1
|
Boldizar H, Friedman A, Stanley T, Padilla M, Galdieri J, Sclar A, Stawicki TM. The role of cilia in the development, survival, and regeneration of hair cells. Biol Open 2024; 13:bio061690. [PMID: 39263863 DOI: 10.1242/bio.061690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024] Open
Abstract
Mutations impacting cilia genes lead to a class of human diseases known as ciliopathies. This is due to the role of cilia in the development, survival, and regeneration of many cell types. We investigated the extent to which disrupting cilia impacted these processes in lateral line hair cells of zebrafish. We found that mutations in two intraflagellar transport (IFT) genes, ift88 and dync2h1, which lead to the loss of kinocilia, caused increased hair cell apoptosis. IFT gene mutants also have a decreased mitochondrial membrane potential, and blocking the mitochondrial uniporter causes a loss of hair cells in wild-type zebrafish but not mutants, suggesting mitochondria dysfunction may contribute to the apoptosis seen in these mutants. These mutants also showed decreased proliferation during hair cell regeneration but did not show consistent changes in support cell number or proliferation during hair cell development. These results show that the loss of hair cells seen following disruption of cilia through either mutations in anterograde or retrograde IFT genes appears to be due to impacts on hair cell survival but not necessarily development in the zebrafish lateral line.
Collapse
Affiliation(s)
- Hope Boldizar
- Neuroscience Program, Lafayette College, Easton, PA 18042, USA
| | - Amanda Friedman
- Neuroscience Program, Lafayette College, Easton, PA 18042, USA
| | - Tess Stanley
- Neuroscience Program, Lafayette College, Easton, PA 18042, USA
| | - María Padilla
- Biology Department, Lafayette College, Easton, PA 18042, USA
| | | | - Arielle Sclar
- Neuroscience Program, Lafayette College, Easton, PA 18042, USA
| | | |
Collapse
|
2
|
Lewis TR, Castillo CM, Klementieva NV, Hsu Y, Hao Y, Spencer WJ, Drack AV, Pazour GJ, Arshavsky VY. Contribution of intraflagellar transport to compartmentalization and maintenance of the photoreceptor cell. Proc Natl Acad Sci U S A 2024; 121:e2408551121. [PMID: 39145934 PMCID: PMC11348033 DOI: 10.1073/pnas.2408551121] [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: 04/29/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
The first steps of vision take place in the ciliary outer segment compartment of photoreceptor cells. The protein composition of outer segments is uniquely suited to perform this function. The most abundant among these proteins is the visual pigment, rhodopsin, whose outer segment trafficking involves intraflagellar transport (IFT). Here, we report three major findings from the analysis of mice in which ciliary transport was acutely impaired by conditional knockouts of IFT-B subunits. First, we demonstrate the existence of a sorting mechanism whereby mislocalized rhodopsin is recruited to and concentrated in extracellular vesicles prior to their release, presumably to protect the cell from adverse effects of protein mislocalization. Second, reducing rhodopsin expression significantly delays photoreceptor degeneration caused by IFT disruption, suggesting that controlling rhodopsin levels may be an effective therapy for some cases of retinal degenerative disease. Last, the loss of IFT-B subunits does not recapitulate a phenotype observed in mutants of the BBSome (another ciliary transport protein complex relying on IFT) in which non-ciliary proteins accumulate in the outer segment. Whereas it is widely thought that the role of the BBSome is to primarily participate in ciliary transport, our data suggest that the BBSome has another major function independent of IFT and possibly related to maintaining the diffusion barrier of the ciliary transition zone.
Collapse
Affiliation(s)
- Tylor R. Lewis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC27710
| | - Carson M. Castillo
- Department of Ophthalmology, Duke University Medical Center, Durham, NC27710
| | | | - Ying Hsu
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA52242
| | - Ying Hao
- Department of Ophthalmology, Duke University Medical Center, Durham, NC27710
| | - William J. Spencer
- Department of Ophthalmology, Duke University Medical Center, Durham, NC27710
| | - Arlene V. Drack
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA52242
| | - Gregory J. Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA01605
| | - Vadim Y. Arshavsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC27710
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC27710
| |
Collapse
|
3
|
Chiang HJ, Nishiwaki Y, Chiang WC, Masai I. Male germ cell-associated kinase is required for axoneme formation during ciliogenesis in zebrafish photoreceptors. Dis Model Mech 2024; 17:dmm050618. [PMID: 38813692 PMCID: PMC11273301 DOI: 10.1242/dmm.050618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 05/16/2024] [Indexed: 05/31/2024] Open
Abstract
Vertebrate photoreceptors are highly specialized retinal neurons that have cilium-derived membrane organelles called outer segments, which function as platforms for phototransduction. Male germ cell-associated kinase (MAK) is a cilium-associated serine/threonine kinase, and its genetic mutation causes photoreceptor degeneration in mice and retinitis pigmentosa in humans. However, the role of MAK in photoreceptors is not fully understood. Here, we report that zebrafish mak mutants show rapid photoreceptor degeneration during embryonic development. In mak mutants, both cone and rod photoreceptors completely lacked outer segments and underwent apoptosis. Interestingly, zebrafish mak mutants failed to generate axonemes during photoreceptor ciliogenesis, whereas basal bodies were specified. These data suggest that Mak contributes to axoneme development in zebrafish, in contrast to mouse Mak mutants, which have elongated photoreceptor axonemes. Furthermore, the kinase activity of Mak was found to be critical in ciliary axoneme development and photoreceptor survival. Thus, Mak is required for ciliogenesis and outer segment formation in zebrafish photoreceptors to ensure intracellular protein transport and photoreceptor survival.
Collapse
Affiliation(s)
- Hung-Ju Chiang
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Tancha, Okinawa 904-0495, Japan
| | - Yuko Nishiwaki
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Tancha, Okinawa 904-0495, Japan
| | - Wei-Chieh Chiang
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Tancha, Okinawa 904-0495, Japan
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Tancha, Okinawa 904-0495, Japan
| |
Collapse
|
4
|
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
|
5
|
Radhakrishnan R, Dronamraju VR, Leung M, Gruesen A, Solanki AK, Walterhouse S, Roehrich H, Song G, da Costa Monsanto R, Cureoglu S, Martin R, Kondkar AA, van Kuijk FJ, Montezuma SR, Knöelker HJ, Hufnagel RB, Lobo GP. The role of motor proteins in photoreceptor protein transport and visual function. Ophthalmic Genet 2022; 43:285-300. [PMID: 35470760 DOI: 10.1080/13816810.2022.2062391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Rods and cones are photoreceptor neurons in the retina that are required for visual sensation in vertebrates, wherein the perception of vision is initiated when these neurons respond to photons in the light stimuli. The photoreceptor cell is structurally studied as outer segments (OS) and inner segments (IS) where proper protein sorting, localization, and compartmentalization are critical for phototransduction, visual function, and survival. In human retinal diseases, improper protein transport to the OS or mislocalization of proteins to the IS and other cellular compartments could lead to impaired visual responses and photoreceptor cell degeneration that ultimately cause loss of visual function. RESULTS Therefore, studying and identifying mechanisms involved in facilitating and maintaining proper protein transport in photoreceptor cells would help our understanding of pathologies involving retinal cell degeneration in inherited retinal dystrophies, age-related macular degeneration, and Usher Syndrome. CONCLUSIONS Our mini-review will discuss mechanisms of protein transport within photoreceptors and introduce a novel role for an unconventional motor protein, MYO1C, in actin-based motor transport of the visual chromophore Rhodopsin to the OS, in support of phototransduction and visual function.
Collapse
Affiliation(s)
- Rakesh Radhakrishnan
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Venkateshwara R Dronamraju
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Matthias Leung
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Andrew Gruesen
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ashish K Solanki
- Department of Medicine, Drug Discovery Building, Medical University of South Carolina, South Carolina, USA
| | - Stephen Walterhouse
- Department of Medicine, Drug Discovery Building, Medical University of South Carolina, South Carolina, USA
| | - Heidi Roehrich
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Grace Song
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rafael da Costa Monsanto
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sebahattin Cureoglu
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - René Martin
- Faculty of Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Altaf A Kondkar
- Department of Ophthalmology.,Glaucoma Research Chair in Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Frederik J van Kuijk
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sandra R Montezuma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Robert B Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Glenn P Lobo
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Medicine, Drug Discovery Building, Medical University of South Carolina, South Carolina, USA.,Department of Ophthalmology, Medical University of South Carolina, South Carolina, USA
| |
Collapse
|
6
|
Comparison of Ciliary Targeting of Two Rhodopsin-Like GPCRs: Role of C-Terminal Localization Sequences in Relation to Cilium Type. J Neurosci 2021; 41:7514-7531. [PMID: 34301828 PMCID: PMC8425976 DOI: 10.1523/jneurosci.0357-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/20/2021] [Accepted: 06/16/2021] [Indexed: 11/21/2022] Open
Abstract
Primary cilia exhibit a distinct complement of proteins, including G-protein-coupled receptors (GPCRs) that mediate sensory and developmental signals. The localization of GPCRs to the ciliary membrane involves ciliary localization sequences (CLSs), but it is not known how CLSs might relate to cilium type. Here, we studied the localization of two rhodopsin (RHO)-like GPCRs, somatostatin receptor (SSTR3) and RHO, in three types of cilia, from inner medullary collecting duct (IMCD3) cells, hTERT-RPE1 cells (possessing pocket cilia), and rod photoreceptors (whose cilia grow into elaborate phototransductive outer segments). SSTR3 was localized specifically to all three types of cilia, whereas RHO showed more selectivity for the photoreceptor cilium. Focusing on C-terminal CLSs, we characterized a novel CLS in the SSTR3 C terminus, which was required for the robust ciliary localization of SSTR3. Replacing the C terminus of RHO with this SSTR3 CLS-enhanced ciliary localization, compared with full-length RHO in IMCD3 and hTERT-RPE1 cells. Addition of the SSTR3 CLS to the single transmembrane protein CD8A enabled ciliary localization. In hTERT-RPE1 cells, a partial SSTR3 CLS added to CD8A effected specific localization to the periciliary (pocket) membrane, demonstrating C-terminal localization sequence targeting to this domain. Using retinas from mice, including both sexes, we show that deletion of the C terminus of RHO reduced the rod outer segment localization and that addition of the SSTR3 C-terminal CLS to the truncated RHO partly rescued this mislocalization. Overall, the study details elements of the different C termini of SSTR3 and RHO that are major effectors in determining specificity of cilium (or pericilium) localization among different types of cilia.SIGNIFICANCE STATEMENT The localization of G-protein-coupled receptors to primary cilia is key to many types of signal transduction. After characterizing a novel C-terminal CLS in SSTR3, we investigated how SSTR3 and RHO localization to the cilium relates to C-terminal CLSs and to cilium type. We found that the SSTR3 C-terminal CLS was effective in three different types of cilia, but the RHO C terminus showed a clear localization preference for the highly elaborate photoreceptor cilium. When added to CD8A, part of the SSTR3 CLS promoted specific periciliary membrane localization in hTERT-RPE1 cells, demonstrating an effective CLS for this domain. Thus, we demonstrate that elements of the C termini of SSTR3 and RHO determine different localization patterns among different types of cilia.
Collapse
|
7
|
Zhu P, Xu J, Wang Y, Zhao C. Loss of Ift74 Leads to Slow Photoreceptor Degeneration and Ciliogenesis Defects in Zebrafish. Int J Mol Sci 2021; 22:ijms22179329. [PMID: 34502236 PMCID: PMC8431285 DOI: 10.3390/ijms22179329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 12/18/2022] Open
Abstract
Cilia are microtubule-based structures projecting from the cell surface that perform diverse biological functions. Ciliary defects can cause a wide range of genetic disorders known collectively as ciliopathies. Intraflagellar transport (IFT) proteins are essential for the assembly and maintenance of cilia by transporting proteins along the axoneme. Here, we report a lack of Ift74, a core IFT-B protein, leading to ciliogenesis defects in multiple organs during early zebrafish development. Unlike rapid photoreceptor cell death in other ift-b mutants, the photoreceptors of ift74 mutants exhibited a slow degeneration process. Further experiments demonstrated that the connecting cilia of ift74 mutants were initially formed but failed to maintain, which resulted in slow opsin transport efficiency and eventually led to photoreceptor cell death. We also showed that the large amount of maternal ift74 transcripts deposited in zebrafish eggs account for the main reason of slow photoreceptor degeneration in the mutants. Together, our data suggested Ift74 is critical for ciliogenesis and that Ift proteins play variable roles in different types of cilia during early zebrafish development. To our knowledge, this is the first study to show ift-b mutant that displays slow photoreceptor degeneration in zebrafish.
Collapse
Affiliation(s)
- Panpan Zhu
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (P.Z.); (J.X.); (Y.W.)
| | - Jingjin Xu
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (P.Z.); (J.X.); (Y.W.)
| | - Yadong Wang
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (P.Z.); (J.X.); (Y.W.)
| | - Chengtian Zhao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (P.Z.); (J.X.); (Y.W.)
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
- Sars-Fang Centre, Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
- Correspondence:
| |
Collapse
|
8
|
Ebke LA, Sinha S, Pauer GJT, Hagstrom SA. Photoreceptor Compartment-Specific TULP1 Interactomes. Int J Mol Sci 2021; 22:ijms22158066. [PMID: 34360830 PMCID: PMC8348715 DOI: 10.3390/ijms22158066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/06/2021] [Accepted: 07/12/2021] [Indexed: 12/16/2022] Open
Abstract
Photoreceptors are highly compartmentalized cells with large amounts of proteins synthesized in the inner segment (IS) and transported to the outer segment (OS) and synaptic terminal. Tulp1 is a photoreceptor-specific protein localized to the IS and synapse. In the absence of Tulp1, several OS-specific proteins are mislocalized and synaptic vesicle recycling is impaired. To better understand the involvement of Tulp1 in protein trafficking, our approach in the current study was to physically isolate Tulp1-containing photoreceptor compartments by serial tangential sectioning of retinas and to identify compartment-specific Tulp1 binding partners by immunoprecipitation followed by liquid chromatography tandem mass spectrometry. Our results indicate that Tulp1 has two distinct interactomes. We report the identification of: (1) an IS-specific interaction between Tulp1 and the motor protein Kinesin family member 3a (Kif3a), (2) a synaptic-specific interaction between Tulp1 and the scaffold protein Ribeye, and (3) an interaction between Tulp1 and the cytoskeletal protein microtubule-associated protein 1B (MAP1B) in both compartments. Immunolocalization studies in the wild-type retina indicate that Tulp1 and its binding partners co-localize to their respective compartments. Our observations are compatible with Tulp1 functioning in protein trafficking in multiple photoreceptor compartments, likely as an adapter molecule linking vesicles to molecular motors and the cytoskeletal scaffold.
Collapse
Affiliation(s)
- Lindsey A. Ebke
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (L.A.E.); (S.S.); (G.J.T.P.)
| | - Satyabrata Sinha
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (L.A.E.); (S.S.); (G.J.T.P.)
| | - Gayle J. T. Pauer
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (L.A.E.); (S.S.); (G.J.T.P.)
| | - Stephanie A. Hagstrom
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (L.A.E.); (S.S.); (G.J.T.P.)
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
9
|
Płochocka AZ, Ramirez Moreno M, Davie AM, Bulgakova NA, Chumakova L. Robustness of the microtubule network self-organization in epithelia. eLife 2021; 10:59529. [PMID: 33522481 PMCID: PMC7920549 DOI: 10.7554/elife.59529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 01/26/2021] [Indexed: 12/24/2022] Open
Abstract
Robustness of biological systems is crucial for their survival, however, for many systems its origin is an open question. Here, we analyze one subcellular level system, the microtubule cytoskeleton. Microtubules self-organize into a network, along which cellular components are delivered to their biologically relevant locations. While the dynamics of individual microtubules is sensitive to the organism’s environment and genetics, a similar sensitivity of the overall network would result in pathologies. Our large-scale stochastic simulations show that the self-organization of microtubule networks is robust in a wide parameter range in individual cells. We confirm this robustness in vivo on the tissue-scale using genetic manipulations of Drosophila epithelial cells. Finally, our minimal mathematical model shows that the origin of robustness is the separation of time-scales in microtubule dynamics rates. Altogether, we demonstrate that the tissue-scale self-organization of a microtubule network depends only on cell geometry and the distribution of the microtubule minus-ends.
Collapse
Affiliation(s)
| | - Miguel Ramirez Moreno
- Department of Biomedical Science, The University of Sheffield, Sheffield, United Kingdom
| | - Alexander M Davie
- Maxwell Institute for Mathematical Sciences, School of Mathematics, Edinburgh University, Edinburgh, United Kingdom
| | - Natalia A Bulgakova
- Department of Biomedical Science, The University of Sheffield, Sheffield, United Kingdom
| | - Lyubov Chumakova
- Maxwell Institute for Mathematical Sciences, School of Mathematics, Edinburgh University, Edinburgh, United Kingdom
| |
Collapse
|
10
|
Burger CA, Albrecht NE, Jiang D, Liang JH, Poché RA, Samuel MA. LKB1 and AMPK instruct cone nuclear position to modify visual function. Cell Rep 2021; 34:108698. [PMID: 33535040 PMCID: PMC7906279 DOI: 10.1016/j.celrep.2021.108698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/06/2020] [Accepted: 01/06/2021] [Indexed: 12/17/2022] Open
Abstract
Cone photoreceptors detect light and are responsible for color vision. These cells display a distinct polarized morphology where nuclei are precisely aligned in the apical retina. However, little is known about the mechanisms involved in cone nuclear positioning or the impact of this organization on retina function. We show that the serine/threonine kinase LKB1 and one of its substrates, AMPK, regulate cone nuclear positioning. In the absence of either molecule, cone nuclei are misplaced along the axon, resulting in altered nuclear lamination. LKB1 is required specifically in cones to mediate this process, and disruptions in nuclear alignment result in reduced cone function. Together, these results identify molecular determinants of cone nuclear position and indicate that cone nuclear position alignment enables proper visual function.
Collapse
Affiliation(s)
- Courtney A Burger
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nicholas E Albrecht
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Danye Jiang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Justine H Liang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ross A Poché
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Melanie A Samuel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
11
|
Meschede IP, Burgoyne T, Tolmachova T, Seabra MC, Futter CE. Chronically shortened rod outer segments accompany photoreceptor cell death in Choroideremia. PLoS One 2020; 15:e0242284. [PMID: 33201897 PMCID: PMC7671558 DOI: 10.1371/journal.pone.0242284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/29/2020] [Indexed: 01/28/2023] Open
Abstract
X-linked choroideremia (CHM) is a disease characterized by gradual retinal degeneration caused by loss of the Rab Escort Protein, REP1. Despite partial compensation by REP2 the disease is characterized by prenylation defects in multiple members of the Rab protein family that are master regulators of membrane traffic. Remarkably, the eye is the only organ affected in CHM patients, possibly because of the huge membrane traffic burden of the post mitotic photoreceptors, which synthesise outer segments, and the adjacent retinal pigment epithelium that degrades the spent portions each day. In this study, we aimed to identify defects in membrane traffic that might lead to photoreceptor cell death in CHM. In a heterozygous null female mouse model of CHM (Chmnull/WT), degeneration of the photoreceptor layer was clearly evident from increased numbers of TUNEL positive cells compared to age matched controls, small numbers of cells exhibiting signs of mitochondrial stress and greatly increased microglial infiltration. However, most rod photoreceptors exhibited remarkably normal morphology with well-formed outer segments and no discernible accumulation of transport vesicles in the inner segment. The major evidence of membrane trafficking defects was a shortening of rod outer segments that was evident at 2 months of age but remained constant over the period during which the cells die. A decrease in rhodopsin density found in the outer segment may underlie the outer segment shortening but does not lead to rhodopsin accumulation in the inner segment. Our data argue against defects in rhodopsin transport or outer segment renewal as triggers of cell death in CHM.
Collapse
Affiliation(s)
| | | | | | - Miguel C. Seabra
- UCL Institute of Ophthalmology, London, United Kingdom
- Imperial College London, London, United Kingdom
- CEDOC, NOVA Universidade Nova de Lisboa, Lisbon, Portugal
| | | |
Collapse
|
12
|
Chen HY, Kelley RA, Li T, Swaroop A. Primary cilia biogenesis and associated retinal ciliopathies. Semin Cell Dev Biol 2020; 110:70-88. [PMID: 32747192 PMCID: PMC7855621 DOI: 10.1016/j.semcdb.2020.07.013] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/14/2020] [Accepted: 07/18/2020] [Indexed: 12/19/2022]
Abstract
The primary cilium is a ubiquitous microtubule-based organelle that senses external environment and modulates diverse signaling pathways in different cell types and tissues. The cilium originates from the mother centriole through a complex set of cellular events requiring hundreds of distinct components. Aberrant ciliogenesis or ciliary transport leads to a broad spectrum of clinical entities with overlapping yet highly variable phenotypes, collectively called ciliopathies, which include sensory defects and syndromic disorders with multi-organ pathologies. For efficient light detection, photoreceptors in the retina elaborate a modified cilium known as the outer segment, which is packed with membranous discs enriched for components of the phototransduction machinery. Retinopathy phenotype involves dysfunction and/or degeneration of the light sensing photoreceptors and is highly penetrant in ciliopathies. This review will discuss primary cilia biogenesis and ciliopathies, with a focus on the retina, and the role of CP110-CEP290-CC2D2A network. We will also explore how recent technologies can advance our understanding of cilia biology and discuss new paradigms for developing potential therapies of retinal ciliopathies.
Collapse
Affiliation(s)
- Holly Y Chen
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA.
| | - Ryan A Kelley
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA
| | - Tiansen Li
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
13
|
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: 2.2] [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
|
14
|
Ojeda Naharros I, Cristian FB, Zang J, Gesemann M, Ingham PW, Neuhauss SCF, Bachmann-Gagescu R. The ciliopathy protein TALPID3/KIAA0586 acts upstream of Rab8 activation in zebrafish photoreceptor outer segment formation and maintenance. Sci Rep 2018; 8:2211. [PMID: 29396404 PMCID: PMC5797153 DOI: 10.1038/s41598-018-20489-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/19/2018] [Indexed: 12/12/2022] Open
Abstract
Ciliopathies are human disorders caused by dysfunction of primary cilia, ubiquitous microtubule-based organelles involved in signal transduction. Cilia are anchored inside the cell through basal bodies (BBs), modified centrioles also acting as microtubule-organization centers. Photoreceptors (PRs) are sensory neurons, whose primary cilium forms a highly specialized compartment called the outer segment (OS) responsible for sensing incoming light. Thus, ciliopathies often present with retinal degeneration. Mutations in KIAA0586/TALPID3 (TA3) cause Joubert syndrome, in which 30% of affected individuals develop retinal involvement. To elucidate the function of TALPID3 in PRs, we studied talpid3 zebrafish mutants and identified a progressive retinal degeneration phenotype. The majority of PRs lack OS development due to defects in BB positioning and docking at the apical cell surface. Intracellular accumulation of the photopigment opsin leads to PR cell death of moderate severity. Electroretinograms demonstrate severe visual impairement. A small subset of PRs display normally docked BBs and extended OSs through rescue by maternally-deposited Talpid3. While localization of the small GTPase Rab8a, which plays an important role in BB docking, appears unaffected in talpid3-/- PRs, overexpression of constitutively active Rab8a rescues OS formation, indicating that the role of Ta3 in early ciliogenesis lies upstream of Rab8a activation in PRs.
Collapse
Affiliation(s)
- Irene Ojeda Naharros
- Institute for Molecular Life Sciences, University of Zurich, 8057, Zurich, Switzerland
| | - Flavia B Cristian
- Institute for Molecular Life Sciences, University of Zurich, 8057, Zurich, Switzerland
- Department of Human Molecular Genetics, Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Jingjing Zang
- Institute for Molecular Life Sciences, University of Zurich, 8057, Zurich, Switzerland
| | - Matthias Gesemann
- Institute for Molecular Life Sciences, University of Zurich, 8057, Zurich, Switzerland
| | - Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University, 639798, Singapore, Singapore
| | - Stephan C F Neuhauss
- Institute for Molecular Life Sciences, University of Zurich, 8057, Zurich, Switzerland
| | - Ruxandra Bachmann-Gagescu
- Institute for Molecular Life Sciences, University of Zurich, 8057, Zurich, Switzerland.
- Institute for Medical Genetics, University of Zurich, 8952, Schlieren, Switzerland.
| |
Collapse
|
15
|
Rpgrip1 is required for rod outer segment development and ciliary protein trafficking in zebrafish. Sci Rep 2017; 7:16881. [PMID: 29203866 PMCID: PMC5715152 DOI: 10.1038/s41598-017-12838-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 09/14/2017] [Indexed: 12/23/2022] Open
Abstract
Mutations in the RPGR-interacting protein 1 (RPGRIP1) gene cause recessive Leber congenital amaurosis (LCA), juvenile retinitis pigmentosa (RP) and cone-rod dystrophy. RPGRIP1 interacts with other retinal disease-causing proteins and has been proposed to have a role in ciliary protein transport; however, its function remains elusive. Here, we describe a new zebrafish model carrying a nonsense mutation in the rpgrip1 gene. Rpgrip1homozygous mutants do not form rod outer segments and display mislocalization of rhodopsin, suggesting a role for RPGRIP1 in rhodopsin-bearing vesicle trafficking. Furthermore, Rab8, the key regulator of rhodopsin ciliary trafficking, was mislocalized in photoreceptor cells of rpgrip1 mutants. The degeneration of rod cells is early onset, followed by the death of cone cells. These phenotypes are similar to that observed in LCA and juvenile RP patients. Our data indicate RPGRIP1 is necessary for rod outer segment development through regulating ciliary protein trafficking. The rpgrip1 mutant zebrafish may provide a platform for developing therapeutic treatments for RP patients.
Collapse
|
16
|
Veleri S, Nellissery J, Mishra B, Manjunath SH, Brooks MJ, Dong L, Nagashima K, Qian H, Gao C, Sergeev YV, Huang XF, Qu J, Lu F, Cideciyan AV, Li T, Jin ZB, Fariss RN, Ratnapriya R, Jacobson SG, Swaroop A. REEP6 mediates trafficking of a subset of Clathrin-coated vesicles and is critical for rod photoreceptor function and survival. Hum Mol Genet 2017; 26:2218-2230. [PMID: 28369466 PMCID: PMC5458339 DOI: 10.1093/hmg/ddx111] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/16/2017] [Indexed: 01/20/2023] Open
Abstract
In retinal photoreceptors, vectorial transport of cargo is critical for transduction of visual signals, and defects in intracellular trafficking can lead to photoreceptor degeneration and vision impairment. Molecular signatures associated with routing of transport vesicles in photoreceptors are poorly understood. We previously reported the identification of a novel rod photoreceptor specific isoform of Receptor Expression Enhancing Protein (REEP) 6, which belongs to a family of proteins involved in intracellular transport of receptors to the plasma membrane. Here we show that loss of REEP6 in mice (Reep6−/−) results in progressive retinal degeneration. Rod photoreceptor dysfunction is observed in Reep6−/− mice as early as one month of age and associated with aberrant accumulation of vacuole-like structures at the apical inner segment and reduction in selected rod phototransduction proteins. We demonstrate that REEP6 is detected in a subset of Clathrin-coated vesicles and interacts with the t-SNARE, Syntaxin3. In concordance with the rod degeneration phenotype in Reep6−/− mice, whole exome sequencing identified homozygous REEP6-E75K mutation in two retinitis pigmentosa families of different ethnicities. Our studies suggest a critical function of REEP6 in trafficking of cargo via a subset of Clathrin-coated vesicles to selected membrane sites in retinal rod photoreceptors.
Collapse
Affiliation(s)
- Shobi Veleri
- Neurobiology Neurodegeneration and Repair Laboratory
| | | | | | | | | | - Lijin Dong
- Genetic Engineering Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kunio Nagashima
- Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Haohua Qian
- Visual Function Core, 5Biological Imaging Core
| | - Chun Gao
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yuri V Sergeev
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiu-Feng Huang
- The Eye Hospital of Wenzhou Medical University, Wenzhou 325027, China and
| | - Jia Qu
- The Eye Hospital of Wenzhou Medical University, Wenzhou 325027, China and
| | - Fan Lu
- The Eye Hospital of Wenzhou Medical University, Wenzhou 325027, China and
| | - Artur V Cideciyan
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tiansen Li
- Neurobiology Neurodegeneration and Repair Laboratory
| | - Zi-Bing Jin
- The Eye Hospital of Wenzhou Medical University, Wenzhou 325027, China and
| | - Robert N Fariss
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Samuel G Jacobson
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anand Swaroop
- Neurobiology Neurodegeneration and Repair Laboratory
| |
Collapse
|
17
|
Feng D, Chen Z, Yang K, Miao S, Xu B, Kang Y, Xie H, Zhao C. The cytoplasmic tail of rhodopsin triggers rapid rod degeneration in kinesin-2 mutants. J Biol Chem 2017; 292:17375-17386. [PMID: 28855254 DOI: 10.1074/jbc.m117.784017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 08/24/2017] [Indexed: 11/06/2022] Open
Abstract
Photoreceptor degeneration can lead to blindness and represents the most common form of neural degenerative disease worldwide. Although many genes involved in photoreceptor degeneration have been identified, the underlying mechanisms remain to be elucidated. Here we examined photoreceptor development in zebrafish kif3a and kif3b mutants, which affect two subunits of the kinesin-2 complex. In both mutants, rods degenerated quickly, whereas cones underwent a slow degeneration process. Notably, the photoreceptor defects were considerably more severe in kif3a mutants than in kif3b mutants. In the cone photoreceptors of kif3a mutants, opsin proteins accumulated in the apical region and formed abnormal membrane structures. In contrast, rhodopsins were enriched in the rod cell body membrane and represented the primary reason for rapid rod degeneration in these mutants. Moreover, removal of the cytoplasmic tail of rhodopsin to reduce its function, but not decreasing rhodopsin expression levels, prevented rod degeneration in both kif3a and kif3b mutants. Of note, overexpression of full-length rhodopsin or its cytoplasmic tail domain, but not of rhodopsin lacking the cytoplasmic tail, exacerbated rod degeneration in kif3a mutants, implying an important role of the cytoplasmic tail in rod degeneration. Finally, we showed that the cytoplasmic tail of rhodopsin might trigger rod degeneration through activating the downstream calcium signaling pathway, as drug treatment with inhibitors of intracellular calcium release prevented rod degeneration in kif3a mutants. Our results demonstrate a previously unknown function of the rhodopsin cytoplasmic domain during opsin-triggered photoreceptor degeneration and may open up new avenues for managing this disease.
Collapse
Affiliation(s)
- Dong Feng
- From the Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Zhe Chen
- From the Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Kuang Yang
- From the Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Shanshan Miao
- From the Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Bolin Xu
- From the Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Yunsi Kang
- From the Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Haibo Xie
- From the Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China
| | - Chengtian Zhao
- From the Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China, .,the Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China, and.,the Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
18
|
Mitchison HM, Valente EM. Motile and non-motile cilia in human pathology: from function to phenotypes. J Pathol 2017; 241:294-309. [PMID: 27859258 DOI: 10.1002/path.4843] [Citation(s) in RCA: 292] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/03/2016] [Accepted: 11/04/2016] [Indexed: 12/13/2022]
Abstract
Ciliopathies are inherited human disorders caused by both motile and non-motile cilia dysfunction that form an important and rapidly expanding disease category. Ciliopathies are complex conditions to diagnose, being multisystem disorders characterized by extensive genetic heterogeneity and clinical variability with high levels of lethality. There is marked phenotypic overlap among distinct ciliopathy syndromes that presents a major challenge for their recognition, diagnosis, and clinical management, in addition to posing an on-going task to develop the most appropriate family counselling. The impact of next-generation sequencing and high-throughput technologies in the last decade has significantly improved our understanding of the biological basis of ciliopathy disorders, enhancing our ability to determine the possible reasons for the extensive overlap in their symptoms and genetic aetiologies. Here, we review the diverse functions of cilia in human health and disease and discuss a growing shift away from the classical clinical definitions of ciliopathy syndromes to a more functional categorization. This approach arises from our improved understanding of this unique organelle, revealed through new genetic and cell biological insights into the discrete functioning of subcompartments of the cilium (basal body, transition zone, intraflagellar transport, motility). Mutations affecting these distinct ciliary protein modules can confer different genetic diseases and new clinical classifications are possible to define, according to the nature and extent of organ involvement. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Hannah M Mitchison
- Genetics and Genomic Medicine Programme, University College London, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Enza Maria Valente
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy.,Neurogenetics Unit, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, 00143, Rome, Italy
| |
Collapse
|
19
|
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: 1.0] [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
|
20
|
Nemet I, Ropelewski P, Imanishi Y. Applications of phototransformable fluorescent proteins for tracking the dynamics of cellular components. Photochem Photobiol Sci 2016; 14:1787-806. [PMID: 26345171 DOI: 10.1039/c5pp00174a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the past few decades, fluorescent proteins have revolutionized the field of cell biology. Phototransformable fluorescent proteins are capable of changing their excitation and emission spectra after being exposed to specific wavelength(s) of light. The majority of phototransformable fluorescent proteins have originated from marine organisms. Genetic engineering of these proteins has made available many choices for different colors, modes of conversion, and other biophysical properties. Their phototransformative property has allowed the highlighting and tracking of subpopulations of cells, organelles, and proteins in living systems. Furthermore, phototransformable fluorescent proteins have offered new methods for superresolution fluorescence microscopy and optogenetics manipulation of proteins. One of the major advantages of phototransformable fluorescent proteins is their applicability for visualizing newly synthesized proteins that are en route to their final destinations. In this paper, we will discuss the biological applications of phototransformable fluorescent proteins with special emphasis on the application of tracking membrane proteins in vertebrate photoreceptor cells.
Collapse
Affiliation(s)
- Ina Nemet
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | | | |
Collapse
|
21
|
Goldberg AFX, Moritz OL, Williams DS. Molecular basis for photoreceptor outer segment architecture. Prog Retin Eye Res 2016; 55:52-81. [PMID: 27260426 DOI: 10.1016/j.preteyeres.2016.05.003] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/27/2016] [Accepted: 05/29/2016] [Indexed: 01/11/2023]
Abstract
To serve vision, vertebrate rod and cone photoreceptors must detect photons, convert the light stimuli into cellular signals, and then convey the encoded information to downstream neurons. Rods and cones are sensory neurons that each rely on specialized ciliary organelles to detect light. These organelles, called outer segments, possess elaborate architectures that include many hundreds of light-sensitive membranous disks arrayed one atop another in precise register. These stacked disks capture light and initiate the chain of molecular and cellular events that underlie normal vision. Outer segment organization is challenged by an inherently dynamic nature; these organelles are subject to a renewal process that replaces a significant fraction of their disks (up to ∼10%) on a daily basis. In addition, a broad range of environmental and genetic insults can disrupt outer segment morphology to impair photoreceptor function and viability. In this chapter, we survey the major progress that has been made for understanding the molecular basis of outer segment architecture. We also discuss key aspects of organelle lipid and protein composition, and highlight distributions, interactions, and potential structural functions of key OS-resident molecules, including: kinesin-2, actin, RP1, prominin-1, protocadherin 21, peripherin-2/rds, rom-1, glutamic acid-rich proteins, and rhodopsin. Finally, we identify key knowledge gaps and challenges that remain for understanding how normal outer segment architecture is established and maintained.
Collapse
Affiliation(s)
- Andrew F X Goldberg
- Eye Research Institute, Oakland University, 417 Dodge Hall, Rochester, MI, 48309, USA.
| | - Orson L Moritz
- Department of Ophthalmology & Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - David S Williams
- Department of Ophthalmology and Jules Stein Eye Institute, Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA
| |
Collapse
|
22
|
Jiang M, Esteve-Rudd J, Lopes VS, Diemer T, Lillo C, Rump A, Williams DS. Microtubule motors transport phagosomes in the RPE, and lack of KLC1 leads to AMD-like pathogenesis. J Cell Biol 2015; 210:595-611. [PMID: 26261180 PMCID: PMC4539993 DOI: 10.1083/jcb.201410112] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 07/02/2015] [Indexed: 11/22/2022] Open
Abstract
The degradation of phagosomes, derived from the ingestion of photoreceptor outer segment (POS) disk membranes, is a major role of the retinal pigment epithelium (RPE). Here, POS phagosomes were observed to associate with myosin-7a, and then kinesin-1, as they moved from the apical region of the RPE. Live-cell imaging showed that the phagosomes moved bidirectionally along microtubules in RPE cells, with kinesin-1 light chain 1 (KLC1) remaining associated in both directions and during pauses. Lack of KLC1 did not inhibit phagosome speed, but run length was decreased, and phagosome localization and degradation were impaired. In old mice, lack of KLC1 resulted in RPE pathogenesis that was strikingly comparable to aspects of age-related macular degeneration (AMD), with an excessive accumulation of RPE and sub-RPE deposits, as well as oxidative and inflammatory stress responses. These results elucidate mechanisms of POS phagosome transport in relation to degradation, and demonstrate that defective microtubule motor transport in the RPE leads to phenotypes associated with AMD.
Collapse
Affiliation(s)
- Mei Jiang
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095
| | - Julian Esteve-Rudd
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095
| | - Vanda S Lopes
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095 Centre of Ophthalmology, Institute for Biomedical Imaging and Life Sciences, Faculty of Medicine, University Coimbra, 3000-548 Coimbra, Portugal
| | - Tanja Diemer
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095
| | - Concepción Lillo
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093
| | - Agrani Rump
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095
| | - David S Williams
- Department of Ophthalmology, Jules Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA 90095 Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095 Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095 Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093 Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
23
|
Rhodopsin Trafficking and Mistrafficking: Signals, Molecular Components, and Mechanisms. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:39-71. [PMID: 26055054 DOI: 10.1016/bs.pmbts.2015.02.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Rhodopsin is a seven-transmembrane G protein-coupled receptor (GPCR) and is the main component of the photoreceptor outer segment (OS), a ciliary compartment essential for vision. Because the OSs are incapable of protein synthesis, rhodopsin must first be synthesized in the inner segments (ISs) and subsequently trafficked across the connecting cilia to the OSs where it participates in the phototransduction cascade. Rapid turnover of the OS necessitates a high rate of synthesis and efficient trafficking of rhodopsin to the cilia. This cilia-targeting mechanism is shared among other ciliary-localized GPCRs. In this review, we will discuss the process of rhodopsin trafficking from the IS to the OS beginning with the trafficking signals present on the protein. Starting from the endoplasmic reticulum and the Golgi apparatus within the IS, we will cover the molecular components assisting the biogenesis and the proper sorting. We will also review the confirmed binding and interacting partners that help target rhodopsin toward the connecting cilium as well as the cilia-localized components which direct proteins into the proper compartments of the OS. While rhodopsin is the most critical and abundant component of the photoreceptor OS, mutations in the rhodopsin gene commonly lead to its mislocalization within the photoreceptors. In addition to covering the trafficking patterns of rhodopsin, we will also review some of the most common rhodopsin mutants which cause mistrafficking and subsequent death of photoreceptors. Toward the goal of understanding the pathogenesis, three major mechanisms of aberrant trafficking as well as putative mechanisms of photoreceptor degeneration will be discussed.
Collapse
|
24
|
Retinal structure and function preservation by polysaccharides of wolfberry in a mouse model of retinal degeneration. Sci Rep 2014; 4:7601. [PMID: 25535040 PMCID: PMC4274520 DOI: 10.1038/srep07601] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 12/03/2014] [Indexed: 12/03/2022] Open
Abstract
Retinitis pigmentosa (RP) is a heterogeneous group of inherited disorders caused by mutations in a variety of genes that are mostly expressed by rod cells, which results in initial death of rod photoreceptors followed by gradual death of cone photoreceptors. RP is currently untreatable and usually leads to partial or complete blindness. Here, we explored the potential neuroprotective effects of polysaccharides of wolfberry, which are long known to possess primary beneficial properties in the eyes, on photoreceptor apoptosis in the rd10 mouse model of RP. We found that these polysaccharides provided long-term morphological and functional preservation of photoreceptors and improved visual behaviors in rd10 mice. Moreover, we demonstrated that polysaccharides exerted neuroprotective effects through antioxidant, anti-inflammatory and anti-apoptotic mechanisms. Furthermore, we identified that polysaccharides modulated inflammation and apoptosis partly through inhibition of NF-κB and HIF-1α expressions, respectively. Overall, we demonstrated the synergistic protective effects of polysaccharides in preserving photoreceptors against degeneration in rd10 mice. Our study provides rationale and scientific support on using polysaccharides of wolfberry as one supplementary treatment of RP patients in the future.
Collapse
|
25
|
Eblimit A, Nguyen TMT, Chen Y, Esteve-Rudd J, Zhong H, Letteboer S, Van Reeuwijk J, Simons DL, Ding Q, Wu KM, Li Y, Van Beersum S, Moayedi Y, Xu H, Pickard P, Wang K, Gan L, Wu SM, Williams DS, Mardon G, Roepman R, Chen R. Spata7 is a retinal ciliopathy gene critical for correct RPGRIP1 localization and protein trafficking in the retina. Hum Mol Genet 2014; 24:1584-601. [PMID: 25398945 DOI: 10.1093/hmg/ddu573] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Leber congenital amaurosis (LCA) and juvenile retinitis pigmentosa (RP) are severe hereditary diseases that causes visual impairment in infants and children. SPATA7 has recently been identified as the LCA3 and juvenile RP gene in humans, whose function in the retina remains elusive. Here, we show that SPATA7 localizes at the primary cilium of cells and at the connecting cilium (CC) of photoreceptor cells, indicating that SPATA7 is a ciliary protein. In addition, SPATA7 directly interacts with the retinitis pigmentosa GTPase regulator interacting protein 1 (RPGRIP1), a key connecting cilium protein that has also been linked to LCA. In the retina of Spata7 null mutant mice, a substantial reduction of RPGRIP1 levels at the CC of photoreceptor cells is observed, suggesting that SPATA7 is required for the stable assembly and localization of the ciliary RPGRIP1 protein complex. Furthermore, our results pinpoint a role of this complex in protein trafficking across the CC to the outer segments, as we identified that rhodopsin accumulates in the inner segments and around the nucleus of photoreceptors. This accumulation then likely triggers the apoptosis of rod photoreceptors that was observed. Loss of Spata7 function in mice indeed results in a juvenile RP-like phenotype, characterized by progressive degeneration of photoreceptor cells and a strongly decreased light response. Together, these results indicate that SPATA7 functions as a key member of a retinal ciliopathy-associated protein complex, and that apoptosis of rod photoreceptor cells triggered by protein mislocalization is likely the mechanism of disease progression in LCA3/ juvenile RP patients.
Collapse
Affiliation(s)
| | - Thanh-Minh T Nguyen
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6525, The Netherlands
| | - Yiyun Chen
- HGSC, Department of Molecular and Human Genetics
| | - Julian Esteve-Rudd
- Jules Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Hua Zhong
- Department of Pathology and Immunology
| | - Stef Letteboer
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6525, The Netherlands
| | - Jeroen Van Reeuwijk
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6525, The Netherlands
| | - David L Simons
- Department of Neuroscience and Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qian Ding
- Department of Ophthalmology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Ka Man Wu
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6525, The Netherlands
| | - Yumei Li
- HGSC, Department of Molecular and Human Genetics
| | - Sylvia Van Beersum
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6525, The Netherlands
| | | | - Huidan Xu
- HGSC, Department of Molecular and Human Genetics
| | | | - Keqing Wang
- HGSC, Department of Molecular and Human Genetics
| | - Lin Gan
- Department of Ophthalmology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Samuel M Wu
- Department of Neuroscience and Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
| | - David S Williams
- Jules Stein Eye Institute, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Graeme Mardon
- Department of Molecular and Human Genetics, Department of Pathology and Immunology, Department of Neuroscience and Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA,
| | - Ronald Roepman
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6525, The Netherlands,
| | - Rui Chen
- HGSC, Department of Molecular and Human Genetics,
| |
Collapse
|
26
|
Tian G, Lodowski KH, Lee R, Imanishi Y. Retrograde intraciliary trafficking of opsin during the maintenance of cone-shaped photoreceptor outer segments of Xenopus laevis. J Comp Neurol 2014; 522:3577-3589. [PMID: 24855015 DOI: 10.1002/cne.23630] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 05/21/2014] [Accepted: 05/21/2014] [Indexed: 12/21/2022]
Abstract
Photoreceptor outer segments (OSs) are essential for our visual perception, and take either rod or cone forms. The cell biological basis for the formation of rods is well established; however, the mechanism of cone formation is ill characterized. While Xenopus rods are called rods, they exhibit cone-shaped OSs during the early process of development. To visualize the dynamic reorganization of disk membranes, opsin and peripherin/rds were fused to a fluorescent protein, Dendra2, and expressed in early developing rod photoreceptors, in which OSs are still cone-shaped. Dendra2 is a fluorescent protein which can be converted from green to red irreversibly, and thus allows spatiotemporal labeling of proteins. Using a photoconversion technique, we found that disk membranes are assembled at the base of cone-shaped OSs. After incorporation into disks, however, Opsin-Dendra2 was also trafficked from old to new disk membranes, consistent with the hypothesis that retrograde trafficking of membrane components contributes to the larger disk membrane observed toward the base of the cone-shaped OS. Such retrograde trafficking is cargo-specific and was not observed for peripherin/rds-Dendra2. The trafficking is unlikely mediated by diffusion, since the disk membranes have a closed configuration, as evidenced by CNGA1 labeling of the plasma membrane. Consistent with retrograde trafficking, the axoneme, which potentially mediates retrograde intraflagellar trafficking, runs through the entire axis of OSs. This study provides an insight into the role of membrane reorganization in developing photoreceptor OSs, and proves that retrograde trafficking of membrane cargoes can occur there.
Collapse
Affiliation(s)
- Guilian Tian
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4965, USA
| | - Kerrie H Lodowski
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4965, USA
| | - Richard Lee
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4965, USA
| | - Yoshikazu Imanishi
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4965, USA
| |
Collapse
|
27
|
Crouse JA, Lopes VS, Sanagustin JT, Keady BT, Williams DS, Pazour GJ. Distinct functions for IFT140 and IFT20 in opsin transport. Cytoskeleton (Hoboken) 2014; 71:302-10. [PMID: 24619649 DOI: 10.1002/cm.21173] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/26/2014] [Accepted: 03/04/2014] [Indexed: 12/20/2022]
Abstract
In the vertebrate retina, light is detected by the outer segments of photoreceptor rods and cones, which are highly modified cilia. Like other cilia, outer segments have no protein synthetic capacity and depend on proteins made in the cell body for their formation and maintenance. The mechanism of transport into the outer segment is not fully understood but intraflagellar transport (IFT) is thought to be a major mechanism for moving protein from the cell body into the cilium. In the case of photoreceptor cells, the high density of receptors and the disk turnover that occurs daily necessitates much higher rates of transport than would be required in other cilia. In this work, we show that the IFT complex A protein IFT140 is required for development and maintenance of outer segments. In earlier work we found that acute deletion of Ift20 caused opsin to accumulate at the Golgi complex. In this work, we find that acute deletion of Ift140 does not cause opsin to accumulate at the Golgi complex but rather it accumulates in the plasma membrane of the inner segments. This work is a strong support of a model of opsin transport where IFT20 is involved in the movement from the Golgi complex to the base of the cilium. Then, once at the base, the opsin is carried through the connecting cilium by an IFT complex that includes IFT140. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jacquelin A Crouse
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Worcester, Massachusetts
| | | | | | | | | | | |
Collapse
|
28
|
Pardo-Diaz C, Jiggins CD. Neighboring genes shaping a single adaptive mimetic trait. Evol Dev 2014; 16:3-12. [DOI: 10.1111/ede.12058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Carolina Pardo-Diaz
- Department of Zoology; University of Cambridge; Downing Street Cambridge CB2 3EJ United Kingdom
| | - Chris D. Jiggins
- Department of Zoology; University of Cambridge; Downing Street Cambridge CB2 3EJ United Kingdom
- Smithsonian Tropical Research Institute; Balboa AA2072 Panama
| |
Collapse
|
29
|
Abstract
Rhodopsin is a cilia-specific GPCR essential for vision. Rhodopsin mislocalization is associated with blinding diseases called retinal ciliopathies. The mechanism by which rhodopsin mislocalizes in rod photoreceptor neurons is not well understood. Therefore, we investigated the roles of trafficking signals in rhodopsin mislocalization. Rhodopsin and its truncation mutants were fused to a photoconvertible fluorescent protein, Dendra2, and expressed in Xenopus laevis rod photoreceptors. Photoconversion of Dendra2 causes a color change from green to red, enabling visualization of the dynamic events associated with rhodopsin trafficking and renewal. We found that rhodopsin mislocalization is a facilitated process for which a signal located within 322-326 aa (CCGKN) is essential. An additional signal within 327-336 aa further facilitated the mislocalization. This collective mistrafficking signal confers toxicity to rhodopsin and causes mislocalization when the VXPX cilia-targeting motif is absent. We also determined that the VXPX motif neutralizes this mistrafficking signal, enhances ciliary targeting at least 10-fold, and accelerates trafficking of post-Golgi vesicular structures. In the absence of the VXPX motif, mislocalized rhodopsin is actively cleared through secretion of vesicles into the extracellular milieu. Therefore, this study unveiled the multiple roles of trafficking signals in rhodopsin localization and renewal.
Collapse
|
30
|
Wang J, Deretic D. Molecular complexes that direct rhodopsin transport to primary cilia. Prog Retin Eye Res 2013; 38:1-19. [PMID: 24135424 DOI: 10.1016/j.preteyeres.2013.08.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 08/13/2013] [Accepted: 08/19/2013] [Indexed: 11/27/2022]
Abstract
Rhodopsin is a key molecular constituent of photoreceptor cells, yet understanding of how it regulates photoreceptor membrane trafficking and biogenesis of light-sensing organelles, the rod outer segments (ROS) is only beginning to emerge. Recently identified sequence of well-orchestrated molecular interactions of rhodopsin with the functional networks of Arf and Rab GTPases at multiple stages of intracellular targeting fits well into the complex framework of the biogenesis and maintenance of primary cilia, of which the ROS is one example. This review will discuss the latest progress in dissecting the molecular complexes that coordinate rhodopsin incorporation into ciliary-targeted carriers with the recruitment and activation of membrane tethering complexes and regulators of fusion with the periciliary plasma membrane. In addition to revealing the fundamental principals of ciliary membrane renewal, recent advances also provide molecular insight into the ways by which disruptions of the exquisitely orchestrated interactions lead to cilia dysfunction and result in human retinal dystrophies and syndromic diseases that affect multiple organs, including the eyes.
Collapse
Affiliation(s)
- Jing Wang
- Department of Surgery, Division of Ophthalmology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Dusanka Deretic
- Department of Surgery, Division of Ophthalmology, University of New Mexico, Albuquerque, NM 87131, USA; Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM 87131, USA.
| |
Collapse
|
31
|
Nagel-Wolfrum K, Wolfrum U. Vesicle transport and photoreceptor death: fishing for molecular links. Dev Cell 2013; 25:435-6. [PMID: 23763944 DOI: 10.1016/j.devcel.2013.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Intracellular vesicle transport defects can induce retinal degeneration and photoreceptor cell death, but the molecular connections between these processes remains poorly understood. Reporting in Developmental Cell, Nishiwaki et al. (2013) suggest that a vesicle fusion cis-SNARE complex component translates vesicular transport defects into photoreceptor cell apoptosis.
Collapse
Affiliation(s)
- Kerstin Nagel-Wolfrum
- Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg University of Mainz, Müllerweg 6, 55099 Mainz, Germany
| | | |
Collapse
|
32
|
Scholey JM. Kinesin-2: a family of heterotrimeric and homodimeric motors with diverse intracellular transport functions. Annu Rev Cell Dev Biol 2013; 29:443-69. [PMID: 23750925 DOI: 10.1146/annurev-cellbio-101512-122335] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Kinesin-2 was first purified as a heterotrimeric, anterograde, microtubule-based motor consisting of two distinct kinesin-related subunits and a novel associated protein (KAP) that is currently best known for its role in intraflagellar transport and ciliogenesis. Subsequent work, however, has revealed diversity in the oligomeric state of different kinesin-2 motors owing to the combinatorial heterodimerization of its subunits and the coexistence of both heterotrimeric and homodimeric kinesin-2 motors in some cells. Although the functional significance of the homo- versus heteromeric organization of kinesin-2 motor subunits and the role of KAP remain uncertain, functional studies suggest that cooperation between different types of kinesin-2 motors or between kinesin-2 and a member of a different motor family can generate diverse patterns of anterograde intracellular transport. Moreover, despite being restricted to ciliated eukaryotes, kinesin-2 motors are now known to drive diverse transport events outside cilia. Here, I review the organization, assembly, phylogeny, biological functions, and motility mechanism of this diverse family of intracellular transport motors.
Collapse
Affiliation(s)
- Jonathan M Scholey
- Department of Molecular and Cell Biology, University of California, Davis, California 95616;
| |
Collapse
|
33
|
Visual arrestin interaction with clathrin adaptor AP-2 regulates photoreceptor survival in the vertebrate retina. Proc Natl Acad Sci U S A 2013; 110:9463-8. [PMID: 23690606 DOI: 10.1073/pnas.1301126110] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Arrestins bind ligand-activated, phosphorylated G protein-coupled receptors (GPCRs) and terminate the activation of G proteins. Additionally, nonvisual arrestin/GPCR complex can initiate G protein-independent intracellular signals through their ability to act as scaffolds that bring other signaling molecules to the internalized GPCR. Like nonvisual arrestins, vertebrate visual arrestin (ARR1) terminates G protein signaling from light-activated, phosphorylated GPCR, rhodopsin. Unlike nonvisual arrestins, its role as a transducer of signaling from internalized rhodopsin has not been reported in the vertebrate retina. Formation of signaling complexes with arrestins often requires recruitment of the endocytic adaptor protein, AP-2. We have previously shown that Lys296 → Glu (K296E), which is a naturally occurring rhodopsin mutation in certain humans diagnosed with autosomal dominant retinitis pigmentosa, causes toxicity through forming a stable complex with ARR1. Here we investigated whether recruitment of AP-2 by the K296E/ARR1 complex plays a role in generating the cell death signal in a transgenic mouse model of retinal degeneration. We measured the binding affinity of ARR1 for AP-2 and found that, although the affinity is much lower than that of the other arrestins, the unusually high concentration of ARR1 in rods would favor this interaction. We further demonstrate that p44, a splice variant of ARR1 that binds light-activated, phosphorylated rhodopsin but lacks the AP-2 binding motif, prevents retinal degeneration and rescues visual function in K296E mice. These results reveal a unique role of ARR1 in a G protein-independent signaling cascade in the vertebrate retina.
Collapse
|
34
|
Tiwari S, Hudson S, Gattone VH, Miller C, Chernoff EAG, Belecky-Adams TL. Meckelin 3 is necessary for photoreceptor outer segment development in rat Meckel syndrome. PLoS One 2013; 8:e59306. [PMID: 23516626 PMCID: PMC3596335 DOI: 10.1371/journal.pone.0059306] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 02/15/2013] [Indexed: 11/20/2022] Open
Abstract
Ciliopathies lead to multiorgan pathologies that include renal cysts, deafness, obesity and retinal degeneration. Retinal photoreceptors have connecting cilia joining the inner and outer segment that are responsible for transport of molecules to develop and maintain the outer segment process. The present study evaluated meckelin (MKS3) expression during outer segment genesis and determined the consequences of mutant meckelin on photoreceptor development and survival in Wistar polycystic kidney disease Wpk/Wpk rat using immunohistochemistry, analysis of cell death and electron microscopy. MKS3 was ubiquitously expressed throughout the retina at postnatal day 10 (P10) and P21. However, in the mature retina, MKS3 expression was restricted to photoreceptors and the retinal ganglion cell layer. At P10, both the wild type and homozygous Wpk mutant retina had all retinal cell types. In contrast, by P21, cells expressing rod- and cone-specific markers were fewer in number and expression of opsins appeared to be abnormally localized to the cell body. Cell death analyses were consistent with the disappearance of photoreceptor-specific markers and showed that the cells were undergoing caspase-dependent cell death. By electron microscopy, P10 photoreceptors showed rudimentary outer segments with an axoneme, but did not develop outer segment discs that were clearly present in the wild type counterpart. At p21 the mutant outer segments appeared much the same as the P10 mutant outer segments with only a short axoneme, while the wild-type controls had developed outer segments with many well-organized discs. We conclude that MKS3 is not important for formation of connecting cilium and rudimentary outer segments, but is critical for the maturation of outer segment processes.
Collapse
Affiliation(s)
- Sarika Tiwari
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
- Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Scott Hudson
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
- Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Vincent H. Gattone
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Caroline Miller
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Ellen A. G. Chernoff
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
- Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Teri L. Belecky-Adams
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
- Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
35
|
Kinesin-2 family motors in the unusual photoreceptor cilium. Vision Res 2012; 75:33-6. [PMID: 23123805 DOI: 10.1016/j.visres.2012.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/11/2012] [Accepted: 10/12/2012] [Indexed: 01/13/2023]
Abstract
This review focuses on recent advances in the understanding of kinesin-2 family motors in vertebrate photoreceptor development. Zebrafish photoreceptors develop by the 3rd day of embryogenesis, making it possible to study mutant phenotypes without the use of conditional alleles. Recent work using a zebrafish kif3b mutant allele validates the concept that the heterotrimeric kinesin II motor is generally required for ciliogenesis. In zebrafish photoreceptors, however, loss of kif3b function delays but does not block cilium formation. This is thought to occur because both kif3b or kif3c can dimerize with kif3a and function redundantly. The second ciliary kinesin thought to function in photoreceptor cells is kif17. Prior work has shown that either morpholino knockdown of this gene or the overexpression of its dominant negative form can reduce or delay photoreceptor cilium development without any evident impact on ciliogenesis in general. This has led to the idea that kif17 may play an important role only in some specialized cilium types, such the one in photoreceptor cells. In a recently identified kif17 mutant, however, photoreceptor outer segments are formed by 5 dpf and an obvious delay of outer segment formation is seen only at the earliest stage analyzed (3 dpf). This work suggests that kif17 plays a significant role mainly at an early stage of photoreceptor development. Taken together, these studies lead to an intriguing concept that as they differentiate photoreceptors alter their kinesin repertoire.
Collapse
|
36
|
Live-cell imaging evidence for the ciliary transport of rod photoreceptor opsin by heterotrimeric kinesin-2. J Neurosci 2012; 32:10587-93. [PMID: 22855808 DOI: 10.1523/jneurosci.0015-12.2012] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Primary cilia detect extracellular signals through membrane receptors and channels. The outer segment of a vertebrate photoreceptor cell represents the most elaborate of all primary cilia, containing extraordinarily large amounts of the visual receptor protein, opsin. Because of its high abundance, opsin represents a potential model system for the study of ciliary membrane receptors, including their transport. Here, we have analyzed the movement of ciliary opsin to test whether the highly conserved intraflagellar transport (IFT), as driven by heterotrimeric kinesin-2, is required. Results show that opsin can enter and move along the primary cilium of a nonphotoreceptor cell (an hTERT-RPE1 epithelial cell), suggesting that it can co-opt the basic anterograde motor system of cilia. Fluorescence recovery after photobleaching analysis of cilia of hTERT-RPE1 cells showed that the movement of ciliary opsin was comparable to that of the IFT protein, IFT88. Moreover, the movement of opsin in these cilia, as well as in cilia of mouse rod photoreceptor cells, was reduced significantly when KIF3A, the obligate motor subunit of heterotrimeric kinesin-2, was deficient. These studies therefore provide evidence from live-cell analysis that the conserved heterotrimeric kinesin-2 is required for the normal transport of opsin along the ciliary plasma membrane.
Collapse
|
37
|
Wang J, Zhang N, Beuve A, Townes-Anderson E. Mislocalized opsin and cAMP signaling: a mechanism for sprouting by rod cells in retinal degeneration. Invest Ophthalmol Vis Sci 2012; 53:6355-69. [PMID: 22899763 DOI: 10.1167/iovs.12-10180] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
PURPOSE In human retinal degeneration, rod photoreceptors reactively sprout neurites. The mechanism is unknown in part because of the paucity of animal models displaying this feature of human pathology. We tested the role of cAMP and opsin in sprouting by tiger salamander rod cells, photoreceptors that can produce reactive growth. METHODS In vitro systems of isolated photoreceptor cells and intact neural retina were used. cAMP signaling was manipulated with nucleotide analogues, enzyme stimulators, agonists for adenosine and dopamine receptors, and the opsin agonist, β-ionone. Levels of cAMP were determined by radioimmunoassay, and protein levels by Western blot and quantitative immunocytochemistry. Neuritic growth was assayed by image analysis and conventional and confocal microscopy. RESULTS cAMP analogues and stimulation of adenylyl cyclase (AC) directly or through G-protein-coupled receptors resulted in significant increases in neuritic growth of isolated rod, but not cone, cells. The signaling pathway included protein kinase A (PKA) and phosphorylation of the transcription factor cAMP response element-binding protein (pCREB). Opsin, a G-linked receptor, is present throughout the plasmalemma of isolated cells; its activation also induced sprouting. In neural retina, rod sprouting was significantly increased by β-ionone with concomitant increases in cAMP, pCREB, and synaptic proteins. Notably, opsin stimulated sprouting only when mislocalized to the plasmalemma of the rod cell body. CONCLUSIONS cAMP causes neuritic sprouting in rod, but not cone, cells through the AC-PKA-CREB pathway known to be associated with synaptic plasticity. We propose that in retinal disease, mislocalized rod opsin gains access to cAMP signaling, which leads to neuritic sprouting.
Collapse
Affiliation(s)
- Jianfeng Wang
- Department of Neurology and Neuroscience, New Jersey Medical School–University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07103, USA
| | | | | | | |
Collapse
|
38
|
Sustained protection against photoreceptor degeneration in tubby mice by intravitreal injection of nanoceria. Biomaterials 2012; 33:8771-81. [PMID: 22959465 DOI: 10.1016/j.biomaterials.2012.08.030] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 08/13/2012] [Indexed: 02/05/2023]
Abstract
We previously reported that nanoceria can slow retinal degeneration in the tubby mouse for two weeks by multiple systemic injections. However, the long-term protection of retinal structure and function by directly deliver of nanoceria to the eye had not been explored. In this study, 172 ng of nanoceria in 1 μl saline (1 mm) were intravitreally injected into tubby P7 pups and assays were performed at P28, P49, P80 and P120. The expression of antioxidant associated genes and photoreceptor-specific genes was significantly up regulated, the mislocalization of rod and cone opsins was decreased, and retinal structure and function were protected. These findings demonstrate that nanoceria can function as catalytic antioxidants in vivo and may be broad spectrum therapeutic agents for multiple types of ocular diseases.
Collapse
|
39
|
Deretic D, Wang J. Molecular assemblies that control rhodopsin transport to the cilia. Vision Res 2012; 75:5-10. [PMID: 22892112 DOI: 10.1016/j.visres.2012.07.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 07/25/2012] [Indexed: 01/09/2023]
Abstract
This review will focus on the conserved molecular mechanisms for the specific targeting of rhodopsin and rhodopsin-like sensory receptors to the primary cilia. We will discuss the molecular assemblies that control the movement of rhodopsin from the central sorting station of the cell, the trans-Golgi network (TGN), into membrane-enclosed rhodopsin transport carriers (RTCs), and their delivery to the primary cilia and the cilia-derived sensory organelle, the rod outer segment (ROS). Recent studies reveal that these processes are initiated by the synergistic interaction of rhodopsin with the active form of the G-protein Arf4 and the Arf GTPase activating protein (GAP) ASAP1. During rhodopsin progression, ASAP1 serves as an activation platform that brings together the proteins necessary for transport to the cilia, including the Rab11a-Rabin8-Rab8 complex involved in ciliogenesis. These specialized molecular assemblies, through successive action of discrete modules, cooperatively determine how rhodopsin and other rhodopsin-like signaling receptors gain access to primary cilia.
Collapse
Affiliation(s)
- Dusanka Deretic
- Department of Surgery, Division of Ophthalmology, University of New Mexico, Albuquerque, NM 87131, United States.
| | | |
Collapse
|
40
|
Cai X, Sezate SA, Seal S, McGinnis JF. WITHDRAWN: Sustained protection against photoreceptor degeneration in tubby mice by intravitreal injection of nanoceria. Biomaterials 2012:S0142-9612(12)00767-3. [PMID: 22871422 DOI: 10.1016/j.biomaterials.2012.06.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 06/30/2012] [Indexed: 11/21/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Xue Cai
- Department of Ophthalmology, Dean A. McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | |
Collapse
|
41
|
Kinesin-2 motors transport IFT-particles, dyneins and tubulin subunits to the tips of Caenorhabditis elegans sensory cilia: relevance to vision research? Vision Res 2012; 75:44-52. [PMID: 22772029 DOI: 10.1016/j.visres.2012.06.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 06/19/2012] [Accepted: 06/19/2012] [Indexed: 10/28/2022]
Abstract
The sensory outer segments (OS) of vertebrate retinal photoreceptors, which detect photons of light, resemble the distal segments of Caenorhabditis elegans sensory cilia, which detect chemical ligands that influence the chemotactic movements of the animal. Based on fluorescence microscopy assays performed in sensory cilia of living, transgenic "wild type" and mutant C. elegans, combined with in vitro motility assays using purified motors, we have proposed that two types of kinesin-2 motor, heterotrimeric kinesin-II and homodimeric OSM-3, cooperate to build amphid and phasmid sensory cilia on chemosensory neurons. Specifically, we propose that these motors function together in a redundant manner to build the axoneme core (aka middle segments (MS)), whereas OSM-3 alone serves to build the distal segments (DS). Furthermore, our data suggest that these motors accomplish this by driving two sequential steps of anterograde transport of cargoes consisting of IFT-particles, retrograde dynein motors, and ciliary tubulin subunits, from the transition zone to the tips of the axonemal microtubules (MTs). Homologs of kinesin-II (KIF3) and OSM-3 (KIF17) are also proposed to contribute to the assembly of vertebrate photoreceptors, although how they do so is currently unclear. Here I review our work on kinesin-2 motors, intraflagellar transport (IFT) and cilium biogenesis in C. elegans sensory cilia, and comment on its possible relevance to current research on vertebrate photoreceptor cilia assembly and function.
Collapse
|
42
|
Hollingsworth TJ, Gross AK. Defective trafficking of rhodopsin and its role in retinal degenerations. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 293:1-44. [PMID: 22251557 DOI: 10.1016/b978-0-12-394304-0.00006-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Retinitis pigmentosa is a retinal degeneration transmitted by varied modes of inheritance and affects approximately 1 in 4000 individuals. The photoreceptors of the outer retina, as well as the retinal pigmented epithelium which supports the outer retina metabolically and structurally, are the retinal regions most affected by the disorder. In several forms of retinitis pigmentosa, the mislocalization of the rod photoreceptor protein rhodopsin is thought to be a contributing factor underlying the pathophysiology seen in patients. The mutations causing this mislocalization often occur in genes coding proteins involved in ciliary formation, vesicular transport, rod outer segment disc formation, and stability, as well as the rhodopsin protein itself. Often, these mutations result in the most early-onset cases of both recessive and dominant retinitis pigmentosa, and the following presents a discussion of the proteins, their degenerative phenotypes, and possible treatments of the disease.
Collapse
Affiliation(s)
- T J Hollingsworth
- Department of Vision Sciences, University of Alabama, Birmingham, Alabama, USA
| | | |
Collapse
|
43
|
Keady BT, Le YZ, Pazour GJ. IFT20 is required for opsin trafficking and photoreceptor outer segment development. Mol Biol Cell 2011; 22:921-30. [PMID: 21307337 PMCID: PMC3069017 DOI: 10.1091/mbc.e10-09-0792] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The light-detecting outer segments of vertebrate photoreceptors are cilia. Like other cilia, all materials needed for assembly and maintenance are synthesized in the cell body and transported into the cilium. The highly elaborated nature of the outer segment and its high rate of turnover necessitate unusually high levels of transport into the cilium. In this work, we examine the role of the IFT20 subunit of the intraflagellar transport (IFT) particle in photoreceptor cells. IFT20 was deleted in developing cones by a cone-specific Cre and in mature rods and cones by a tamoxifen-activatable Cre. Loss of IFT20 during cone development leads to opsin accumulation in the inner segment even when the connecting cilium and outer segment are still intact. With time this causes cone cell degeneration. Similarly, deletion of IFT20 in mature rods causes rapid accumulation of rhodopsin in the cell body, where it is concentrated at the Golgi complex. We further show that IFT20, acting both as part of the IFT particle and independent of the particle, binds to rhodopsin and RG-opsin. Since IFT20 dynamically moves between the Golgi complex and the connecting cilium, the current work suggests that rhodopsin and opsins are cargo for IFT transport.
Collapse
Affiliation(s)
- Brian T Keady
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | |
Collapse
|