regulation among primary cilia and sensory cilia. Here we discuss the physiological demands on photoreceptors that have driven their evolution into neurons that rely on a highly specialized cilium for signaling changes in light intensity. We explore what is known and what is not known about how that specialization appears to have driven unique mechanisms for photoreceptor protein and membrane compartmentalization.
Collapse
Affiliation(s)
| | | | - Peter D. Calvert
- Department of Ophthalmology and Visual Sciences, Center for Vision Research, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
19
|
Birkner K, Wasser B, Ruck T, Thalman C, Luchtman D, Pape K, Schmaul S, Bitar L, Krämer-Albers EM, Stroh A, Meuth SG, Zipp F, Bittner S. β1-Integrin- and KV1.3 channel-dependent signaling stimulates glutamate release from Th17 cells. J Clin Invest 2020; 130:715-732. [PMID: 31661467 DOI: 10.1172/jci126381] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 10/22/2019] [Indexed: 12/22/2022] Open
Abstract
Although the impact of Th17 cells on autoimmunity is undisputable, their pathogenic effector mechanism is still enigmatic. We discovered soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) complex proteins in Th17 cells that enable a vesicular glutamate release pathway that induces local intracytoplasmic calcium release and subsequent damage in neurons. This pathway is glutamine dependent and triggered by binding of β1-integrin to vascular cell adhesion molecule 1 (VCAM-1) on neurons in the inflammatory context. Glutamate secretion could be blocked by inhibiting either glutaminase or KV1.3 channels, which are known to be linked to integrin expression and highly expressed on stimulated T cells. Although KV1.3 is not expressed in CNS tissue, intrathecal administration of a KV1.3 channel blocker or a glutaminase inhibitor ameliorated disability in experimental neuroinflammation. In humans, T cells from patients with multiple sclerosis secreted higher levels of glutamate, and cerebrospinal fluid glutamine levels were increased. Altogether, our findings demonstrate that β1-integrin- and KV1.3 channel-dependent signaling stimulates glutamate release from Th17 cells upon direct cell-cell contact between Th17 cells and neurons.
Collapse
Affiliation(s)
- Katharina Birkner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Beatrice Wasser
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Tobias Ruck
- Department of Neurology, University of Muenster, Muenster, Germany
| | - Carine Thalman
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Dirk Luchtman
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Katrin Pape
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Samantha Schmaul
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lynn Bitar
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Albrecht Stroh
- Institute for Pathophysiology, FTN, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sven G Meuth
- Department of Neurology, University of Muenster, Muenster, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
20
|
Zimmermann C, Krämer N, Krauter S, Strand D, Sehn E, Wolfrum U, Freiwald A, Butter F, Plachter B. Autophagy interferes with human cytomegalovirus genome replication, morphogenesis, and progeny release. Autophagy 2020; 17:779-795. [PMID: 32079454 DOI: 10.1080/15548627.2020.1732686] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Viral infections are often accompanied by the induction of autophagy as an intrinsic cellular defense mechanism. Herpesviruses have developed strategies to evade autophagic degradation and to manipulate autophagy of the host cells to their benefit. Here we addressed the role of macroautophagy/autophagy in human cytomegalovirus replication and for particle morphogenesis. We found that proteins of the autophagy machinery localize to cytoplasmic viral assembly compartments and enveloped virions in the cytoplasm. Surprisingly, the autophagy receptor SQSTM1/p62 was also found to colocalize with HCMV capsids in the nucleus of infected cells. This finding indicates that the autophagy machinery interacts with HCMV already at the early nuclear stages of particle morphogenesis. The membrane-bound form of LC3 and several autophagy receptors were packaged into extracellular HCMV virions. This suggested that autophagic membranes were included during secondary envelopment of HCMV virions. To further address the importance of autophagy in HCMV infection, we generated an HCMV mutant that expressed a dominant-negative version of the protease ATG4B (BAD-ATG4BC74A). The proteolytic activity of ATG4B is required for LC3 cleavage, priming it for membrane conjugation. Surprisingly, both genome replication and virus release were enhanced in cells infected with BAD-ATG4BC74A, compared to control strains. These results show that autophagy operates as an antiviral process during HCMV infection but is dispensable for secondary HCMV particle envelopment.Abbreviations: ATG: autophagy-related; BAC: bacterial artificial chromosome; BECN1: beclin 1; CPE: cytopathic effect; cVACs: cytoplasmic viral assembly compartments; d.p.i.: days post-infection; DB: dense body; EBV: Epstein-Barr virus; galK: galactokinase; HCMV: human cytomegalovirus; HFF: human foreskin fibroblasts; IE: immediate-early; IRS: internal repeat short; LC3: MAP1LC3A/B; m.o.i.; multiplicity of infection; MCP: major capsid protein; Pp: phosphoprotein; sCP/UL48a: smallest capsid protein; TRS: terminal repeat short; UL: unique long; US: unique short.
Collapse
Affiliation(s)
- Christine Zimmermann
- Institute for Virology , University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Nadine Krämer
- Institute for Virology , University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Steffi Krauter
- Institute for Virology , University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Dennis Strand
- I. Medical Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Elisabeth Sehn
- Institute of Molecular Physiology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Anja Freiwald
- Institute for Molecular Biology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Falk Butter
- Institute for Molecular Biology, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Bodo Plachter
- Institute for Virology , University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
21
|
Oswald F, Prevo B, Acar S, Peterman EJG. Interplay between Ciliary Ultrastructure and IFT-Train Dynamics Revealed by Single-Molecule Super-resolution Imaging. Cell Rep 2020; 25:224-235. [PMID: 30282031 DOI: 10.1016/j.celrep.2018.09.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 08/08/2018] [Accepted: 09/07/2018] [Indexed: 01/10/2023] Open
Abstract
Cilia are built and maintained by intraflagellar transport (IFT), driving IFT trains back and forth along the ciliary axoneme. How IFT brings about the intricate ciliary structure and how this structure affects IFT are not well understood. We identify, using single-molecule super-resolution imaging of IFT components in living C. elegans, ciliary subdomains, enabling correlation of IFT-train dynamics to ciliary ultra-structure. In the transition zone, IFT dynamics are impaired, resulting in frequent pauses. At the ciliary base and tip, IFT trains show intriguing turnaround dynamics. Surprisingly, deletion of IFT motor kinesin-II not only affects IFT-train dynamics but also alters ciliary structure. Super-resolution imaging in these mutant animals suggests that the arrangement of IFT trains with respect to the axonemal microtubules is different than in wild-type animals. Our results reveal a complex, mutual interplay between ciliary ultrastructure and IFT-train dynamics, highlighting the importance of physical cues in the control of IFT dynamics.
Collapse
Affiliation(s)
- Felix Oswald
- Department of Physics and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, the Netherlands
| | - Bram Prevo
- Department of Physics and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, the Netherlands
| | - Seyda Acar
- Department of Physics and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, the Netherlands
| | - Erwin J G Peterman
- Department of Physics and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, 1081HV, the Netherlands.
| |
Collapse
|
22
|
Sorusch N, Yildirim A, Knapp B, Janson J, Fleck W, Scharf C, Wolfrum U. SANS (USH1G) Molecularly Links the Human Usher Syndrome Protein Network to the Intraflagellar Transport Module by Direct Binding to IFT-B Proteins. Front Cell Dev Biol 2019; 7:216. [PMID: 31637240 PMCID: PMC6787559 DOI: 10.3389/fcell.2019.00216] [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: 06/07/2019] [Accepted: 09/18/2019] [Indexed: 01/19/2023] Open
Abstract
The human Usher syndrome (USH) is a retinal ciliopathy, characterized by profound congenital deafness, variable vestibular dysfunction and pre-pubertal onset of retinitis pigmentosa. In the effected sensory cells, USH protein networks are assumed to function in ciliary transport processes. The USH1G protein SANS is a scaffold of the ciliary/periciliary USH protein network of photoreceptor cells. Moreover, SANS is associated with microtubules, the transport routes for protein delivery toward the cilium. To enlighten the role of SANS in ciliary transport processes, we aimed to identify transport related proteins associated with SANS. The intraflagellar transport (IFT) system is a conserved mechanism for bi-directional transport toward and through primary cilia. Thus, we tested the direct binding of SANS to IFT molecules, namely IFT20, IFT57, and IFT74 in 1:1 yeast-two-hybrid assay. The identified SANS-IFT interactions were validated in vitro via independent complementary interaction assays and in cells by applying membrane targeting assays. Quantitative immunofluorescence microscopy revealed the co-localization of SANS with IFT20, IFT52, and IFT57 particularly at ciliary base of wild type mouse photoreceptor cells. Analysis of photoreceptor cells of SANS knock out mice revealed the decrease of IFTs in the ciliary compartment indicating a role of SANS in the proper positioning of IFT-B molecules in primary cilia. Our study demonstrated direct binding of IFT complex B proteins IFT52 and IFT57 to the N-terminal ankyrin repeats and the central domain of SANS. Our data also indicate that pathologic mutations in the N-terminus of SANS lead to the loos of SANS binding to IFT-B molecules. Our findings provide direct evidence for a molecular link between the ciliary USH protein network and the IFT transport module in primary cilia.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Uwe Wolfrum
- Molecular Cell Biology, Institute of Molecular Physiology, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
23
|
Falasconi A, Biagioni M, Novelli E, Piano I, Gargini C, Strettoi E. Retinal Phenotype in the rd9 Mutant Mouse, a Model of X-Linked RP. Front Neurosci 2019; 13:991. [PMID: 31607844 PMCID: PMC6761883 DOI: 10.3389/fnins.2019.00991] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/03/2019] [Indexed: 12/25/2022] Open
Abstract
Retinal degeneration 9 (rd9) mice carry a mutation in the retina specific “Retinitis Pigmentosa GTPase Regulator (RPGR)” Open Reading Frame (ORF) 15 gene, located on the X chromosome and represent a rare model of X-linked Retinitis Pigmentosa (XLRP), a common and severe form of retinal degeneration (Wright et al., 2010; Tsang and Sharma, 2018). The rd9 RPGR-ORF15 mutation in mice causes lack of the protein in photoreceptors and a slow degeneration of these cells with consequent decrease in Outer Nuclear Layer (ONL) thickness and amplitude of ERG responses, as previously described (Thompson et al., 2012). However, relative rates of rod and cone photoreceptor loss, as well as secondary alterations occurring in neuronal and non-neuronal retinal cell types of rd9 mutants remain to be assessed. Aim of this study is to extend phenotype analysis of the rd9 mouse retina focusing on changes occurring in cells directly interacting with photoreceptors. To this purpose, first we estimated rod and cone survival and its degree of intraretinal variation over time; then, we studied the morphology of horizontal and bipolar cells and of the retinal pigment epithelium (RPE), extending our observations to glial cell reactivity. We found that in rd9 retinas rod (but not cone) death is the main cause of decrease in ONL thickness and that degeneration shows a high degree of intraretinal variation. Rod loss drives remodeling in the outer retina, with sprouting of second-order neurons of the rod-pathway and relative sparing of cone pathway elements. Remarkably, despite cone survival, functional defects can be clearly detected in ERG recordings in both scotopic and photopic conditions. Moderate levels of Muller cells and microglial reactivity are sided by striking attenuation of staining for RPE tight junctions, suggesting altered integrity of the outer Blood Retina Barrier (BRB). Because of many features resembling slowly progressing photoreceptor degeneration paradigms or early stages of more aggressive forms of RP, the rd9 mouse model can be considered a rare and useful tool to investigate retinal changes associated to a process of photoreceptor death sustained throughout life and to reveal disease biomarkers (e.g., BRB alterations) of human XLRP.
Collapse
Affiliation(s)
- Antonio Falasconi
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy.,Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Martina Biagioni
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Elena Novelli
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Enrica Strettoi
- Institute of Neuroscience, National Research Council (CNR), Pisa, Italy
| |
Collapse
|
24
|
Takahashi VKL, Xu CL, Takiuti JT, Apatoff MBL, Duong JK, Mahajan VB, Tsang SH. Comparison of structural progression between ciliopathy and non-ciliopathy associated with autosomal recessive retinitis pigmentosa. Orphanet J Rare Dis 2019; 14:187. [PMID: 31370859 PMCID: PMC6676605 DOI: 10.1186/s13023-019-1163-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 07/22/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND To evaluate and compare the progression of ciliopathy and non-ciliopathy autosomal recessive Retinitis Pigmentosa patients (arRP) by measuring the constriction of hyperautofluorescent rings in fundus autofluorescence (FAF) images and the progressive shortening of the ellipsoid zone line width obtained by spectral-domain optical coherence tomography (SD-OCT). RESULTS For the ciliopathy group, the estimated mean shortening of the ellipsoid zone line was 259 μm per year and the ring area decreased at a rate of 2.46 mm2 per year. For the non-ciliopathy group, the estimated mean shortening of the ellipsoid zone line was 84 μm per year and the ring area decreased at a rate of 0.7 mm2 per year. CONCLUSIONS Our study was able to quantify and compare the loss of EZ line width and short-wavelength autofluorescence (SW-AF) ring constriction progression over time for ciliopathy and non-ciliopathy arRP genes. These results may serve as a basis for modeling RP disease progression, and furthermore, they could potentially be used as endpoints in clinical trials seeking to promote cone and rod survival in RP patients.
Collapse
Affiliation(s)
- Vitor K L Takahashi
- Department of Ophthalmology, Columbia University, New York, NY, USA.,Jonas Children's Vision Care, and the Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology & Cell Biology, Columbia Stem Cell Initiative, Institute of Human Nutrition, Columbia University, New York, NY, USA.,Department of Ophthalmology, Federal University of São Paulo, São Paulo, Brazil
| | - Christine L Xu
- Department of Ophthalmology, Columbia University, New York, NY, USA.,Jonas Children's Vision Care, and the Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology & Cell Biology, Columbia Stem Cell Initiative, Institute of Human Nutrition, Columbia University, New York, NY, USA
| | - Júlia T Takiuti
- Department of Ophthalmology, Columbia University, New York, NY, USA.,Jonas Children's Vision Care, and the Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology & Cell Biology, Columbia Stem Cell Initiative, Institute of Human Nutrition, Columbia University, New York, NY, USA.,Division of Ophthalmology, University of São Paulo Medical School, São Paulo, Brazil
| | - Mary Ben L Apatoff
- Department of Ophthalmology, Columbia University, New York, NY, USA.,Jonas Children's Vision Care, and the Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology & Cell Biology, Columbia Stem Cell Initiative, Institute of Human Nutrition, Columbia University, New York, NY, USA
| | - Jimmy K Duong
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Vinit B Mahajan
- Byers Eye Institute, Omics Laboratory, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Columbia University, New York, NY, USA. .,Jonas Children's Vision Care, and the Bernard & Shirlee Brown Glaucoma Laboratory, Departments of Ophthalmology, Pathology & Cell Biology, Columbia Stem Cell Initiative, Institute of Human Nutrition, Columbia University, New York, NY, USA. .,Department of Pathology & Cell Biology, Stem Cell Initiative (CSCI), Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY, USA. .,Harkness Eye Institute, Columbia University Medical Center, 635 West 165th Street, Box 212, New York, NY, 10032, USA.
| |
Collapse
|
25
|
Abstract
Rods and cones are retinal photoreceptor neurons required for our visual sensation. Because of their highly polarized structures and well-characterized processes of G protein-coupled receptor-mediated phototransduction signaling, these photoreceptors have been excellent models for studying the compartmentalization and sorting of proteins. Rods and cones have a modified ciliary compartment called the outer segment (OS) as well as non-OS compartments. The distinct membrane protein compositions between OS and non-OS compartments suggest that the OS is separated from the rest of the cellular compartments by multiple barriers or gates that are selectively permissive to specific cargoes. This review discusses the mechanisms of protein sorting and compartmentalization in photoreceptor neurons. Proper sorting and compartmentalization of membrane proteins are required for signal transduction and transmission. This review also discusses the roles of compartmentalized signaling, which is compromised in various retinal ciliopathies.
Collapse
Affiliation(s)
- Yoshikazu Imanishi
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA;
| |
Collapse
|
26
|
Chadha A, Volland S, Baliaouri NV, Tran EM, Williams DS. The route of the visual receptor rhodopsin along the cilium. J Cell Sci 2019; 132:jcs.229526. [PMID: 30975916 DOI: 10.1242/jcs.229526] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/02/2019] [Indexed: 11/20/2022] Open
Abstract
The photoreceptor outer segment is the most elaborate primary cilium, containing large amounts of rhodopsin (RHO) in disk membranes that grow from a connecting cilium. The movement of RHO along the connecting cilium precedes formation of the disk membranes. However, the route that RHO takes has not been clearly determined; some reports suggest that it follows an intracellular, vesicular route along the axoneme, possibly as an adaptation for the high load of delivery or the morphogenesis of the disk endomembranes. We addressed this question by studying RHO in cilia of IMCD3 cells and mouse rod photoreceptors. In IMCD3 cilia, fluorescence recovery after photobleaching (FRAP) experiments with fluorescently tagged RHO supported the idea of RHO motility in the ciliary plasma membrane and was inconsistent with the hypothesis of RHO motility within the lumen of the cilium. In rod photoreceptors, FRAP of RHO-EGFP was altered by externally applied lectin, supporting the idea of plasmalemmal RHO dynamics. Quantitative immunoelectron microscopy corroborated our live-cell conclusions, as RHO was found to be distributed along the plasma membrane of the connecting cilium, with negligible labeling within the axoneme. Taken together, the present findings demonstrate RHO trafficking entirely via the ciliary plasma membrane.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Abhishek Chadha
- Departments of Ophthalmology and Neurobiology, Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095, USA
| | - Stefanie Volland
- Departments of Ophthalmology and Neurobiology, Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095, USA
| | - Natella V Baliaouri
- Departments of Ophthalmology and Neurobiology, Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095, USA
| | - Elaine M Tran
- Departments of Ophthalmology and Neurobiology, Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095, USA
| | - David S Williams
- Departments of Ophthalmology and Neurobiology, Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095, USA .,Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA.,Brain Research Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
27
|
Photoreceptor cell development requires prostaglandin signaling in the zebrafish retina. Biochem Biophys Res Commun 2019; 510:230-235. [DOI: 10.1016/j.bbrc.2019.01.073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 01/16/2019] [Indexed: 01/02/2023]
|
28
|
Photoreceptor actin dysregulation in syndromic and non-syndromic retinitis pigmentosa. Biochem Soc Trans 2018; 46:1463-1473. [PMID: 30464047 DOI: 10.1042/bst20180138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/26/2018] [Accepted: 10/12/2018] [Indexed: 01/11/2023]
Abstract
Retinitis pigmentosa (RP) is the leading cause of inherited blindness. RP is a genetically heterogeneous disorder, with more than 100 different causal genes identified in patients. Central to disease pathogenesis is the progressive loss of retinal photoreceptors. Photoreceptors are specialised sensory neurons that exhibit a complex and highly dynamic morphology. The highly polarised and elaborated architecture of photoreceptors requires precise regulation of numerous cytoskeletal elements. In recent years, significant work has been placed on investigating the role of microtubules (specifically, the acetylated microtubular axoneme of the photoreceptor connecting cilium) and their role in normal photoreceptor function. This has been driven by the emerging field of ciliopathies, human diseases arising from mutations in genes required for cilia formation or function, of which RP is a frequently reported phenotype. Recent studies have highlighted an intimate relationship between cilia and the actin cystoskeleton. This review will focus on the role of actin in photoreceptors, examining the connection between actin dysregulation in RP.
Collapse
|
29
|
Ta-Shma A, Hjeij R, Perles Z, Dougherty GW, Abu Zahira I, Letteboer SJF, Antony D, Darwish A, Mans DA, Spittler S, Edelbusch C, Cindrić S, Nöthe-Menchen T, Olbrich H, Stuhlmann F, Aprea I, Pennekamp P, Loges NT, Breuer O, Shaag A, Rein AJJT, Gulec EY, Gezdirici A, Abitbul R, Elias N, Amirav I, Schmidts M, Roepman R, Elpeleg O, Omran H. Homozygous loss-of-function mutations in MNS1 cause laterality defects and likely male infertility. PLoS Genet 2018; 14:e1007602. [PMID: 30148830 PMCID: PMC6128653 DOI: 10.1371/journal.pgen.1007602] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 09/07/2018] [Accepted: 08/02/2018] [Indexed: 12/22/2022] Open
Abstract
The clinical spectrum of ciliopathies affecting motile cilia spans impaired mucociliary clearance in the respiratory system, laterality defects including heart malformations, infertility and hydrocephalus. Using linkage analysis and whole exome sequencing, we identified two recessive loss-of-function MNS1 mutations in five individuals from four consanguineous families: 1) a homozygous nonsense mutation p.Arg242* in four males with laterality defects and infertility and 2) a homozygous nonsense mutation p.Gln203* in one female with laterality defects and recurrent respiratory infections additionally carrying homozygous mutations in DNAH5. Consistent with the laterality defects observed in these individuals, we found Mns1 to be expressed in mouse embryonic ventral node. Immunofluorescence analysis further revealed that MNS1 localizes to the axonemes of respiratory cilia as well as sperm flagella in human. In-depth ultrastructural analyses confirmed a subtle outer dynein arm (ODA) defect in the axonemes of respiratory epithelial cells resembling findings reported in Mns1-deficient mice. Ultrastructural analyses in the female carrying combined mutations in MNS1 and DNAH5 indicated a role for MNS1 in the process of ODA docking (ODA-DC) in the distal respiratory axonemes. Furthermore, co-immunoprecipitation and yeast two hybrid analyses demonstrated that MNS1 dimerizes and interacts with the ODA docking complex component CCDC114. Overall, we demonstrate that MNS1 deficiency in humans causes laterality defects (situs inversus) and likely male infertility and that MNS1 plays a role in the ODA-DC assembly.
Collapse
Affiliation(s)
- Asaf Ta-Shma
- Department of Pediatric Cardiology, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
- Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Rim Hjeij
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Zeev Perles
- Department of Pediatric Cardiology, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Gerard W. Dougherty
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Ibrahim Abu Zahira
- Department of Pediatric Cardiology, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Stef J. F. Letteboer
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Dinu Antony
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands
- Pediatric Genetics Division, Center for Pediatrics and Adolescent Medicine, Faculty of Medicine, Freiburg University, Freiburg, Germany
| | - Alaa Darwish
- Department of Pediatric Cardiology, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Dorus A. Mans
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Sabrina Spittler
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Christine Edelbusch
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Sandra Cindrić
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Tabea Nöthe-Menchen
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Heike Olbrich
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Friederike Stuhlmann
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Isabella Aprea
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Petra Pennekamp
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Niki T. Loges
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
| | - Oded Breuer
- Pediatric Pulmonology Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Avraham Shaag
- Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Azaria J. J. T. Rein
- Department of Pediatric Cardiology, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Elif Yilmaz Gulec
- University of Health Sciences, Kanuni Sultan Suleyman, Training and Research Hospital, Department of Medical Genetics, Istanbul, Turkey
| | - Alper Gezdirici
- University of Health Sciences, Kanuni Sultan Suleyman, Training and Research Hospital, Department of Medical Genetics, Istanbul, Turkey
| | - Revital Abitbul
- Pediatric Department, Ziv Medical Center, Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Nael Elias
- Saint Vincent Hospital, Nazareth, Faculty of Medicine, Bar Ilan University, Israel
| | - Israel Amirav
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
- Pediatric Pulmonology Unit, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Miriam Schmidts
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands
- Pediatric Genetics Division, Center for Pediatrics and Adolescent Medicine, Faculty of Medicine, Freiburg University, Freiburg, Germany
| | - Ronald Roepman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Orly Elpeleg
- Monique and Jacques Roboh Department of Genetic Research, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Heymut Omran
- Department of General Pediatrics, University Hospital Muenster, Muenster, Germany
- * E-mail:
| |
Collapse
|
30
|
Müller K, Schnatz A, Schillner M, Woertge S, Müller C, von Graevenitz I, Waisman A, van Minnen J, Vogelaar CF. A predominantly glial origin of axonal ribosomes after nerve injury. Glia 2018; 66:1591-1610. [DOI: 10.1002/glia.23327] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 01/24/2023]
Affiliation(s)
- Kerstin Müller
- Institute for Microanatomy and Neurobiology, University Medical Center of the Johannes Gutenberg University Mainz; Mainz 55131 Germany
| | - Andrea Schnatz
- Institute for Microanatomy and Neurobiology, University Medical Center of the Johannes Gutenberg University Mainz; Mainz 55131 Germany
- Institute of Developmental Biology and Neurobiology, Section Cellular Neurobiology, Johannes Gutenberg University Mainz; Mainz 55099 Germany
| | - Miriam Schillner
- Department of Neurology, Section Neuroimmunology; University Medical Center of the Johannes Gutenberg University Mainz; Mainz 55131 Germany
| | - Simone Woertge
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz; Mainz 55131 Germany
| | - Christina Müller
- Institute of Developmental Biology and Neurobiology, Section Cellular Neurobiology, Johannes Gutenberg University Mainz; Mainz 55099 Germany
| | - Ilse von Graevenitz
- Institute for Microanatomy and Neurobiology, University Medical Center of the Johannes Gutenberg University Mainz; Mainz 55131 Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz; Mainz 55131 Germany
| | - Jan van Minnen
- Hotchkiss Brain Institute and Cumming School of Medicine; University of Calgary, 3330 Hospital Drive NW; Calgary Alberta T2N 4N1 Canada
| | - Christina F. Vogelaar
- Institute for Microanatomy and Neurobiology, University Medical Center of the Johannes Gutenberg University Mainz; Mainz 55131 Germany
- Department of Neurology, Section Neuroimmunology; University Medical Center of the Johannes Gutenberg University Mainz; Mainz 55131 Germany
| |
Collapse
|
31
|
Gelsolin dysfunction causes photoreceptor loss in induced pluripotent cell and animal retinitis pigmentosa models. Nat Commun 2017; 8:271. [PMID: 28814713 PMCID: PMC5559447 DOI: 10.1038/s41467-017-00111-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/02/2017] [Indexed: 12/27/2022] Open
Abstract
Mutations in the Retinitis Pigmentosa GTPase Regulator (RPGR) cause X-linked RP (XLRP), an untreatable, inherited retinal dystrophy that leads to premature blindness. RPGR localises to the photoreceptor connecting cilium where its function remains unknown. Here we show, using murine and human induced pluripotent stem cell models, that RPGR interacts with and activates the actin-severing protein gelsolin, and that gelsolin regulates actin disassembly in the connecting cilium, thus facilitating rhodopsin transport to photoreceptor outer segments. Disease-causing RPGR mutations perturb this RPGR-gelsolin interaction, compromising gelsolin activation. Both RPGR and Gelsolin knockout mice show abnormalities of actin polymerisation and mislocalisation of rhodopsin in photoreceptors. These findings reveal a clinically-significant role for RPGR in the activation of gelsolin, without which abnormalities in actin polymerisation in the photoreceptor connecting cilia cause rhodopsin mislocalisation and eventual retinal degeneration in XLRP. Mutations in the Retinitis Pigmentosa GTPase Regulator (RPGR) cause retinal dystrophy, but how this arises at a molecular level is unclear. Here, the authors show in induced pluripotent stem cells and mouse knockouts that RPGR mediates actin dynamics in photoreceptors via the actin-severing protein, gelsolin.
Collapse
|
32
|
Disease mechanisms of X-linked retinitis pigmentosa due to RP2 and RPGR mutations. Biochem Soc Trans 2017; 44:1235-1244. [PMID: 27911705 DOI: 10.1042/bst20160148] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 01/24/2023]
Abstract
Photoreceptor degeneration is the prominent characteristic of retinitis pigmentosa (RP), a heterogeneous group of inherited retinal dystrophies resulting in blindness. Although abnormalities in many pathways can cause photoreceptor degeneration, one of the most important causes is defective protein transport through the connecting cilium, the structure that connects the biosynthetic inner segment with the photosensitive outer segment of the photoreceptors. The majority of patients with X-linked RP have mutations in the retinitis pigmentosa GTPase regulator (RPGR) or RP2 genes, the protein products of which are both components of the connecting cilium and associated with distinct mechanisms of protein delivery to the outer segment. RP2 and RPGR proteins are associated with severe diseases ranging from classic RP to atypical forms. In this short review, we will summarise current knowledge generated by experimental studies and knockout animal models, compare and discuss the prominent hypotheses about the two proteins' functions in retinal cell biology.
Collapse
|
33
|
May-Simera H, Nagel-Wolfrum K, Wolfrum U. Cilia - The sensory antennae in the eye. Prog Retin Eye Res 2017; 60:144-180. [PMID: 28504201 DOI: 10.1016/j.preteyeres.2017.05.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 12/21/2022]
Abstract
Cilia are hair-like projections found on almost all cells in the human body. Originally believed to function merely in motility, the function of solitary non-motile (primary) cilia was long overlooked. Recent research has demonstrated that primary cilia function as signalling hubs that sense environmental cues and are pivotal for organ development and function, tissue hoemoestasis, and maintenance of human health. Cilia share a common anatomy and their diverse functional features are achieved by evolutionarily conserved functional modules, organized into sub-compartments. Defects in these functional modules are responsible for a rapidly growing list of human diseases collectively termed ciliopathies. Ocular pathogenesis is common in virtually all classes of syndromic ciliopathies, and disruptions in cilia genes have been found to be causative in a growing number of non-syndromic retinal dystrophies. This review will address what is currently known about cilia contribution to visual function. We will focus on the molecular and cellular functions of ciliary proteins and their role in the photoreceptor sensory cilia and their visual phenotypes. We also highlight other ciliated cell types in tissues of the eye (e.g. lens, RPE and Müller glia cells) discussing their possible contribution to disease progression. Progress in basic research on the cilia function in the eye is paving the way for therapeutic options for retinal ciliopathies. In the final section we describe the latest advancements in gene therapy, read-through of non-sense mutations and stem cell therapy, all being adopted to treat cilia dysfunction in the retina.
Collapse
Affiliation(s)
- Helen May-Simera
- Institute of Molecular Physiology, Cilia Biology, Johannes Gutenberg-Universität Mainz, 55099 Mainz, Germany
| | - Kerstin Nagel-Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg-Universität Mainz, 55099 Mainz, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg-Universität Mainz, 55099 Mainz, Germany.
| |
Collapse
|
34
|
Jovancevic N, Dendorfer A, Matzkies M, Kovarova M, Heckmann JC, Osterloh M, Boehm M, Weber L, Nguemo F, Semmler J, Hescheler J, Milting H, Schleicher E, Gelis L, Hatt H. Medium-chain fatty acids modulate myocardial function via a cardiac odorant receptor. Basic Res Cardiol 2017; 112:13. [PMID: 28116519 PMCID: PMC5258789 DOI: 10.1007/s00395-017-0600-y] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 01/03/2017] [Indexed: 12/17/2022]
Abstract
Several studies have demonstrated the expression of odorant receptors (OR) in various human tissues and their involvement in different physiological and pathophysiological processes. However, the functional role of ORs in the human heart is still unclear. Here, we firstly report the functional characterization of an OR in the human heart. Initial next-generation sequencing analysis revealed the OR expression pattern in the adult and fetal human heart and identified the fatty acid-sensing OR51E1 as the most highly expressed OR in both cardiac development stages. An extensive characterization of the OR51E1 ligand profile by luciferase reporter gene activation assay identified 2-ethylhexanoic acid as a receptor antagonist and various structurally related fatty acids as novel OR51E1 ligands, some of which were detected at receptor-activating concentrations in plasma and epicardial adipose tissue. Functional investigation of the endogenous receptor was carried out by Ca2+ imaging of human stem cell-derived cardiomyocytes. Application of OR51E1 ligands induced negative chronotropic effects that depended on activation of the OR. OR51E1 activation also provoked a negative inotropic action in cardiac trabeculae and slice preparations of human explanted ventricles. These findings indicate that OR51E1 may play a role as metabolic regulator of cardiac function.
Collapse
Affiliation(s)
- Nikolina Jovancevic
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany.
| | - A Dendorfer
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-University, 80336, Munich, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - M Matzkies
- Institute for Neurophysiology, University of Cologne, 50931, Cologne, Germany
| | - M Kovarova
- Division of Pathobiochemistry and Clinical Chemistry, University of Tuebingen, 72076, Tuebingen, Germany
| | - J C Heckmann
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| | - M Osterloh
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| | - M Boehm
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| | - L Weber
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| | - F Nguemo
- Institute for Neurophysiology, University of Cologne, 50931, Cologne, Germany
| | - J Semmler
- Institute for Neurophysiology, University of Cologne, 50931, Cologne, Germany
| | - J Hescheler
- Institute for Neurophysiology, University of Cologne, 50931, Cologne, Germany
| | - H Milting
- Erich and Hanna Klessmann Institute, Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, Ruhr-University Bochum, 32545, Bad Oeynhausen, Germany
| | - E Schleicher
- Division of Pathobiochemistry and Clinical Chemistry, University of Tuebingen, 72076, Tuebingen, Germany
| | - L Gelis
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| | - H Hatt
- Department of Cell Physiology, Ruhr-University Bochum, 44801, Bochum, Germany
| |
Collapse
|
35
|
Wensel TG, Zhang Z, Anastassov IA, Gilliam JC, He F, Schmid MF, Robichaux MA. Structural and molecular bases of rod photoreceptor morphogenesis and disease. Prog Retin Eye Res 2016; 55:32-51. [PMID: 27352937 PMCID: PMC5112133 DOI: 10.1016/j.preteyeres.2016.06.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/15/2022]
Abstract
The rod cell has an extraordinarily specialized structure that allows it to carry out its unique function of detecting individual photons of light. Both the structural features of the rod and the metabolic processes required for highly amplified light detection seem to have rendered the rod especially sensitive to structural and metabolic defects, so that a large number of gene defects are primarily associated with rod cell death and give rise to blinding retinal dystrophies. The structures of the rod, especially those of the sensory cilium known as the outer segment, have been the subject of structural, biochemical, and genetic analysis for many years, but the molecular bases for rod morphogenesis and for cell death in rod dystrophies are still poorly understood. Recent developments in imaging technology, such as cryo-electron tomography and super-resolution fluorescence microscopy, in gene sequencing technology, and in gene editing technology are rapidly leading to new breakthroughs in our understanding of these questions. A summary is presented of our current understanding of selected aspects of these questions, highlighting areas of uncertainty and contention as well as recent discoveries that provide new insights. Examples of structural data from emerging imaging technologies are presented.
Collapse
Affiliation(s)
- Theodore G Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Zhixian Zhang
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ivan A Anastassov
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jared C Gilliam
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Feng He
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael F Schmid
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael A Robichaux
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
36
|
Jaiswal M, Fansa EK, Kösling SK, Mejuch T, Waldmann H, Wittinghofer A. Novel Biochemical and Structural Insights into the Interaction of Myristoylated Cargo with Unc119 Protein and Their Release by Arl2/3. J Biol Chem 2016; 291:20766-78. [PMID: 27481943 PMCID: PMC5034065 DOI: 10.1074/jbc.m116.741827] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 07/29/2016] [Indexed: 11/06/2022] Open
Abstract
Primary cilia are highly specialized small antenna-like cellular protrusions that extend from the cell surface of many eukaryotic cell types. The protein content inside cilia and cytoplasm is very different, but details of the sorting process are not understood for most ciliary proteins. Recently, we have shown that prenylated proteins are sorted according to their affinity to the carrier protein PDE6δ and the ability of Arl3 but not Arl2 to release high affinity cargo inside the cilia (Fansa, E. K., Kösling, S. K., Zent, E., Wittinghofer, A., and Ismail, S. (2016) Nat. Commun. 7, 11366). Here we address the question whether a similar principle governs the transport of myristoylated cargo by the carrier proteins Unc119a and Unc119b. We thus analyzed the binding strength of N-terminal myristoylated cargo peptides (GNAT1, NPHP3, Cystin1, RP2, and Src) to Unc119a and Unc119b proteins. The affinity between myristoylated cargo and carrier protein, Unc119, varies between subnanomolar and micromolar. Peptides derived from ciliary localizing proteins (GNAT1, NPHP3, and Cystin1) bind with high affinity to Unc119 proteins, whereas a peptide derived from a non-ciliary localizing protein (Src) has low affinity. The peptide with intermediate affinity (RP2) is localized at the ciliary transition zone as a gate keeper. We show that the low affinity peptides are released by both Arl2·GppNHp and Arl3·GppNHp, whereas the high affinity peptides are exclusively released by only Arl3·GppNHp. Determination of the x-ray structure of myristoylated NPHP3 peptide in complex with Unc119a reveals the molecular details of high affinity binding and suggests the importance of the residues at the +2 and +3 positions relative to the myristoylated glycine for high and low affinities. The mutational analysis of swapping the residues at the +2 and +3 positions between high and low affinity peptides results in reversing their affinities for Unc119a and leads to a partial mislocalization of a low affinity mutant of NPHP3.
Collapse
Affiliation(s)
| | | | | | - Tom Mejuch
- the Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | - Herbert Waldmann
- the Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
| | | |
Collapse
|
37
|
A FRAP-Based Method for Monitoring Molecular Transport in Ciliary Photoreceptor Cells In Vivo. Methods Mol Biol 2016. [PMID: 27514918 DOI: 10.1007/978-1-4939-3789-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The outer segment of rod and cone photoreceptor cells represents a highly modified primary sensory cilium. It renews on a daily basis throughout lifetime and effective vectorial transport to the cilium is essential for the maintenance of the photoreceptor cell function. Defects in molecules of transport modules lead to severe retinal ciliopathies. We have recently established a fluorescence recovery after photobleaching (FRAP)-based method to monitor molecular trafficking in living rodent photoreceptor cells. We irreversibly bleach the fluorescence of tagged molecules (e.g. eGFP-Rhodopsin) in photoreceptor cells of native vibratome sections through the retina by high laser intensity. In the laser scanning microscope, the recovery of the fluorescent signal is monitored over time and the kinetics of movements of molecules can be quantitatively ascertained.
Collapse
|
38
|
Raghupathy RK, Zhang X, Alhasani RH, Zhou X, Mullin M, Reilly J, Li W, Liu M, Shu X. Abnormal photoreceptor outer segment development and early retinal degeneration in kif3a mutant zebrafish. Cell Biochem Funct 2016; 34:429-40. [PMID: 27470972 DOI: 10.1002/cbf.3205] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 01/09/2023]
Abstract
Photoreceptors are highly specialized sensory neurons that possess a modified primary cilium called the outer segment. Photoreceptor outer segment formation and maintenance require highly active protein transport via a process known as intraflagellar transport. Anterograde transport in outer segments is powered by the heterotrimeric kinesin II and coordinated by intraflagellar transport proteins. Here, we describe a new zebrafish model carrying a nonsense mutation in the kinesin II family member 3A (kif3a) gene. Kif3a mutant zebrafish exhibited curved body axes and kidney cysts. Outer segments were not formed in most parts of the mutant retina, and rhodopsin was mislocalized, suggesting KIF3A has a role in rhodopsin trafficking. Both rod and cone photoreceptors degenerated rapidly between 4 and 9 days post fertilization, and electroretinography response was not detected in 7 days post fertilization mutant larvae. Loss of KIF3A in zebrafish also resulted in an intracellular transport defect affecting anterograde but not retrograde transport of organelles. Our results indicate KIF3A plays a conserved role in photoreceptor outer segment formation and intracellular transport.
Collapse
Affiliation(s)
| | - Xun Zhang
- Department of Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Reem H Alhasani
- Department of Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Xinzhi Zhou
- Department of Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | | | - James Reilly
- Department of Life Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Wenchang Li
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xinhua Shu
- Department of Life Sciences, Glasgow Caledonian University, Glasgow, UK
| |
Collapse
|
39
|
Burgoyne T, Meschede IP, Futter CE. New light on photoreceptor renewal. Cell Cycle 2016; 15:1389-90. [PMID: 27153199 DOI: 10.1080/15384101.2016.1164579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- T Burgoyne
- a UCL Institute of Ophthalmology , London , UK
| | | | - C E Futter
- a UCL Institute of Ophthalmology , London , UK
| |
Collapse
|
40
|
Burgoyne T, Meschede IP, Burden JJ, Bailly M, Seabra MC, Futter CE. Rod disc renewal occurs by evagination of the ciliary plasma membrane that makes cadherin-based contacts with the inner segment. Proc Natl Acad Sci U S A 2015; 112:15922-7. [PMID: 26668363 PMCID: PMC4702997 DOI: 10.1073/pnas.1509285113] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The outer segments of vertebrate rod photoreceptors are renewed every 10 d. Outer segment components are transported from the site of synthesis in the inner segment through the connecting cilium, followed by assembly of the highly ordered discs. Two models of assembly of discrete discs involving either successive fusion events between intracellular rhodopsin-bearing vesicles or the evagination of the plasma membrane followed by fusion of adjacent evaginations have been proposed. Here we use immuno-electron microscopy and electron tomography to show that rhodopsin is transported from the inner to the outer segment via the ciliary plasma membrane, subsequently forming successive evaginations that "zipper" up proximally, but at their leading edges are free to make junctions containing the protocadherin, PCDH21, with the inner segment plasma membrane. Given the physical dimensions of the evaginations, coupled with likely instability of the membrane cortex at the distal end of the connecting cilium, we propose that the evagination occurs via a process akin to blebbing and is not driven by actin polymerization. Disassembly of these junctions is accompanied by fusion of the leading edges of successive evaginations to form discrete discs. This fusion is topologically different to that mediated by the membrane fusion proteins, SNAREs, as initial fusion is between exoplasmic leaflets, and is accompanied by gain of the tetraspanin rim protein, peripherin.
Collapse
Affiliation(s)
- Thomas Burgoyne
- Institute of Ophthalmology, University College London, London, EC1V9EL, United Kingdom
| | - Ingrid P Meschede
- Institute of Ophthalmology, University College London, London, EC1V9EL, United Kingdom
| | - Jemima J Burden
- Laboratory for Molecular and Cell Biology, University College London, London, WC1E6BT, United Kingdom
| | - Maryse Bailly
- Institute of Ophthalmology, University College London, London, EC1V9EL, United Kingdom
| | - Miguel C Seabra
- National Heart and Lung Institute, Imperial College, London, SW72AZ, United Kingdom; Centro de Estudos de Doenças Crónicas, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | - Clare E Futter
- Institute of Ophthalmology, University College London, London, EC1V9EL, United Kingdom;
| |
Collapse
|
41
|
Zaki MS, Heller R, Thoenes M, Nürnberg G, Stern-Schneider G, Nürnberg P, Karnati S, Swan D, Fateen E, Nagel-Wolfrum K, Mostafa MI, Thiele H, Wolfrum U, Baumgart-Vogt E, Bolz HJ. PEX6 is Expressed in Photoreceptor Cilia and Mutated in Deafblindness with Enamel Dysplasia and Microcephaly. Hum Mutat 2015; 37:170-4. [PMID: 26593283 DOI: 10.1002/humu.22934] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/12/2015] [Indexed: 02/06/2023]
Abstract
Deafblindness is part of several genetic disorders. We investigated a consanguineous Egyptian family with two siblings affected by congenital hearing loss and retinal degeneration, initially diagnosed as Usher syndrome type 1. At teenage, severe enamel dysplasia, developmental delay, and microcephaly became apparent. Genome-wide homozygosity mapping and whole-exome sequencing detected a homozygous missense mutation, c.1238G>T (p.Gly413Val), affecting a highly conserved residue of peroxisomal biogenesis factor 6, PEX6. Biochemical profiling of the siblings revealed abnormal and borderline plasma phytanic acid concentration, and cerebral imaging revealed white matter disease in both. We show that Pex6 localizes to the apical extensions of secretory ameloblasts and differentiated odontoblasts at early stages of dentin synthesis in mice, and to cilia of retinal photoreceptor cells. We propose PEX6, and possibly other peroxisomal genes, as candidate for the rare cooccurrence of deafblindness and enamel dysplasia. Our study for the first time links peroxisome biogenesis disorders to retinal ciliopathies.
Collapse
Affiliation(s)
- Maha S Zaki
- Department of Clinical Genetics, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Raoul Heller
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany
| | - Michaela Thoenes
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany
| | - Gudrun Nürnberg
- Cologne Center for Genomics (CCG) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Gabi Stern-Schneider
- Department of Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Srikanth Karnati
- Institute for Anatomy and Cell Biology, Justus Liebig University, Giessen, Germany
| | - Daniel Swan
- Computational Biology Group, Oxford Gene Technology, Oxford, United Kingdom
| | - Ekram Fateen
- Department of Biochemical Genetics, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Kerstin Nagel-Wolfrum
- Department of Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Mostafa I Mostafa
- Department of Orodental Genetics, Orodental Research Division, National Research Centre, Cairo, Egypt
| | - Holger Thiele
- Cologne Center for Genomics (CCG) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Uwe Wolfrum
- Department of Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | | | - Hanno J Bolz
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany.,Bioscientia Center for Human Genetics, Ingelheim, Germany
| |
Collapse
|
42
|
Jansen F, Kalbe B, Scholz P, Mikosz M, Wunderlich KA, Kurtenbach S, Nagel-Wolfrum K, Wolfrum U, Hatt H, Osterloh S. Impact of the Usher syndrome on olfaction. Hum Mol Genet 2015; 25:524-33. [PMID: 26620972 DOI: 10.1093/hmg/ddv490] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/24/2015] [Indexed: 12/21/2022] Open
Abstract
Usher syndrome is a genetically and clinically heterogeneous disease in humans, characterized by sensorineural hearing loss, retinitis pigmentosa and vestibular dysfunction. This disease is caused by mutations in genes encoding proteins that form complex networks in different cellular compartments. Currently, it remains unclear whether the Usher proteins also form networks within the olfactory epithelium (OE). Here, we describe Usher gene expression at the mRNA and protein level in the OE of mice and showed interactions between these proteins and olfactory signaling proteins. Additionally, we analyzed the odor sensitivity of different Usher syndrome mouse models using electro-olfactogram recordings and monitored significant changes in the odor detection capabilities in mice expressing mutant Usher proteins. Furthermore, we observed changes in the expression of signaling proteins that might compensate for the Usher protein deficiency. In summary, this study provides novel insights into the presence and purpose of the Usher proteins in olfactory signal transduction.
Collapse
Affiliation(s)
- Fabian Jansen
- Cell Physiology, Faculty for Biology and Biotechnology, Ruhr-University Bochum, 44801 Bochum, Germany and
| | - Benjamin Kalbe
- Cell Physiology, Faculty for Biology and Biotechnology, Ruhr-University Bochum, 44801 Bochum, Germany and
| | - Paul Scholz
- Cell Physiology, Faculty for Biology and Biotechnology, Ruhr-University Bochum, 44801 Bochum, Germany and
| | - Marta Mikosz
- Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg-University, 55099 Mainz, Germany
| | - Kirsten A Wunderlich
- Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg-University, 55099 Mainz, Germany
| | - Stefan Kurtenbach
- Cell Physiology, Faculty for Biology and Biotechnology, Ruhr-University Bochum, 44801 Bochum, Germany and
| | - Kerstin Nagel-Wolfrum
- Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg-University, 55099 Mainz, Germany
| | - Uwe Wolfrum
- Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg-University, 55099 Mainz, Germany
| | - Hanns Hatt
- Cell Physiology, Faculty for Biology and Biotechnology, Ruhr-University Bochum, 44801 Bochum, Germany and
| | - Sabrina Osterloh
- Cell Physiology, Faculty for Biology and Biotechnology, Ruhr-University Bochum, 44801 Bochum, Germany and
| |
Collapse
|
43
|
On the Ultrastructure and Function of Rhogocytes from the Pond Snail Lymnaea stagnalis. PLoS One 2015; 10:e0141195. [PMID: 26488403 PMCID: PMC4619347 DOI: 10.1371/journal.pone.0141195] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 10/06/2015] [Indexed: 11/19/2022] Open
Abstract
Rhogocytes, also termed "pore cells", occur as solitary or clustered cells in the connective tissue of gastropod molluscs. Rhogocytes possess an enveloping lamina of extracellular matrix and enigmatic extracellular lacunae bridged by cytoplasmic bars that form 20 nm diaphragmatic slits likely to act as a molecular sieve. Recent papers highlight the embryogenesis and ultrastructure of these cells, and their role in heavy metal detoxification. Rhogocytes are the site of hemocyanin or hemoglobin biosynthesis in gastropods. Based on electron microscopy, we recently proposed a possible pathway of hemoglobin exocytosis through the slit apparatus, and provided molecular evidence of a common phylogenetic origin of molluscan rhogocytes, insect nephrocytes and vertebrate podocytes. However, the previously proposed secretion mode of the respiratory proteins into the hemolymph is still rather hypothetical, and the possible role of rhogocytes in detoxification requires additional data. Although our previous study on rhogocytes of the red-blooded (hemoglobin-containing) freshwater snail Biomphalaria glabrata provided much new information, a disadvantage was that the hemoglobin molecules were not unequivocally defined in the electron microscope. This made it difficult to trace the exocytosis pathway of this protein. Therefore, we have now performed a similar study on the rhogocytes of the blue-blooded (hemocyanin-containing) freshwater snail Lymnaea stagnalis. The intracellular hemocyanin could be identified in the electron microscope, either as individual molecules or as pseudo-crystalline arrays. Based on 3D-electron microscopy, and supplemented by in situ hybridization, immunocytochemistry and stress response experiments, we provide here additional details on the structure and hemocyanin biosynthesis of rhogocytes, and on their response in animals under cadmium and starvation stress. Moreover, we present an advanced model on the release of synthesized hemocyanin molecules through the slit apparatus into the hemolymph, and the uptake of much smaller particles such as cadmium ions from the hemolymph through the slit apparatus into the cytoplasm.
Collapse
|
44
|
Jiang L, Tam BM, Ying G, Wu S, Hauswirth WW, Frederick JM, Moritz OL, Baehr W. Kinesin family 17 (osmotic avoidance abnormal-3) is dispensable for photoreceptor morphology and function. FASEB J 2015; 29:4866-80. [PMID: 26229057 DOI: 10.1096/fj.15-275677] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/27/2015] [Indexed: 01/22/2023]
Abstract
In Caenorhabditis elegans, homodimeric [kinesin family (KIF) 17, osmotic avoidance abnormal-3 (OSM-3)] and heterotrimeric (KIF3) kinesin-2 motors are required to establish sensory cilia by intraflagellar transport (IFT) where KIF3 and KIF17 cooperate to build the axoneme core and KIF17 builds the distal segments. However, the function of KIF17 in vertebrates is unresolved. We expressed full-length and motorless KIF17 constructs in mouse rod photoreceptors using adeno-associated virus in Xenopus laevis rod photoreceptors using a transgene and in ciliated IMCD3 cells. We found that tagged KIF17 localized along the rod outer segment axoneme when expressed in mouse and X. laevis photoreceptors, whereas KIF3A was restricted to the proximal axoneme. Motorless KIF3A and KIF17 mutants caused photoreceptor degeneration, likely through dominant negative effects on IFT. KIF17 mutant lacking the motor domain translocated to nuclei after exposure of a C-terminal nuclear localization signal. Germ-line deletion of Kif17 in mouse did not affect photoreceptor function. A rod-specific Kif3/Kif17 double knockout mouse demonstrated that KIF17 and KIF3 do not act synergistically and did not prevent rhodopsin trafficking to rod outer segments. In summary, the nematode model of KIF3/KIF17 cooperation apparently does not apply to mouse photoreceptors in which the photosensory cilium is built exclusively by KIF3.
Collapse
Affiliation(s)
- Li Jiang
- *Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, and Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah, USA; Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada; State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China; Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, USA; and Department of Biology, University of Utah, Salt Lake City, Utah, USA
| | - Beatrice M Tam
- *Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, and Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah, USA; Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada; State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China; Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, USA; and Department of Biology, University of Utah, Salt Lake City, Utah, USA
| | - Guoxing Ying
- *Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, and Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah, USA; Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada; State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China; Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, USA; and Department of Biology, University of Utah, Salt Lake City, Utah, USA
| | - Sen Wu
- *Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, and Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah, USA; Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada; State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China; Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, USA; and Department of Biology, University of Utah, Salt Lake City, Utah, USA
| | - William W Hauswirth
- *Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, and Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah, USA; Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada; State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China; Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, USA; and Department of Biology, University of Utah, Salt Lake City, Utah, USA
| | - Jeanne M Frederick
- *Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, and Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah, USA; Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada; State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China; Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, USA; and Department of Biology, University of Utah, Salt Lake City, Utah, USA
| | - Orson L Moritz
- *Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, and Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah, USA; Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada; State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China; Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, USA; and Department of Biology, University of Utah, Salt Lake City, Utah, USA
| | - Wolfgang Baehr
- *Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, and Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah, USA; Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, British Columbia, Canada; State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China; Department of Ophthalmology, University of Florida College of Medicine, Gainesville, Florida, USA; and Department of Biology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
45
|
Abstract
Inherited retinal degeneration (IRD) may occur in isolation or as part of a multi-systemic condition. Ocular manifestations may be the presenting symptom of a syndromic disease and can include retinitis pigmentosa, cone-rod dystrophy, or maculopathy. Alternatively, patients affected with syndromic disease may already have other systemic manifestations at the time retinal disease is diagnosed. Some of these systemic diseases can cause significant morbidity. Here, we review several of these syndromic IRDs and their underlying genetic causes. Early recognition and referral for systemic evaluation and surveillance may lead to early intervention and an improved outcome. Obtaining a molecular diagnosis can be beneficial in securing a definitive diagnosis, especially in cases with atypical presentations. A genetic diagnosis may also be informative with regard to prognosis and potential therapies. Effective management and rehabilitation for patients with syndromic retinal dystrophy requires a comprehensive genetic-based team approach involving patients, family members, ophthalmologists, primary care physicians, and geneticists.
Collapse
Affiliation(s)
- Xiang Q Werdich
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School , Boston, Massachusetts , USA
| | | | | |
Collapse
|
46
|
Chuang JZ, Hsu YC, Sung CH. Ultrastructural visualization of trans-ciliary rhodopsin cargoes in mammalian rods. Cilia 2015; 4:4. [PMID: 25664179 PMCID: PMC4320831 DOI: 10.1186/s13630-015-0013-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 01/15/2015] [Indexed: 11/28/2022] Open
Abstract
Background Cilia are vital to various cellular and sensory functions. The pathway by which ciliary membrane proteins translocate through the transition zone is not well understood. Direct morphological characterization of ciliary cargoes in transit remains lacking. In the vertebrate photoreceptor, rhodopsin is synthesized and transported from the inner segment to the disc membranes of the outer segment (OS), which is a modified cilium. To date, the membrane topology of the basal OS and the mechanisms by which rhodopsin is transported through the transition zone (i.e., connecting cilium) and by which nascent disc membranes are formed remain controversial. Results Using an antibody recognizing its cytoplasmic C-terminus, we localize rhodopsin on both the plasma membrane and lumen of the connecting cilium by immuno-electron microscopy (EM). We also use transmission EM to visualize the electron-dense enzymatic products derived from the rhodopsin-horseradish peroxidase (HRP) fusion in transfected rodent rods. In the connecting cilium, rhodopsin is not only expressed in the plasma membrane but also in the lumen on two types of membranous carriers, long smooth tubules and small, coated, filament-bound vesicles. Additionally, membrane-bound rhodopsin carriers are also found in close proximity to the nascent discs at the basal OS axoneme and in the distal inner segment. This topology-indicative HRP-rhodopsin reporter shows that the nascent basalmost discs and the mature discs have the same membrane topology, with no indication of evagination or invagination from the basal OS plasma membranes. Serial block face and focus ion beam scanning EM analyses both indicate that the transport carriers enter the connecting cilium lumen from either the basal body lumen or cytoplasmic space between the axonemal microtubules and the ciliary plasma membrane. Conclusions Our results suggest the existence of multiple ciliary gate entry pathways in rod photoreceptors. Rhodopsin is likely transported across the connecting cilium on the plasma membrane and through the lumens on two types of tubulovesicular carriers produced in the inner segment. Our findings agree with a previous model that rhodopsin carriers derived from the cell body may fuse directly onto nascent discs as they grow and mature. Electronic supplementary material The online version of this article (doi:10.1186/s13630-015-0013-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jen-Zen Chuang
- Department of Ophthalmology, Dyson Vision Research Institute, Weill Medical College of Cornell University, New York, NY 10065 USA
| | - Ya-Chu Hsu
- Department of Ophthalmology, Dyson Vision Research Institute, Weill Medical College of Cornell University, New York, NY 10065 USA
| | - Ching-Hwa Sung
- Department of Ophthalmology, Dyson Vision Research Institute, Weill Medical College of Cornell University, New York, NY 10065 USA ; Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY 10067 USA ; The Margaret M. Dyson Vision Research Institute, Weill Medical College of Cornell University, 1300 York Avenue, LC313, New York, NY 10065 USA
| |
Collapse
|
47
|
Usher syndrome protein network functions in the retina and their relation to other retinal ciliopathies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 801:527-33. [PMID: 24664740 DOI: 10.1007/978-1-4614-3209-8_67] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human Usher syndrome (USH) is the most frequent cause of combined hereditary deaf-blindness. USH is genetically and clinically heterogeneous: 15 chromosomal loci assigned to 3 clinical types, USH1-3. All USH1 and 2 proteins are organized into protein networks by the scaffold proteins harmonin (USH1C), whirlin (USH2D) and SANS (USH1G). This has contributed essentially to our current understanding of the USH protein function in the eye and the ear and explains why defects in proteins of different families cause very similar phenotypes. Ongoing in depth analyses of USH protein networks in the eye indicated cytoskeletal functions as well as roles in molecular transport processes and ciliary cargo delivery in photoreceptor cells. The analysis of USH protein networks revealed molecular links of USH to other ciliopathies, including non-syndromic inner ear defects and isolated retinal dystrophies but also to kidney diseases and syndromes like the Bardet-Biedl syndrome. These findings provide emerging evidence that USH is a ciliopathy molecularly related to other ciliopathies, which opens an avenue for common therapy strategies to treat these diseases.
Collapse
|
48
|
Kokkinopoulou M, Güler MA, Lieb B, Barbeck M, Ghanaati S, Markl J. 3D-ultrastructure, functions and stress responses of gastropod (Biomphalaria glabrata) rhogocytes. PLoS One 2014; 9:e101078. [PMID: 24971744 PMCID: PMC4074132 DOI: 10.1371/journal.pone.0101078] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 06/03/2014] [Indexed: 11/18/2022] Open
Abstract
Rhogocytes are pore cells scattered among the connective tissue of different body parts of gastropods and other molluscs, with great variation in their number, shape and size. They are enveloped by a lamina of extracellular matrix. Their most characteristic feature is the "slit apparatus", local invaginations of the plasma membrane bridged by cytoplasmic bars, forming slits of ca. 20 nm width. A slit diaphragm creates a molecular sieve with permeation holes of 20×20 nm. In blue-blooded gastropods, rhogocytes synthesize and secrete the respiratory protein hemocyanin, and it has been proposed-though not proven-that in the rare red-blooded snail species they might synthesize and secrete the hemoglobin. However, the cellular secretion pathway for respiratory proteins, and the functional role(s) of the enigmatic rhogocyte slit apparatus are still unclear. Additional functions for rhogocytes have been proposed, notably a role in protein uptake and degradation, and in heavy metal detoxification. Here we provide new structural and functional information on the rhogocytes of the red-blooded freshwater snail Biomphalaria glabrata. By in situ hybridization of mantle tissues, we prove that rhogocytes indeed synthesize hemoglobin. By electron tomography, the first three dimensional (3D) reconstructions of the slit apparatus are provided, showing detail of highly dense material in the cytoplasmic bars close to the slits. By immunogold labelling, we collected evidence that a major component of this material is actin. By genome databank mining, the complete sequence of a B. glabrata nephrin was obtained, and localized to the rhogocytes by immunofluorescence microscopy. The presence of both proteins fit the ultrastructure-based hypothesis that rhogocytes are related to mammalian podocytes and insect nephrocytes. Reactions of the rhogocytes to deprivation of food and cadmium toxification are also documented, and a possible secretion pathway of newly synthesized respiratory proteins through the slit apparatus is discussed.
Collapse
Affiliation(s)
| | | | - Bernhard Lieb
- Institute of Zoology, Johannes Gutenberg University, Mainz, Germany
| | - Mike Barbeck
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Shahram Ghanaati
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jürgen Markl
- Institute of Zoology, Johannes Gutenberg University, Mainz, Germany
- * E-mail:
| |
Collapse
|
49
|
Becirovic E, Nguyen ONP, Paparizos C, Butz ES, Stern-Schneider G, Wolfrum U, Hauck SM, Ueffing M, Wahl-Schott C, Michalakis S, Biel M. Peripherin-2 couples rhodopsin to the CNG channel in outer segments of rod photoreceptors. Hum Mol Genet 2014; 23:5989-97. [PMID: 24963162 DOI: 10.1093/hmg/ddu323] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Outer segments (OSs) of rod photoreceptors are cellular compartments specialized in the conversion of light into electrical signals. This process relies on the light-triggered change in the intracellular levels of cyclic guanosine monophosphate, which in turn controls the activity of cyclic nucleotide-gated (CNG) channels in the rod OS plasma membrane. The rod CNG channel is a macromolecular complex that in its core harbors the ion-conducting CNGA1 and CNGB1a subunits. To identify additional proteins of the complex that interact with the CNGB1a core subunit, we applied affinity purification of mouse retinal proteins followed by mass spectrometry. In combination with in vitro and in vivo co-immunoprecipitation and fluorescence resonance energy transfer (FRET), we found that the tetraspanin peripherin-2 links CNGB1a to the light-detector rhodopsin. Using immunoelectron microscopy, we found that this peripherin-2/rhodopsin/CNG channel complex localizes to the contact region between the disk rims and the plasma membrane. FRET measurements revealed that the fourth transmembrane domain (TM4) of peripherin-2 is required for the interaction with rhodopsin. Quantitatively, the binding affinity of the peripherin-2/rhodopsin interaction was in a similar range as that observed for rhodopsin dimers. Finally, we demonstrate that the p.G266D retinitis pigmentosa mutation found within TM4 selectively abolishes the binding of peripherin-2 to rhodopsin. This finding suggests that the specific disruption of the rhodopsin/peripherin-2 interaction in the p.G266D mutant might contribute to the pathophysiology in affected persons.
Collapse
Affiliation(s)
- Elvir Becirovic
- Munich Center for Integrated Protein Science CIPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, München, Germany
| | - O N Phuong Nguyen
- Munich Center for Integrated Protein Science CIPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, München, Germany
| | - Christos Paparizos
- Munich Center for Integrated Protein Science CIPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, München, Germany
| | - Elisabeth S Butz
- Munich Center for Integrated Protein Science CIPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, München, Germany
| | - Gabi Stern-Schneider
- Cell and Matrix Biology, Institute of Zoology, Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Uwe Wolfrum
- Cell and Matrix Biology, Institute of Zoology, Johannes-Gutenberg Universität Mainz, Mainz, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, München, Germany and
| | - Marius Ueffing
- Research Unit Protein Science, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, München, Germany and Center for Ophthalmology, Institute for Ophthalmic Research, Tübingen, Germany
| | - Christian Wahl-Schott
- Munich Center for Integrated Protein Science CIPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, München, Germany
| | - Stylianos Michalakis
- Munich Center for Integrated Protein Science CIPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, München, Germany
| | - Martin Biel
- Munich Center for Integrated Protein Science CIPSM and Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, München, Germany,
| |
Collapse
|
50
|
Karlstetter M, Sorusch N, Caramoy A, Dannhausen K, Aslanidis A, Fauser S, Boesl MR, Nagel-Wolfrum K, Tamm ER, Jägle H, Stoehr H, Wolfrum U, Langmann T. Disruption of the retinitis pigmentosa 28 gene Fam161a in mice affects photoreceptor ciliary structure and leads to progressive retinal degeneration. Hum Mol Genet 2014; 23:5197-210. [DOI: 10.1093/hmg/ddu242] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|