1
|
Katada Y, Yoshida K, Serizawa N, Lee D, Kobayashi K, Negishi K, Okano H, Kandori H, Tsubota K, Kurihara T. Highly sensitive visual restoration and protection via ectopic expression of chimeric rhodopsin in mice. iScience 2023; 26:107716. [PMID: 37720108 PMCID: PMC10504486 DOI: 10.1016/j.isci.2023.107716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/22/2023] [Accepted: 08/22/2023] [Indexed: 09/19/2023] Open
Abstract
Photoreception requires amplification by mammalian rhodopsin through G protein activation, which requires a visual cycle. To achieve this in retinal gene therapy, we incorporated human rhodopsin cytoplasmic loops into Gloeobacter rhodopsin, thereby generating Gloeobacter and human chimeric rhodopsin (GHCR). In a murine model of inherited retinal degeneration, we induced retinal GHCR expression by intravitreal injection of a recombinant adeno-associated virus vector. Retinal explant and visual thalamus electrophysiological recordings, behavioral tests, and histological analysis showed that GHCR restored dim-environment vision and prevented the progression of retinal degeneration. Thus, GHCR may be a potent clinical tool for the treatment of retinal disorders.
Collapse
Affiliation(s)
- Yusaku Katada
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazuho Yoshida
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Aichi 466-0061, Japan
| | - Naho Serizawa
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Nutritional Sciences, Toyo University, Kita-ku, Tokyo 115-8650, Japan
| | - Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideki Kandori
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Aichi 466-0061, Japan
| | - Kazuo Tsubota
- Tsubota Laboratory, Inc., Shinjuku-ku, Tokyo 160-0016, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
2
|
Liu Y, Wang X, Gong R, Xu G, Zhu M. Overexpression of Rhodopsin or Its Mutants Leads to Energy Metabolism Dysfunction in 661w Cells. Invest Ophthalmol Vis Sci 2022; 63:2. [PMID: 36469028 PMCID: PMC9730732 DOI: 10.1167/iovs.63.13.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Retinitis pigmentosa (RP) is a heterogeneous group of inherited disorders characterized by photoreceptor degeneration. The rhodopsin gene (RHO) is the most frequent cause of autosomal dominant RP (ADRP), yet it remains unclear how RHO mutations cause heterogeneous phenotypes. Energy failure is a main cause of the secondary cone death during RP progression; however, its role in primary rod death induced by ADRP RHO mutants is unknown. Methods Three RHO missense mutations were chosen from different clinical classes. Wild-type (WT) RHO and its mutants, P23H (class B1), R135L (class A), and G188R (class B2), were overexpressed in 661w cells, a mouse photoreceptor cell line, and their effects on oxidative phosphorylation (OXPHOS) and aerobic glycolysis were compared separately. Results Here, we report that energy failure is an early event in the cell death caused by overexpression of WT RHO and its mutants. RHO overexpression leads to OXPHOS deficiency, which might be a result of mitochondrial loss. Nonetheless, only in WT RHO and P23H groups, energy stress triggers AMP-activated protein kinase activation and metabolic reprogramming to increase glycolysis. Metabolic reprogramming impairment in R135L and G188R groups might be the reason why energy failure and cell injury are much more severe in those groups. Conclusions Our results imply that overexpression of RHO missense mutants have distinct impacts on the two energy metabolic pathways, which might be related to their heterogeneous phenotypes.
Collapse
Affiliation(s)
- Yang Liu
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Fudan University, Shanghai, China,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xin Wang
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Fudan University, Shanghai, China,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Ruowen Gong
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Fudan University, Shanghai, China,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Gezhi Xu
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Fudan University, Shanghai, China,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Min Zhu
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Fudan University, Shanghai, China,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
3
|
Intartaglia D, Giamundo G, Naso F, Nusco E, Di Giulio S, Salierno FG, Polishchuk E, Conte I. Induction of Autophagy Promotes Clearance of RHOP23H Aggregates and Protects From Retinal Degeneration. Front Aging Neurosci 2022; 14:878958. [PMID: 35847673 PMCID: PMC9281868 DOI: 10.3389/fnagi.2022.878958] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/19/2022] [Indexed: 12/16/2022] Open
Abstract
Autophagy is a critical metabolic process that acts as a major self-digestion and recycling pathway contributing to maintain cellular homeostasis. An emerging field of research supports the therapeutic modulation of autophagy for treating human neurodegenerative disorders, in which toxic aggregates are accumulated in neurons. Our previous study identified Ezrin protein as an inhibitor of autophagy and lysosomal functions in the retina; thus, in turn, identifying it as a potential pharmacological target for increasing retinal cell clearance to treat inherited retinal dystrophies in which misfolded proteins have accumulated. This study aimed to verify the therapeutic inhibition of Ezrin to induce clearance of toxic aggregates in a mouse model for a dominant form of retinitis pigmentosa (i.e., RHOP23H/+). We found that daily inhibition of Ezrin significantly decreased the accumulation of misfolded RHOP23H aggregates. Remarkably, induction of autophagy, by a drug-mediated pulsatile inhibition of Ezrin, promoted the lysosomal clearance of disease-linked RHOP23H aggregates. This was accompanied with a reduction of endoplasmic reticulum (ER)-stress, robust decrease of photoreceptors' cell death, amelioration in both retinal morphology and function culminating in a better preservation of vision. Our study opens new perspectives for a pulsatile pharmacological induction of autophagy as a mutation-independent therapy paving the way toward a more effective therapeutic strategy to treat these devastating retinal disorders due to an accumulation of intracellular toxic aggregates.
Collapse
Affiliation(s)
| | - Giuliana Giamundo
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Federica Naso
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | - Edoardo Nusco
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
| | | | | | | | - Ivan Conte
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Biology, University of Naples Federico II, Naples, Italy
- *Correspondence: Ivan Conte
| |
Collapse
|
4
|
Jiang K, Fairless E, Kanda A, Gotoh N, Cogliati T, Li T, Swaroop A. Divergent Effects of HSP70 Overexpression in Photoreceptors During Inherited Retinal Degeneration. Invest Ophthalmol Vis Sci 2021; 61:25. [PMID: 33107904 PMCID: PMC7594617 DOI: 10.1167/iovs.61.12.25] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Purpose Disruption of proteostasis is a key event in many neurodegenerative diseases. Heat shock proteins (HSPs) participate in multiple functions associated with intracellular transport and proteostasis. We evaluated the effect of augmented HSP70 expression in mutant photoreceptors of mouse retinal degeneration models to test the hypothesis that failure to sustain HSP70 expression contributes to photoreceptor cell death. Methods We examined HSP70 expression in retinas of wild-type and mutant mice by RNA and protein analysis. A transgenic mouse line, TgCrx-Hspa1a-Flag, was generated to express FLAG-tagged full-length HSP70 protein under control of a 2.3 kb mouse Crx promoter. This line was crossed to three distinct retinal degeneration mouse models. Retinal structure and function were evaluated by histology, immunohistochemistry, and electroretinography. Results In seven different mouse models of retinal degeneration, we detected transient elevation of endogenous HSP70 expression at early stages, followed by a dramatic reduction as cell death ensues, suggesting an initial adaptive response to cellular stress. Augmented expression of HSP70 in RHOT17M mice, in which mutant rhodopsin is misfolded, marginally improved photoreceptor survival, whereas elevated HSP70 led to more severe retinal degeneration in rd10 mutants that produce a partially functional PDE6B. In Rpgrip1−/− mice that display a ciliary defect, higher HSP70 had no impact on photoreceptor survival or function. Conclusions HSP70 overexpression has divergent effects in photoreceptors determined, at least in part, by the nature of the mutant protein each model carries. Additional investigations on HSP pathways and associated chaperone networks in photoreceptors are needed before designing therapeutic strategies targeting proteostasis.
Collapse
Affiliation(s)
- Ke Jiang
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Elizabeth Fairless
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Atsuhiro Kanda
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Norimoto Gotoh
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Tiziana Cogliati
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Tiansen Li
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Anand Swaroop
- Neurobiology, Neurodegeneration, and Repair Laboratory (NNRL), National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
5
|
Kakavand K, Jobling AI, Greferath U, Vessey KA, de Iongh RU, Fletcher EL. Photoreceptor Degeneration in Pro23His Transgenic Rats (Line 3) Involves Autophagic and Necroptotic Mechanisms. Front Neurosci 2020; 14:581579. [PMID: 33224023 PMCID: PMC7670078 DOI: 10.3389/fnins.2020.581579] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 10/02/2020] [Indexed: 01/09/2023] Open
Abstract
Photoreceptor death contributes to 50% of irreversible vision loss in the western world. Pro23His (P23H) transgenic albino rat strains are widely used models for the most common rhodopsin gene mutation associated with the autosomal dominant form of retinitis pigmentosa. However, the mechanism(s) by which photoreceptor death occurs are not well understood and were the principal aim of this study. We first used electroretinogram recording and optical coherence tomography to confirm the time course of functional and structural loss. Electroretinogram analyses revealed significantly decreased rod photoreceptor (a-wave), bipolar cell (b-wave) and amacrine cell responses (oscillatory potentials) from P30 onward. The cone-mediated b-wave was also decreased from P30. TUNEL analysis showed extensive cell death at P18, with continued labeling detected until P30. Focused gene expression arrays indicated activation of, apoptosis, autophagy and necroptosis in whole retina from P14-18. However, analysis of mitochondrial permeability changes (ΔΨm) using JC-1 dye, combined with immunofluorescence markers for caspase-dependent (cleaved caspase-3) and caspase-independent (AIF) cell death pathways, indicated mitochondrial-mediated cell death was not a major contributor to photoreceptor death. By contrast, reverse-phase protein array data combined with RIPK3 and phospho-MLKL immunofluorescence indicated widespread necroptosis as the predominant mechanism of photoreceptor death. These findings highlight the complexity of mechanisms involved in photoreceptor death in the Pro23His rat model of degeneration and suggest therapies that target necroptosis should be considered for their potential to reduce photoreceptor death.
Collapse
Affiliation(s)
- Kiana Kakavand
- Visual Neuroscience Laboratory, Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew I Jobling
- Visual Neuroscience Laboratory, Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Ursula Greferath
- Visual Neuroscience Laboratory, Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Kirstan A Vessey
- Visual Neuroscience Laboratory, Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Robb U de Iongh
- Ocular Development Laboratory, Department Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Erica L Fletcher
- Visual Neuroscience Laboratory, Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
6
|
Disorders of FZ-CRD; insights towards FZ-CRD folding and therapeutic landscape. Mol Med 2019; 26:4. [PMID: 31892318 PMCID: PMC6938638 DOI: 10.1186/s10020-019-0129-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 12/13/2019] [Indexed: 02/08/2023] Open
Abstract
The ER is hub for protein folding. Proteins that harbor a Frizzled cysteine-rich domain (FZ-CRD) possess 10 conserved cysteine motifs held by a unique disulfide bridge pattern which attains a correct fold in the ER. Little is known about implications of disease-causing missense mutations within FZ-CRD families. Mutations in FZ-CRD of Frizzled class receptor 4 (FZD4) and Muscle, skeletal, receptor tyrosine kinase (MuSK) and Receptor tyrosine kinase-like orphan receptor 2 (ROR2) cause Familial Exudative Vitreoretinopathy (FEVR), Congenital Myasthenic Syndrome (CMS), and Robinow Syndrome (RS) respectively. We highlight reported pathogenic inherited missense mutations in FZ-CRD of FZD4, MuSK and ROR2 which misfold, and traffic abnormally in the ER, with ER-associated degradation (ERAD) as a common pathogenic mechanism for disease. Our review shows that all studied FZ-CRD mutants of RS, FEVR and CMS result in misfolded proteins and/or partially misfolded proteins with an ERAD fate, thus we coin them as “disorders of FZ-CRD”. Abnormal trafficking was demonstrated in 17 of 29 mutants studied; 16 mutants were within and/or surrounding the FZ-CRD with two mutants distant from FZ-CRD. These ER-retained mutants were improperly N-glycosylated confirming ER-localization. FZD4 and MuSK mutants were tagged with polyubiquitin chains confirming targeting for proteasomal degradation. Investigating the cellular and molecular mechanisms of these mutations is important since misfolded protein and ER-targeted therapies are in development. The P344R-MuSK kinase mutant showed around 50% of its in-vitro autophosphorylation activity and P344R-MuSK increased two-fold on proteasome inhibition. M105T-FZD4, C204Y-FZD4, and P344R-MuSK mutants are thermosensitive and therefore, might benefit from extending the investigation to a larger number of chemical chaperones and/or proteasome inhibitors. Nonetheless, FZ-CRD ER-lipidation it less characterized in the literature and recent structural data sheds light on the importance of lipidation in protein glycosylation, proper folding, and ER trafficking. Current treatment strategies in-place for the conformational disease landscape is highlighted. From this review, we envision that disorders of FZ-CRD might be receptive to therapies that target FZ-CRD misfolding, regulation of fatty acids, and/or ER therapies; thus paving the way for a newly explored paradigm to treat different diseases with common defects.
Collapse
|
7
|
Scirè A, Cianfruglia L, Minnelli C, Bartolini D, Torquato P, Principato G, Galli F, Armeni T. Glutathione compartmentalization and its role in glutathionylation and other regulatory processes of cellular pathways. Biofactors 2019; 45:152-168. [PMID: 30561781 DOI: 10.1002/biof.1476] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/20/2022]
Abstract
Glutathione is considered the major non-protein low molecular weight modulator of redox processes and the most important thiol reducing agent of the cell. The biosynthesis of glutathione occurs in the cytosol from its constituent amino acids, but this tripeptide is also present in the most important cellular districts, such as mitochondria, nucleus, and endoplasmic reticulum, thus playing a central role in several metabolic pathways and cytoprotection mechanisms. Indeed, glutathione is involved in the modulation of various cellular processes and, not by chance, it is a ubiquitous determinant for redox signaling, xenobiotic detoxification, and regulation of cell cycle and death programs. The balance between its concentration and redox state is due to a complex series of interactions between biosynthesis, utilization, degradation, and transport. All these factors are of great importance to understand the significance of cellular redox balance and its relationship with physiological responses and pathological conditions. The purpose of this review is to give an overview on glutathione cellular compartmentalization. Information on its subcellular distribution provides a deeper understanding of glutathione-dependent processes and reflects the importance of compartmentalization in the regulation of specific cellular pathways. © 2018 BioFactors, 45(2):152-168, 2019.
Collapse
Affiliation(s)
- Andrea Scirè
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Laura Cianfruglia
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Cristina Minnelli
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Desirée Bartolini
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Pierangelo Torquato
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Giovanni Principato
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Galli
- Clinical Biochemistry and Human Nutrition Labs, Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Tatiana Armeni
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
8
|
Sullivan LS, Bowne SJ, Koboldt DC, Cadena EL, Heckenlively JR, Branham KE, Wheaton DH, Jones KD, Ruiz RS, Pennesi ME, Yang P, Davis-Boozer D, Northrup H, Gurevich VV, Chen R, Xu M, Li Y, Birch DG, Daiger SP. A Novel Dominant Mutation in SAG, the Arrestin-1 Gene, Is a Common Cause of Retinitis Pigmentosa in Hispanic Families in the Southwestern United States. Invest Ophthalmol Vis Sci 2017; 58:2774-2784. [PMID: 28549094 PMCID: PMC5455168 DOI: 10.1167/iovs.16-21341] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/23/2017] [Indexed: 01/22/2023] Open
Abstract
Purpose To identify the causes of autosomal dominant retinitis pigmentosa (adRP) in a cohort of families without mutations in known adRP genes and consequently to characterize a novel dominant-acting missense mutation in SAG. Methods Patients underwent ophthalmologic testing and were screened for mutations using targeted-capture and whole-exome next-generation sequencing. Confirmation and additional screening were done by Sanger sequencing. Haplotypes segregating with the mutation were determined using short tandem repeat and single nucleotide variant polymorphisms. Genealogies were established by interviews of family members. Results Eight families in a cohort of 300 adRP families, and four additional families, were found to have a novel heterozygous mutation in the SAG gene, c.440G>T; p.Cys147Phe. Patients exhibited symptoms of retinitis pigmentosa and none showed symptoms characteristic of Oguchi disease. All families are of Hispanic descent and most were ascertained in Texas or California. A single haplotype including the SAG mutation was identified in all families. The mutation dramatically alters a conserved amino acid, is extremely rare in global databases, and was not found in 4000+ exomes from Hispanic controls. Molecular modeling based on the crystal structure of bovine arrestin-1 predicts protein misfolding/instability. Conclusions This is the first dominant-acting mutation identified in SAG, a founder mutation possibly originating in Mexico several centuries ago. The phenotype is clearly adRP and is distinct from the previously reported phenotypes of recessive null mutations, that is, Oguchi disease and recessive RP. The mutation accounts for 3% of the 300 families in the adRP Cohort and 36% of Hispanic families in this cohort.
Collapse
Affiliation(s)
- Lori S. Sullivan
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center, Houston, Texas, United States
| | - Sara J. Bowne
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center, Houston, Texas, United States
| | | | - Elizabeth L. Cadena
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center, Houston, Texas, United States
| | | | - Kari E. Branham
- Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, United States
| | | | - Kaylie D. Jones
- Retina Foundation of the Southwest, Dallas, Texas, United States
| | - Richard S. Ruiz
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, United States
| | - Mark E. Pennesi
- Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Paul Yang
- Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - David Davis-Boozer
- Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States
| | - Hope Northrup
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, United States
| | | | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Mingchu Xu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Yumei Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - David G. Birch
- Retina Foundation of the Southwest, Dallas, Texas, United States
| | - Stephen P. Daiger
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center, Houston, Texas, United States
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, United States
| |
Collapse
|
9
|
Beerepoot P, Nazari R, Salahpour A. Pharmacological chaperone approaches for rescuing GPCR mutants: Current state, challenges, and screening strategies. Pharmacol Res 2017; 117:242-251. [DOI: 10.1016/j.phrs.2016.12.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 12/11/2022]
|
10
|
Khanam H, Ali A, Asif M, Shamsuzzaman. Neurodegenerative diseases linked to misfolded proteins and their therapeutic approaches: A review. Eur J Med Chem 2016; 124:1121-1141. [DOI: 10.1016/j.ejmech.2016.08.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/02/2016] [Accepted: 08/05/2016] [Indexed: 12/11/2022]
|
11
|
Gene therapy restores vision in rd1 mice after removal of a confounding mutation in Gpr179. Nat Commun 2015; 6:6006. [PMID: 25613321 PMCID: PMC4354202 DOI: 10.1038/ncomms7006] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 12/01/2014] [Indexed: 12/21/2022] Open
Abstract
The rd1 mouse with a mutation in the Pde6b gene was the first strain of mice identified with a retinal degeneration. However, AAV-mediated gene supplementation of rd1 mice only results in structural preservation of photoreceptors, and restoration of the photoreceptor-mediated a-wave, but not in restoration of the bipolar cell-mediated b-wave. Here we show that a mutation in Gpr179 prevents the full restoration of vision in rd1 mice. Backcrossing rd1 with C57BL6 mice reveals the complete lack of b-wave in a subset of mice, consistent with an autosomal recessive Mendelian inheritance pattern. We identify a mutation in the Gpr179 gene, which encodes for a G-protein coupled receptor localized to the dendrites of ON-bipolar cells. Gene replacement in rd1 mice that are devoid of the mutation in Gpr179 successfully restores the function of both photoreceptors and bipolar cells, which is maintained for up to 13 months. Our discovery may explain the failure of previous gene therapy attempts in rd1 mice, and we propose that Grp179 mutation status should be taken into account in future studies involving rd1 mice.
Collapse
|
12
|
Jiang H, Xiong S, Xia X. Retinitis pigmentosa‑associated rhodopsin mutant T17M induces endoplasmic reticulum (ER) stress and sensitizes cells to ER stress-induced cell death. Mol Med Rep 2014; 9:1737-42. [PMID: 24573320 DOI: 10.3892/mmr.2014.1987] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 02/14/2014] [Indexed: 11/06/2022] Open
Abstract
Retinitis pigmentosa (RP) is a group of inherited diseases that primarily affect light‑sensitive rods and cones in the retina. Rhodopsin mutations, including the T17M mutation, are associated with the autosomal dominant form of retinitis pigmentosa (ADRP) and have been linked to abnormal protein folding. However, the molecular mechanisms underlying T17M rhodopsin‑induced retinal degeneration are yet to be elucidated. In the present study, Human embryonic kidney (HEK) 293 and ARPE‑19 cells were transfected with myc‑tagged wild‑type (WT) and T17M rhodopsin constructs. Cells were fixed and stained with anti‑myc antibodies and the localization of WT and T17M rhodopsin was visualized using immunofluorescence microscopy. Turnover rates of WT and T17M rhodopsin were measured using western blot analysis. In addition, endoplasmic reticulum (ER) stress‑induced cell death was analyzed in WT and T17M rhodopsin‑transfected cells using nuclear staining. Misfolded T17M rhodopsin was observed to be abnormally localized in the ER, while WT rhodopsin was predominantly found at the plasma membrane. Protein turnover analysis revealed that T17M rhodopsin was more rapidly degraded by proteasomes than WT rhodopsin. Furthermore, overexpression of T17M rhodopsin was observed to induce cell death and increase cytotoxicity; predisposing cells to ER stress‑induced cell death. These findings show novel insight into the properties of T17M rhodopsin and highlight the role of ER stress in T17M‑associated RP.
Collapse
Affiliation(s)
- Haibo Jiang
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Siqi Xiong
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Xiaobo Xia
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
13
|
Nguyen ATH, Campbell M, Kiang AS, Humphries MM, Humphries P. Current therapeutic strategies for P23H RHO-linked RP. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 801:471-6. [PMID: 24664733 DOI: 10.1007/978-1-4614-3209-8_60] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The first autosomal dominant mutation identified to cause retinitis pigmentosa in the North American population was the substitution of proline to histidine at position 23 of the rhodopsin gene (P23H RHO). Many biochemical studies have demonstrated that P23H mutation induces rhodopsin (RHO) misfolding leading to endoplasmic reticulum stress. Herein, we review current thinking of this topic.
Collapse
Affiliation(s)
- Anh T H Nguyen
- The Ocular Genetics Unit, Smurfit Institute of Genetics, Trinity College Dublin, College Green, Dublin 2, Ireland,
| | | | | | | | | |
Collapse
|
14
|
Abstract
The ARF-like (ARL) proteins, within the ARF family, are a collection of functionally diverse GTPases that share extensive (>40 %) identity with the ARFs and each other and are assumed to share basic mechanisms of regulation and a very incompletely documented degree of overlapping regulators. At least four ARLs were already present in the last eukaryotic common ancestor, along with one ARF, and these have been expanded to >20 members in mammals. We know little about the majority of these proteins so our review will focus on those about which the most is known, including ARL1, ARL2, ARL3, ARL4s, ARL6, ARL13s, and ARFRP1. From this fragmentary information we extract some generalizations and conclusions regarding the sources and extent of specificity and functions of the ARLs.
Collapse
Affiliation(s)
- Alfred Wittinghofer
- Max-Planck-Institute of Molecular Physiology, Dortmund, Nordrhein-Westfalen Germany
| |
Collapse
|
15
|
Tastan O, Dutta A, Booth P, Klein-Seetharaman J. Retinal proteins as model systems for membrane protein folding. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2013; 1837:656-63. [PMID: 24333783 DOI: 10.1016/j.bbabio.2013.11.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 11/19/2013] [Accepted: 11/28/2013] [Indexed: 11/30/2022]
Abstract
Experimental folding studies of membrane proteins are more challenging than water-soluble proteins because of the higher hydrophobicity content of membrane embedded sequences and the need to provide a hydrophobic milieu for the transmembrane regions. The first challenge is their denaturation: due to the thermodynamic instability of polar groups in the membrane, secondary structures in membrane proteins are more difficult to disrupt than in soluble proteins. The second challenge is to refold from the denatured states. Successful refolding of membrane proteins has almost always been from very subtly denatured states. Therefore, it can be useful to analyze membrane protein folding using computational methods, and we will provide results obtained with simulated unfolding of membrane protein structures using the Floppy Inclusions and Rigid Substructure Topography (FIRST) method. Computational methods have the advantage that they allow a direct comparison between diverse membrane proteins. We will review here both, experimental and FIRST studies of the retinal binding proteins bacteriorhodopsin and mammalian rhodopsin, and discuss the extension of the findings to deriving hypotheses on the mechanisms of folding of membrane proteins in general. This article is part of a Special Issue entitled: Retinal Proteins-You can teach an old dog new tricks.
Collapse
Affiliation(s)
- Oznur Tastan
- Department of Computer Engineering, Bilkent University, Ankara, Turkey
| | - Arpana Dutta
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, USA
| | - Paula Booth
- School of Biochemistry, University of Bristol, UK
| | - Judith Klein-Seetharaman
- Division of Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, UK.
| |
Collapse
|
16
|
Mori R, Toda T. The dual role of fission yeast Tbc1/cofactor C orchestrates microtubule homeostasis in tubulin folding and acts as a GAP for GTPase Alp41/Arl2. Mol Biol Cell 2013; 24:1713-24, S1-8. [PMID: 23576550 PMCID: PMC3667724 DOI: 10.1091/mbc.e12-11-0792] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 02/22/2013] [Accepted: 03/29/2013] [Indexed: 11/11/2022] Open
Abstract
Supplying the appropriate amount of correctly folded α/β-tubulin heterodimers is critical for microtubule dynamics. Formation of assembly-competent heterodimers is remarkably elaborate at the molecular level, in which the α- and β-tubulins are separately processed in a chaperone-dependent manner. This sequential step is performed by the tubulin-folding cofactor pathway, comprising a specific set of regulatory proteins: cofactors A-E. We identified the fission yeast cofactor: the orthologue of cofactor C, Tbc1. In addition to its roles in tubulin folding, Tbc1 acts as a GAP in regulating Alp41/Arl2, a highly conserved small GTPase. Of interest, the expression of GDP- or GTP-bound Alp41 showed the identical microtubule loss phenotype, suggesting that continuous cycling between these forms is important for its functions. In addition, we found that Alp41 interacts with Alp1(D), the orthologue of cofactor D, specifically when in the GDP-bound form. Intriguingly, Alp1(D) colocalizes with microtubules when in excess, eventually leading to depolymerization, which is sequestered by co-overproducing GDP-bound Alp41. We present a model of the final stages of the tubulin cofactor pathway that includes a dual role for both Tbc1 and Alp1(D) in opposing regulation of the microtubule.
Collapse
Affiliation(s)
- Risa Mori
- Cell Regulation Laboratory, Cancer Research UK, London Research Institute, Lincoln's Inn Fields Laboratories, London WC2A 3LY, United Kingdom
| | - Takashi Toda
- Cell Regulation Laboratory, Cancer Research UK, London Research Institute, Lincoln's Inn Fields Laboratories, London WC2A 3LY, United Kingdom
| |
Collapse
|
17
|
Expression of wild-type Rp1 protein in Rp1 knock-in mice rescues the retinal degeneration phenotype. PLoS One 2012; 7:e43251. [PMID: 22927954 PMCID: PMC3424119 DOI: 10.1371/journal.pone.0043251] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 07/18/2012] [Indexed: 01/31/2023] Open
Abstract
Mutations in the retinitis pigmentosa 1 (RP1) gene are a common cause of autosomal dominant retinitis pigmentosa (adRP), and have also been found to cause autosomal recessive RP (arRP) in a few families. The 33 dominant mutations and 6 recessive RP1 mutations identified to date are all nonsense or frameshift mutations, and almost exclusively (38 out of 39) are located in the 4th and final exon of RP1. To better understand the underlying disease mechanisms of and help develop therapeutic strategies for RP1 disease, we performed a series of human genetic and animal studies using gene targeted and transgenic mice. Here we report that a frameshift mutation in the 3rd exon of RP1 (c.686delC; p.P229QfsX35) found in a patient with recessive RP1 disease causes RP in the homozygous state, whereas the heterozygous carriers are unaffected, confirming that haploinsufficiency is not the causative mechanism for RP1 disease. We then generated Rp1 knock-in mice with a nonsense Q662X mutation in exon 4, as well as Rp1 transgenic mice carrying a wild-type BAC Rp1 transgene. The Rp1-Q662X allele produces a truncated Rp1 protein, and homozygous Rp1-Q662X mice experience a progressive photoreceptor degeneration characterized disorganization of photoreceptor outer segments. This phenotype could be prevented by expression of a normal amount of Rp1 protein from the BAC transgene without removal of the mutant Rp1-Q662X protein. Over-expression of Rp1 protein in additional BAC Rp1 transgenic lines resulted in retinal degeneration. These findings suggest that the truncated Rp1-Q662X protein does not exert a toxic gain-of-function effect. These results also imply that in principle gene augmentation therapy could be beneficial for both recessive and dominant RP1 patients, but the levels of RP1 protein delivered for therapy will have to be carefully controlled.
Collapse
|
18
|
Hibbard KL, O’Tousa JE. A role for the cytoplasmic DEAD box helicase Dbp21E2 in rhodopsin maturation and photoreceptor viability. J Neurogenet 2012; 26:177-88. [PMID: 22794106 PMCID: PMC3680124 DOI: 10.3109/01677063.2012.692412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The Dbp21E2 (DEAD box protein 21E2) is a member of a family of DEAD box helicases active in RNA processing and stability. The authors used genetic mosaics to identify mutants in Dbp21E2 that affect rhodopsin biogenesis and the maintenance of photoreceptor structure. Analysis of a green fluorescent protein (GFP)-tagged Rh1 rhodopsin construct placed under control of a heat shock promoter showed that Dbp21E21 fails to efficiently transport Rh1 from the photoreceptor cell body to the rhabdomere. Retinal degeneration is not dependent on the Rh1 transport defects. The authors also showed that GFP- and red fluorescent protein (RFP)-tagged Dbp21E2 proteins are localized to discrete cytoplasmic structures that are not associated with organelles known to be active in rhodopsin transport. The molecular genetic analysis described here reveals an unexpected role for the Dbp21E2 helicase and provides an experimental system to further characterize its function.
Collapse
Affiliation(s)
- Karen L. Hibbard
- Dept. of Biological Sciences, Univ. of Notre Dame, Notre Dame, IN, USA
| | | |
Collapse
|
19
|
Pennesi ME, Stover NB, Stone EM, Chiang PW, Weleber RG. Residual electroretinograms in young Leber congenital amaurosis patients with mutations of AIPL1. Invest Ophthalmol Vis Sci 2011; 52:8166-73. [PMID: 21900377 DOI: 10.1167/iovs.11-8298] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE To describe in detail the clinical phenotype and electrophysiological features of three patients with Leber congenital amaurosis caused by mutations of AIPL1. METHODS Ophthalmologic examination, color fundus photography, detailed electrophysiological assessment, and screening of AIPL1 were undertaken in three subjects. One patient also underwent visual field testing and spectral domain-optical coherence tomography. RESULTS All three patients, two of whom were siblings, had histories consistent with Leber congenital amaurosis (severely reduced vision, poorly responsive pupils, and nystagmus presenting within the first year of life). However, each patient had recordable and similar electroretinograms (ERGs), which demonstrated absent cone-driven responses and slow insensitive scotopic responses. The first patient was found to have a homozygous Trp278 stop mutation in AIPL1, whereas the siblings were each found to have novel heterozygous mutations in AIPL1 (Leu17Pro and Lys214Asn). CONCLUSIONS Patients with mutations in AIPL1 may present with Leber congenital amaurosis and residual ERGs characterized by slow insensitive scotopic responses. Such responses are likely seen only in very young patients and may not be seen with the typical filter settings recommended by the ISCEV standards because of low-pass filtering. Progressive loss of residual ERG activity in young LCA patients with AIPL1 mutations suggests that gene replacement therapy will likely have to be performed early.
Collapse
Affiliation(s)
- Mark E Pennesi
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97239, USA.
| | | | | | | | | |
Collapse
|
20
|
Sullivan JM, Yau EH, Kolniak TA, Sheflin LG, Taggart RT, Abdelmaksoud HE. Variables and strategies in development of therapeutic post-transcriptional gene silencing agents. J Ophthalmol 2011; 2011:531380. [PMID: 21785698 PMCID: PMC3138052 DOI: 10.1155/2011/531380] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 02/17/2011] [Accepted: 02/28/2011] [Indexed: 11/24/2022] Open
Abstract
Post-transcriptional gene silencing (PTGS) agents such as ribozymes, RNAi and antisense have substantial potential for gene therapy of human retinal degenerations. These technologies are used to knockdown a specific target RNA and its cognate protein. The disease target mRNA may be a mutant mRNA causing an autosomal dominant retinal degeneration or a normal mRNA that is overexpressed in certain diseases. All PTGS technologies depend upon the initial critical annealing event of the PTGS ligand to the target RNA. This event requires that the PTGS agent is in a conformational state able to support hybridization and that the target have a large and accessible single-stranded platform to allow rapid annealing, although such platforms are rare. We address the biocomplexity that currently limits PTGS therapeutic development with particular emphasis on biophysical variables that influence cellular performance. We address the different strategies that can be used for development of PTGS agents intended for therapeutic translation. These issues apply generally to the development of PTGS agents for retinal, ocular, or systemic diseases. This review should assist the interested reader to rapidly appreciate critical variables in PTGS development and facilitate initial design and testing of such agents against new targets of clinical interest.
Collapse
Affiliation(s)
- Jack M. Sullivan
- Department of Ophthalmology, University at Buffalo SUNY, Buffalo, NY 14214, USA
- Department of Pharmacology and Toxicology, University at Buffalo SUNY, Buffalo, NY 14214, USA
- Department of Physiology and Biophysics, University at Buffalo SUNY, Buffalo, NY 14214, USA
- Neuroscience Program, University at Buffalo SUNY, Buffalo, NY 14214, USA
- Ross Eye Institute, University at Buffalo SUNY, Buffalo, NY 14209, USA
- Veterans Administration Western New York Healthcare System, Medical Research, Buffalo, NY 14215, USA
| | - Edwin H. Yau
- Department of Ophthalmology, University at Buffalo SUNY, Buffalo, NY 14214, USA
- Department of Pharmacology and Toxicology, University at Buffalo SUNY, Buffalo, NY 14214, USA
| | - Tiffany A. Kolniak
- Department of Ophthalmology, University at Buffalo SUNY, Buffalo, NY 14214, USA
- Neuroscience Program, University at Buffalo SUNY, Buffalo, NY 14214, USA
| | - Lowell G. Sheflin
- Department of Ophthalmology, University at Buffalo SUNY, Buffalo, NY 14214, USA
- Veterans Administration Western New York Healthcare System, Medical Research, Buffalo, NY 14215, USA
| | - R. Thomas Taggart
- Department of Ophthalmology, University at Buffalo SUNY, Buffalo, NY 14214, USA
| | - Heba E. Abdelmaksoud
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY 13215, USA
| |
Collapse
|
21
|
Sakamoto K, Mori A, Nakahara T, Ishii K. [Cause of retinitis pigmentosa and new therapeutics under development]. Nihon Yakurigaku Zasshi 2011; 137:22-26. [PMID: 21233585 DOI: 10.1254/fpj.137.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
|
22
|
|
23
|
Lopes VS, Jimeno D, Khanobdee K, Song X, Chen B, Nusinowitz S, Williams DS. Dysfunction of heterotrimeric kinesin-2 in rod photoreceptor cells and the role of opsin mislocalization in rapid cell death. Mol Biol Cell 2010; 21:4076-88. [PMID: 20926680 PMCID: PMC2993738 DOI: 10.1091/mbc.e10-08-0715] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Loss of kinesin-2 function causes rapid death of rod photoreceptors. The cell death is dependent on the expression of opsin, which first accumulates along the route to the outer segment, but not on signaling by opsin-arrestin complexes or by light activation; the key element appears to be the accumulation of excessive protein in the wrong place. Due to extensive elaboration of the photoreceptor cilium to form the outer segment, axonemal transport (IFT) in photoreceptors is extraordinarily busy, and retinal degeneration is a component of many ciliopathies. Functional loss of heterotrimeric kinesin-2, a major anterograde IFT motor, causes mislocalized opsin, followed by rapid cell death. Here, we have analyzed the nature of protein mislocalization and the requirements for the death of kinesin-2-mutant rod photoreceptors. Quantitative immuno EM showed that opsin accumulates initially within the inner segment, and then in the plasma membrane. The light-activated movement of arrestin to the outer segment is also impaired, but this defect likely results secondarily from binding to mislocalized opsin. Unlike some other retinal degenerations, neither opsin–arrestin complexes nor photoactivation were necessary for cell loss. In contrast, reduced rod opsin expression provided enhanced rod and cone photoreceptor survival and function, as measured by photoreceptor cell counts, apoptosis assays, and ERG analysis. The cell death incurred by loss of kinesin-2 function was almost completely negated by Rho−/−. Our results indicate that mislocalization of opsin is a major cause of photoreceptor cell death from kinesin-2 dysfunction and demonstrate the importance of accumulating mislocalized protein per se, rather than specific signaling properties of opsin, stemming from photoactivation or arrestin binding.
Collapse
Affiliation(s)
- Vanda S Lopes
- Departments of Ophthalmology and Neurobiology, UCLA School of Medicine, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Ohgane K, Dodo K, Hashimoto Y. Retinobenzaldehydes as proper-trafficking inducers of folding-defective P23H rhodopsin mutant responsible for Retinitis Pigmentosa. Bioorg Med Chem 2010; 18:7022-8. [DOI: 10.1016/j.bmc.2010.08.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 08/04/2010] [Accepted: 08/05/2010] [Indexed: 10/19/2022]
|
25
|
Griciuc A, Aron L, Roux MJ, Klein R, Giangrande A, Ueffing M. Inactivation of VCP/ter94 suppresses retinal pathology caused by misfolded rhodopsin in Drosophila. PLoS Genet 2010; 6:e1001075. [PMID: 20865169 PMCID: PMC2928793 DOI: 10.1371/journal.pgen.1001075] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 07/20/2010] [Indexed: 11/30/2022] Open
Abstract
The most common Rhodopsin (Rh) mutation associated with autosomal dominant retinitis pigmentosa (ADRP) in North America is the substitution of proline 23 by histidine (Rh(P23H)). Unlike the wild-type Rh, mutant Rh(P23H) exhibits folding defects and forms intracellular aggregates. The mechanisms responsible for the recognition and clearance of misfolded Rh(P23H) and their relevance to photoreceptor neuron (PN) degeneration are poorly understood. Folding-deficient membrane proteins are subjected to Endoplasmic Reticulum (ER) quality control, and we have recently shown that Rh(P23H) is a substrate of the ER-associated degradation (ERAD) effector VCP/ter94, a chaperone that extracts misfolded proteins from the ER (a process called retrotranslocation) and facilitates their proteasomal degradation. Here, we used Drosophila, in which Rh1(P37H) (the equivalent of mammalian Rh(P23H)) is expressed in PNs, and found that the endogenous Rh1 is required for Rh1(P37H) toxicity. Genetic inactivation of VCP increased the levels of misfolded Rh1(P37H) and further activated the Ire1/Xbp1 ER stress pathway in the Rh1(P37H) retina. Despite this, Rh1(P37H) flies with decreased VCP function displayed a potent suppression of retinal degeneration and blindness, indicating that VCP activity promotes neurodegeneration in the Rh1(P37H) retina. Pharmacological treatment of Rh1(P37H) flies with the VCP/ERAD inhibitor Eeyarestatin I or with the proteasome inhibitor MG132 also led to a strong suppression of retinal degeneration. Collectively, our findings raise the possibility that excessive retrotranslocation and/or degradation of visual pigment is a primary cause of PN degeneration.
Collapse
Affiliation(s)
- Ana Griciuc
- Department of Protein Science, Helmholtz Zentrum Muenchen-German Research Center for Environmental Health, Neuherberg, Germany
| | - Liviu Aron
- Department of Molecular Neurobiology, Max-Planck-Institute of Neurobiology, Martinsried, Germany
| | - Michel J. Roux
- Department of Neurobiology and Genetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, Illkirch, France
| | - Rüdiger Klein
- Department of Molecular Neurobiology, Max-Planck-Institute of Neurobiology, Martinsried, Germany
| | - Angela Giangrande
- Department of Cell and Developmental Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/ULP, Illkirch, France
| | - Marius Ueffing
- Department of Protein Science, Helmholtz Zentrum Muenchen-German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
26
|
Babizhayev MA. Designation of imidazole-containing dipeptides as pharmacological chaperones. Hum Exp Toxicol 2010; 30:736-61. [PMID: 20656726 DOI: 10.1177/0960327110377526] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
We review the dichotomous regulatory roles of natural imidazole-containing peptidomimetics (N-acetylcarnosine [NAC], carcinine, non-hydrolized carnosine) in maintaining skin homeostasis that determines whether keratinocytes survive or undergo apoptosis in response to various insults and in the development of skin diseases. General strategies addressing common ground techniques to improve absorption of usually active chaperone proteins or their dipeptide inducer (usually poorly absorbed) compounds include encapsulation into hydrophobic carriers, combination with penetration enhancers, active electrical transport or chemical modification to increase hydrophobicity. A growing evidence is presented that demonstrates the ability of NAC (lubricant eye drops) or carcinine to act as pharmacological chaperones, or being synergistically coupled in patented formulations with another imidazole-containing peptidomimetic (such as, Leucyl-histidylhydrazide), to decrease oxidative stress and ameliorate oxidative and excessive glycation stress-related eye disease phenotypes, suggesting that the field of chaperone therapy might hold novel treatments for age-related cataracts, glaucoma, age-related macular degeneration (AMD), and ocular complications of diabetes (OCD). Current efforts are being directed towards exploring therapeutic approaches of pharmacological targeting and human drug delivery for chaperone molecules based on innovative patented strategies.
Collapse
Affiliation(s)
- Mark A Babizhayev
- Innovative Vision Products Inc, County of New Castle, Delaware, USA.
| |
Collapse
|
27
|
Restoration of visual function in P23H rhodopsin transgenic rats by gene delivery of BiP/Grp78. Proc Natl Acad Sci U S A 2010; 107:5961-6. [PMID: 20231467 DOI: 10.1073/pnas.0911991107] [Citation(s) in RCA: 229] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The P23H mutation within the rhodopsin gene (RHO) causes rhodopsin misfolding, endoplasmic reticulum (ER) stress, and activates the unfolded protein response (UPR), leading to rod photoreceptor degeneration and autosomal dominant retinitis pigmentosa (ADRP). Grp78/BiP is an ER-localized chaperone that is induced by UPR signaling in response to ER stress. We have previously demonstrated that BiP mRNA levels are selectively reduced in animal models of ADRP arising from P23H rhodopsin expression at ages that precede photoreceptor degeneration. We have now overexpressed BiP to test the hypothesis that this chaperone promotes the trafficking of P23H rhodopsin to the cell membrane, reprograms the UPR favoring the survival of photoreceptors, blocks apoptosis, and, ultimately, preserves vision in ADRP rats. In cell culture, increasing levels of BiP had no impact on the localization of P23H rhodopsin. However, BiP overexpression alleviated ER stress by reducing levels of cleaved pATF6 protein, phosphorylated eIF2alpha and the proapoptotic protein CHOP. In P23H rats, photoreceptor levels of cleaved ATF6, pEIF2alpha, CHOP, and caspase-7 were much higher than those of wild-type rats. Subretinal delivery of AAV5 expressing BiP to transgenic rats led to reduction in CHOP and photoreceptor apoptosis and to a sustained increase in electroretinogram amplitudes. We detected complexes between BiP, caspase-12, and the BH3-only protein BiK that may contribute to the antiapoptotic activity of BiP. Thus, the preservation of photoreceptor function resulting from elevated levels of BiP is due to suppression of apoptosis rather than to a promotion of rhodopsin folding.
Collapse
|
28
|
Conformational diseases: looking into the eyes. Brain Res Bull 2010; 81:12-24. [PMID: 19808079 DOI: 10.1016/j.brainresbull.2009.09.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 09/11/2009] [Accepted: 09/29/2009] [Indexed: 01/09/2023]
Abstract
Conformational diseases, a general term comprising more than 40 disorders are caused by the accumulation of unfolded or misfolded proteins. Improper protein folding (misfolding) as well as accrual of unfolded proteins can lead to the formation of disordered (amorphous) or ordered (amyloid fibril) aggregates. The gradual accumulation of protein aggregates and the acceleration of their formation by stress explain the characteristic late or episodic onset of the diseases. The best studied in this group are neurodegenerative diseases and amyloidosis accompanied by the deposition of a specific aggregation-prone proteins or protein fragments and formation of insoluble fibrils. Amyloidogenic protein accumulation often occurs in the brain tissues, e.g. in Alzheimer's disease with the deposition of amyloid-beta and Tau, in scrapie and bovine spongiform encephalopathy with the accumulation of prion protein, in Parkinson's disease with the deposition of alpha-synuclein. Other examples of amyloid proteins are transthyretin, immunoglobulin light chain, gelsolin, etc. In addition to the brain, the accumulation of unfolded or misfolded proteins leading to pathology takes place in a wide variety of organs and tissues, including different parts of the eye. The best studied ocular conformational diseases are cataract in the lens and retinitis pigmentosa in the retina, but accumulation of misfolded proteins also occurs in other parts of the eye causing various disorders. Furthermore, ocular manifestation of systemic amyloidosis often causes the deposition of amyloidogenic proteins in different ocular tissues. Here we present the data regarding naturally unfolded and misfolded proteins in eye tissues, their structure-function relationships, and molecular mechanisms underlying their involvement in diseases. We also summarize the etiology of ocular conformational diseases and discuss approaches to their treatment.
Collapse
|
29
|
Griciuc A, Aron L, Piccoli G, Ueffing M. Clearance of Rhodopsin(P23H) aggregates requires the ERAD effector VCP. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1803:424-34. [PMID: 20097236 DOI: 10.1016/j.bbamcr.2010.01.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2009] [Revised: 12/29/2009] [Accepted: 01/15/2010] [Indexed: 11/15/2022]
Abstract
Dominant mutations in the visual pigment Rhodopsin (Rh) cause retinitis pigmentosa (RP) characterized by progressive blindness and retinal degeneration. The most common Rh mutation, Rh(P23H) forms aggregates in the endoplasmic reticulum (ER) and impairs the proteasome; however, the mechanisms linking Rh aggregate formation to proteasome dysfunction and photoreceptor cell loss remain unclear. Using mammalian cell cultures, we provide the first evidence that misfolded Rh(P23H) is a substrate of the ERAD effector VCP, an ATP-dependent chaperone that extracts misfolded proteins from the ER and escorts them for proteasomal degradation. VCP co-localizes with misfolded Rh(P23H) in retinal cells and requires functional N-terminal and D1 ATPase domains to form a complex with Rh(P23H) aggregates. Furthermore, VCP uses its D2 ATPase activity to promote Rh(P23H) aggregate retrotranslocation and proteasomal delivery. Our results raise the possibility that modulation of VCP and ERAD activity might have potential therapeutic significance for RP.
Collapse
Affiliation(s)
- Ana Griciuc
- Department of Protein Science, Helmholtz Zentrum Muenchen-German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | | | | | | |
Collapse
|
30
|
Pharmacological manipulation of rhodopsin retinitis pigmentosa. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 664:317-23. [PMID: 20238031 DOI: 10.1007/978-1-4419-1399-9_36] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Mutations in rhodopsin cause autosomal dominant retinitis pigmentosa. The majority of these mutations (class II) lead to protein misfolding. The misfolded protein is retained in the ER then retrotranslocated into the cytoplasm for degradation by the proteasome. If degradation fails, the protein can aggregate to form intracellular inclusions. In addition, the mutant rod opsin exerts a dominant negative effect on the wild-type protein. Here, we review these pathways and how different drug treatments can affect mutant rod opsin. Interestingly, drugs targeted at general protein stability (kosmotropes) or improving the cellular folding and degradation machinery (molecular chaperone inducers and autophagy induction) reduced P23H rod opsin aggregation and inclusion formation together with associated caspase activation and cell death, but did not enhance mutant protein processing or reduce the dominant negative effects. In contrast, pharmacological chaperones (retinoids) enhanced P23H folding and reduced the dominant negative effects, as well as reducing the other gains of function. Therefore, targeting the toxic gain of function did not require improved folding, whereas reducing the dominant negative effects required improved folding. These studies suggest that some forms of rhodopsin retinitis pigmentosa could be treated by targeting protein folding and/or reducing protein aggregation.
Collapse
|
31
|
Bondarenko VA, Hayashi F, Usukura J, Yamazaki A. Involvement of rhodopsin and ATP in the activation of membranous guanylate cyclase in retinal photoreceptor outer segments (ROS-GC) by GC-activating proteins (GCAPs): a new model for ROS-GC activation and its link to retinal diseases. Mol Cell Biochem 2009; 334:125-39. [PMID: 19941040 DOI: 10.1007/s11010-009-0323-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2008] [Accepted: 11/04/2009] [Indexed: 11/27/2022]
Abstract
Membranous guanylate cyclase in retinal photoreceptor outer segments (ROS-GC), a key enzyme for the recovery of photoreceptors to the dark state, has a topology identical to and cytoplasmic domains homologous to those of peptide-regulated GCs. However, under the prevailing concept, its activation mechanism is significantly different from those of peptide-regulated GCs: GC-activating proteins (GCAPs) function as the sole activator of ROS-GC in a Ca(2+)-sensitive manner, and neither reception of an outside signal by the extracellular domain (ECD) nor ATP binding to the kinase homology domain (KHD) is required for its activation. We have recently shown that ATP pre-binding to the KHD in ROS-GC drastically enhances its GCAP-stimulated activity, and that rhodopsin illumination, as the outside signal, is required for the ATP pre-binding. These results indicate that illuminated rhodopsin is involved in ROS-GC activation in two ways: to initiate ATP binding to ROS-GC for preparation of its activation and to reduce [Ca(2+)] through activation of cGMP phosphodiesterase. These two signal pathways are activated in a parallel and proportional manner and finally converge for strong activation of ROS-GC by Ca(2+)-free GCAPs. These results also suggest that the ECD receives the signal for ATP binding from illuminated rhodopsin. The ECD is projected into the intradiscal space, i.e., an intradiscal domain(s) of rhodopsin is also involved in the signal transfer. Many retinal disease-linked mutations are found in these intradiscal domains; however, their consequences are often unclear. This model will also provide novel insights into causal relationship between these mutations and certain retinal diseases.
Collapse
|
32
|
Sun X, Pawlyk B, Xu X, Liu X, Bulgakov OV, Adamian M, Sandberg MA, Khani SC, Tan MH, Smith AJ, Ali RR, Li T. Gene therapy with a promoter targeting both rods and cones rescues retinal degeneration caused by AIPL1 mutations. Gene Ther 2009; 17:117-31. [PMID: 19710705 PMCID: PMC2804971 DOI: 10.1038/gt.2009.104] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIPL1 is required for the biosynthesis of photoreceptor phosphodiesterase (PDE)1–3. Gene defects in AIPL1 cause a heterogeneous set of conditions ranging from Leber Congenital Amaurosis (LCA), the severest form of early-onset retinal degeneration, to milder forms such as retinitis pigmentosa (RP) and cone-rod dystrophy1,4,5. In mice, null and hypomorphic alleles cause retinal degeneration similar to human LCA and RP, respectively2,3,6. Thus these mouse models represent two ends of the disease spectrum associated with AIPL1 gene defects in humans. We evaluated whether adeno-associated virus (AAV)-mediated gene replacement therapy in these models could restore PDE biosynthesis in rods and cones and thereby improve photoreceptor survival. We validated the efficacy of human AIPL1 (isoform 1) replacement gene controlled by a promoter derived from the human rhodopsin kinase (RK) gene which is active in both rods and cones7. We found substantial and long-term rescue of the disease phenotype as a result of transgene expression. This is the first gene therapy study in which both rods and cones were targeted successfully with a single photoreceptor-specific promoter. We propose that the vector and construct design used in this study could serve as a prototype for a human clinical trial.
Collapse
Affiliation(s)
- X Sun
- Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Disrupted plasma membrane localization and loss of function reveal regions of human equilibrative nucleoside transporter 1 involved in structural integrity and activity. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:2326-34. [PMID: 19699178 DOI: 10.1016/j.bbamem.2009.08.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 07/16/2009] [Accepted: 08/12/2009] [Indexed: 01/08/2023]
Abstract
Human Equilibrative Nucleoside Transporter 1 (hENT1) is an integral membrane protein that transports nucleosides and analog drugs across cellular membranes. Very little is known about intracellular processing and localization of hENT1. Here we show that disruption of a highly conserved triplet (PWN) near the N-terminus, or the last eight C-terminal residues (two hydrophobic triplets separated by a positive arginine) result in loss of plasma membrane localization and/or transport function. To understand the role of specific residues within these regions, we studied the localization patterns of N- or C-terminal deletion and/or substitution mutants of GFP-hENT1 using confocal microscopy. Quantification of GFP-hENT1 (mutant and wildtype) protein at the plasma membrane was conducted using nitrobenzylthioinosine (NBTI) binding. Functionality of the GFP-hENT1 mutants was determined by heterologous expression in Xenopus laevis oocytes followed by measurement of uridine uptake. Mutation of the proline within the PWN motif disrupts plasma membrane localization. C-terminal mutations (primarily within the hydrophobic triplets) lead to hENT1 retention within the cell (e.g. in the ER). Some mutants still localize to the plasma membrane but show reduced transport activity. These data suggest that these two regions contribute to the structural integrity and thus correct processing and function of hENT1.
Collapse
|
34
|
Tan MH, Smith AJ, Pawlyk B, Xu X, Liu X, Bainbridge JB, Basche M, McIntosh J, Tran HV, Nathwani A, Li T, Ali RR. Gene therapy for retinitis pigmentosa and Leber congenital amaurosis caused by defects in AIPL1: effective rescue of mouse models of partial and complete Aipl1 deficiency using AAV2/2 and AAV2/8 vectors. Hum Mol Genet 2009; 18:2099-114. [PMID: 19299492 PMCID: PMC2722233 DOI: 10.1093/hmg/ddp133] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Accepted: 03/17/2009] [Indexed: 01/19/2023] Open
Abstract
Defects in the photoreceptor-specific gene encoding aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) are clinically heterogeneous and present as Leber Congenital Amaurosis, the severest form of early-onset retinal dystrophy and milder forms of retinal dystrophies such as juvenile retinitis pigmentosa and dominant cone-rod dystrophy. [Perrault, I., Rozet, J.M., Gerber, S., Ghazi, I., Leowski, C., Ducroq, D., Souied, E., Dufier, J.L., Munnich, A. and Kaplan, J. (1999) Leber congenital amaurosis. Mol. Genet. Metab., 68, 200-208.] Although not yet fully elucidated, AIPL1 is likely to function as a specialized chaperone for rod phosphodiesterase (PDE). We evaluate whether AAV-mediated gene replacement therapy is able to improve photoreceptor function and survival in retinal degeneration associated with AIPL1 defects. We used two mouse models of AIPL1 deficiency simulating three different rates of photoreceptor degeneration. The Aipl1 hypomorphic (h/h) mouse has reduced Aipl1 levels and a relatively slow degeneration. Under light acceleration, the rate of degeneration in the Aipl1 h/h mouse is increased by 2-3-fold. The Aipl1-/- mouse has no functional Aipl1 and has a very rapid retinal degeneration. To treat the different rates of degeneration, two pseudotypes of recombinant adeno-associated virus (AAV) exhibiting different transduction kinetics are used for gene transfer. We demonstrate restoration of cellular function and preservation of photoreceptor cells and retinal function in Aipl1 h/h mice following gene replacement therapy using an AAV2/2 vector and in the light accelerated Aipl1 h/h model and Aipl1-/- mice using an AAV2/8 vector. We have thus established the potential of gene replacement therapy in varying rates of degeneration that reflect the clinical spectrum of disease. This is the first gene replacement study to report long-term rescue of a photoreceptor-specific defect and to demonstrate effective rescue of a rapid photoreceptor degeneration.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Dependovirus/genetics
- Disease Models, Animal
- Genetic Therapy
- Genetic Vectors/genetics
- Humans
- Mice
- Mice, Transgenic
- Optic Atrophy, Hereditary, Leber/genetics
- Optic Atrophy, Hereditary, Leber/physiopathology
- Optic Atrophy, Hereditary, Leber/therapy
- Photoreceptor Cells, Vertebrate/metabolism
- Retinitis Pigmentosa/genetics
- Retinitis Pigmentosa/physiopathology
- Retinitis Pigmentosa/therapy
Collapse
Affiliation(s)
- Mei Hong Tan
- Institute of Ophthalmology, NIHR Biomedical research Centre, University College London, London, UK
| | - Alexander J. Smith
- Institute of Ophthalmology, NIHR Biomedical research Centre, University College London, London, UK
| | - Basil Pawlyk
- Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - Xiaoyun Xu
- Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - Xiaoqing Liu
- Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - James B. Bainbridge
- Institute of Ophthalmology, NIHR Biomedical research Centre, University College London, London, UK
| | - Mark Basche
- Institute of Ophthalmology, NIHR Biomedical research Centre, University College London, London, UK
| | - Jenny McIntosh
- Cancer Research Institute, University College London, London, UK
| | - Hoai Viet Tran
- Institute of Ophthalmology, NIHR Biomedical research Centre, University College London, London, UK
| | - Amit Nathwani
- Cancer Research Institute, University College London, London, UK
| | - Tiansen Li
- Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
| | - Robin R. Ali
- Institute of Ophthalmology, NIHR Biomedical research Centre, University College London, London, UK
| |
Collapse
|
35
|
Tan MH, Smith AJ, Pawlyk B, Xu X, Liu X, Bainbridge JB, Basche M, McIntosh J, Tran HV, Nathwani A, Li T, Ali RR. Gene therapy for retinitis pigmentosa and Leber congenital amaurosis caused by defects in AIPL1: effective rescue of mouse models of partial and complete Aipl1 deficiency using AAV2/2 and AAV2/8 vectors. Hum Mol Genet 2009. [PMID: 19299492 DOI: 10.1093/hgm/ddp133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Defects in the photoreceptor-specific gene encoding aryl hydrocarbon receptor-interacting protein-like 1 (AIPL1) are clinically heterogeneous and present as Leber Congenital Amaurosis, the severest form of early-onset retinal dystrophy and milder forms of retinal dystrophies such as juvenile retinitis pigmentosa and dominant cone-rod dystrophy. [Perrault, I., Rozet, J.M., Gerber, S., Ghazi, I., Leowski, C., Ducroq, D., Souied, E., Dufier, J.L., Munnich, A. and Kaplan, J. (1999) Leber congenital amaurosis. Mol. Genet. Metab., 68, 200-208.] Although not yet fully elucidated, AIPL1 is likely to function as a specialized chaperone for rod phosphodiesterase (PDE). We evaluate whether AAV-mediated gene replacement therapy is able to improve photoreceptor function and survival in retinal degeneration associated with AIPL1 defects. We used two mouse models of AIPL1 deficiency simulating three different rates of photoreceptor degeneration. The Aipl1 hypomorphic (h/h) mouse has reduced Aipl1 levels and a relatively slow degeneration. Under light acceleration, the rate of degeneration in the Aipl1 h/h mouse is increased by 2-3-fold. The Aipl1-/- mouse has no functional Aipl1 and has a very rapid retinal degeneration. To treat the different rates of degeneration, two pseudotypes of recombinant adeno-associated virus (AAV) exhibiting different transduction kinetics are used for gene transfer. We demonstrate restoration of cellular function and preservation of photoreceptor cells and retinal function in Aipl1 h/h mice following gene replacement therapy using an AAV2/2 vector and in the light accelerated Aipl1 h/h model and Aipl1-/- mice using an AAV2/8 vector. We have thus established the potential of gene replacement therapy in varying rates of degeneration that reflect the clinical spectrum of disease. This is the first gene replacement study to report long-term rescue of a photoreceptor-specific defect and to demonstrate effective rescue of a rapid photoreceptor degeneration.
Collapse
Affiliation(s)
- Mei Hong Tan
- Institute of Ophthalmology, NIHR Biomedical research Centre, University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kosmaoglou M, Schwarz N, Bett JS, Cheetham ME. Molecular chaperones and photoreceptor function. Prog Retin Eye Res 2008; 27:434-49. [PMID: 18490186 PMCID: PMC2568879 DOI: 10.1016/j.preteyeres.2008.03.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Molecular chaperones facilitate and regulate protein conformational
change within cells. This encompasses many fundamental cellular processes:
including the correct folding of nascent chains; protein transport and
translocation; signal transduction and protein quality control. Chaperones are,
therefore, important in several forms of human disease, including
neurodegeneration. Within the retina, the highly specialized photoreceptor cell
presents a fascinating paradigm to investigate the specialization of molecular
chaperone function and reveals unique chaperone requirements essential to
photoreceptor function. Mutations in several photoreceptor proteins lead to
protein misfolding mediated neurodegeneration. The best characterized of these
are mutations in the molecular light sensor, rhodopsin, which cause autosomal
dominant retinitis pigmentosa. Rhodopsin biogenesis is likely to require
chaperones, while rhodopsin misfolding involves molecular chaperones in quality
control and the cellular response to protein aggregation. Furthermore, the
specialization of components of the chaperone machinery to photoreceptor
specific roles has been revealed by the identification of mutations in molecular
chaperones that cause inherited retinal dysfunction and degeneration. These
chaperones are involved in several important cellular pathways and further
illuminate the essential and diverse roles of molecular
chaperones.
Collapse
Affiliation(s)
- Maria Kosmaoglou
- Division of Molecular and Cellular Neuroscience, UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1 V 9EL, UK
| | | | | | | |
Collapse
|
37
|
Müller DJ, Wu N, Palczewski K. Vertebrate membrane proteins: structure, function, and insights from biophysical approaches. Pharmacol Rev 2008; 60:43-78. [PMID: 18321962 DOI: 10.1124/pr.107.07111] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Membrane proteins are key targets for pharmacological intervention because they are vital for cellular function. Here, we analyze recent progress made in the understanding of the structure and function of membrane proteins with a focus on rhodopsin and development of atomic force microscopy techniques to study biological membranes. Membrane proteins are compartmentalized to carry out extra- and intracellular processes. Biological membranes are densely populated with membrane proteins that occupy approximately 50% of their volume. In most cases membranes contain lipid rafts, protein patches, or paracrystalline formations that lack the higher-order symmetry that would allow them to be characterized by diffraction methods. Despite many technical difficulties, several crystal structures of membrane proteins that illustrate their internal structural organization have been determined. Moreover, high-resolution atomic force microscopy, near-field scanning optical microscopy, and other lower resolution techniques have been used to investigate these structures. Single-molecule force spectroscopy tracks interactions that stabilize membrane proteins and those that switch their functional state; this spectroscopy can be applied to locate a ligand-binding site. Recent development of this technique also reveals the energy landscape of a membrane protein, defining its folding, reaction pathways, and kinetics. Future development and application of novel approaches during the coming years should provide even greater insights to the understanding of biological membrane organization and function.
Collapse
Affiliation(s)
- Daniel J Müller
- Biotechnology Center, University of Technology, Dresden, Germany
| | | | | |
Collapse
|
38
|
Roberts L, Rebello G, Ramesar R, Greenberg J. Management of a South African family with retinitis pigmentosa-should potential therapy influence translational research protocols? J Ocul Biol Dis Infor 2008; 1:55-8. [PMID: 20072635 PMCID: PMC2802413 DOI: 10.1007/s12177-008-9001-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Accepted: 02/13/2008] [Indexed: 11/28/2022] Open
Abstract
Mutation analysis of retinal candidate genes is performed as part of an ongoing research to identify the causative genetic defect in South African families with retinal degenerative disorders (RDDs). A translational research protocol has been established whereby probands are counseled and given their molecular genetic results to take back to other family members, who can then request individual diagnostic testing. A Thr17Met mutation of the rhodopsin gene was identified in a Caucasian South African family with autosomal dominant retinitis pigmentosa. Patients with this mutation appear to benefit from treatment using oral vitamin A supplementation. This family has been informed that a molecular diagnosis is available; however, one individual has refused testing and none of the younger generation has shown interest in receiving molecular results or genetic counseling. Adapting the established protocol for the translation of RDD research results and contacting mutation positive individuals may be justifiable in light of the potential benefit of therapy.
Collapse
|
39
|
Rohrer B, Crouch R. Rod and cone pigment regeneration in RPE65-/- mice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 572:101-7. [PMID: 17249562 DOI: 10.1007/0-387-32442-9_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Affiliation(s)
- Baerbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | |
Collapse
|
40
|
Van der Spuy J, Cheetham ME. The chaperone function of the LCA protein AIPL1. AIPL1 chaperone function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 572:471-6. [PMID: 17249611 DOI: 10.1007/0-387-32442-9_65] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
|
41
|
Organisciak D, Darrow R, Gu X, Barsalou L, Crabb JW. Genetic, age and light mediated effects on crystallin protein expression in the retina. Photochem Photobiol 2007; 82:1088-96. [PMID: 16602829 DOI: 10.1562/2005-06-30-ra-599] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To probe for possible relationships between retinal crystallins and retinal degenerations, protein expression was compared in normal Sprague-Dawley rats, treated or not with intense light, Royal College of Surgeons (RCS) rats and transgenic rats expressing rhodopsin mutations. Rats were reared in dim cyclic light for 21-75 days. Photoreceptor cell DNA levels were determined at various ages to assess the rates of visual cell loss. 1D- and 2D-gel electrophoresis was used to profile retinal protein expression. Crystallins were identified by western analysis and by tandem mass spectrometry. In normal rat retinas, alpha, beta and gamma crystallins were present, although alphaA- and gamma-crystallins exhibited some increase with age. As measured by DNA levels, the rate of genetically induced photoreceptor cell loss was greater in rats with faster degenerating retinas (RCS, S334-ter Line 4, P23H Line 3) than in rats with slower degenerating retinas (S334-ter Line 9, P23H Line 2). In genetic models of retinal degeneration increased levels of immunoreactivity for all crystallins, especially alphaA-insert, correlated with the different rates of photoreceptor loss. In the light induced degeneration model alphaA-insert was unchanged, truncated alphaB-crystallin levels were increased and gamma-crystallins were greatly reduced. In the RCS rat retina 16 different crystallins were identified. Our data suggests that an increase in crystallin expression occurs during various retinal degenerations and that the increases may be related to the severity, type and onset of retinal degeneration.
Collapse
Affiliation(s)
- Daniel Organisciak
- Petticrew Research Laboratory, School of Medicine, Wright State University, Dayton, OH, USA.
| | | | | | | | | |
Collapse
|
42
|
Surguchov A. Focus on molecules: the synucleins: "When friends become foes". Exp Eye Res 2006; 86:1-2. [PMID: 17049462 DOI: 10.1016/j.exer.2006.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 09/01/2006] [Accepted: 09/06/2006] [Indexed: 11/26/2022]
Affiliation(s)
- Andrei Surguchov
- Retinal Biology Laboratory, VA Medical Center, Kansas City, 4801 Linwood blvd, Kansas City, MO 64128, USA.
| |
Collapse
|
43
|
Abstract
A large number of neurodegenerative diseases in humans result from protein misfolding and aggregation. Protein misfolding is believed to be the primary cause of Alzheimer's disease, Parkinson's disease, Huntington's disease, Creutzfeldt-Jakob disease, cystic fibrosis, Gaucher's disease and many other degenerative and neurodegenerative disorders. Cellular molecular chaperones, which are ubiquitous, stress-induced proteins, and newly found chemical and pharmacological chaperones have been found to be effective in preventing misfolding of different disease-causing proteins, essentially reducing the severity of several neurodegenerative disorders and many other protein-misfolding diseases. In this review, we discuss the probable mechanisms of several protein-misfolding diseases in humans, as well as therapeutic approaches for countering them. The role of molecular, chemical and pharmacological chaperones in suppressing the effect of protein misfolding-induced consequences in humans is explained in detail. Functional aspects of the different types of chaperones suggest their uses as potential therapeutic agents against different types of degenerative diseases, including neurodegenerative disorders.
Collapse
Affiliation(s)
- Tapan K Chaudhuri
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, India.
| | | |
Collapse
|
44
|
Zhu L, Imanishi Y, Filipek S, Alekseev A, Jastrzebska B, Sun W, Saperstein DA, Palczewski K. Autosomal recessive retinitis pigmentosa and E150K mutation in the opsin gene. J Biol Chem 2006; 281:22289-22298. [PMID: 16737970 PMCID: PMC1618956 DOI: 10.1074/jbc.m602664200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Retinitis pigmentosa (RP) is a heterogeneous group of hereditary disorders of the retina caused by mutation in genes of the photoreceptor proteins with an autosomal dominant (adRP), autosomal recessive (arRP), or X-linked pattern of inheritance. Although there are over 100 identified mutations in the opsin gene associated with RP, only a few of them are inherited with the arRP pattern. E150K is the first reported missense mutation associated with arRP. This opsin mutation is located in the second cytoplasmic loop of this G protein-coupled receptor. E150K opsin expressed in HEK293 cells and reconstituted with 11-cis-retinal displayed an absorption spectrum similar to the wild type (WT) counterpart and activated G protein transducin slightly faster than WT receptor. However, the majority of E150K opsin showed a higher apparent molecular mass in SDS-PAGE and was resistant to endoglycosidase H deglycosidase. Instead of being transported to the plasma membrane, E150K opsin is partially colocalized with the cis/medial Golgi compartment markers such as GM130 and Vti1b but not with the trans-Golgi network. In contrast to the endoplasmic reticulum-retained adRP mutant, P23H opsin, Golgi-retained E150K opsin did not influence the proper transport of the WT opsin when coexpressed in HEK293 cells. This result is consistent with the recessive pattern of inheritance of this mutation. Thus, our study reveals a novel molecular mechanism for retinal degeneration that results from deficient export of opsin from the Golgi apparatus.
Collapse
Affiliation(s)
- Li Zhu
- Department of Chemistry, University of Washington, Seattle, Washington 98195
| | - Yoshikazu Imanishi
- Department of Pharmacology, Case School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Sławomir Filipek
- International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Andrei Alekseev
- Department of Ophthalmology, University of Washington, Seattle, Washington 98195
| | - Beata Jastrzebska
- Department of Pharmacology, Case School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - Wenyu Sun
- Department of Pharmacology, Case School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106
| | - David A Saperstein
- Department of Ophthalmology, University of Washington, Seattle, Washington 98195
| | - Krzysztof Palczewski
- Department of Pharmacology, Case School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106.
| |
Collapse
|
45
|
Banh A, Bantseev V, Choh V, Moran KL, Sivak JG. The lens of the eye as a focusing device and its response to stress. Prog Retin Eye Res 2006; 25:189-206. [PMID: 16330238 DOI: 10.1016/j.preteyeres.2005.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The continued peripheral growth of the lens, resulting in the concentration of older tissue toward the center, has the important optical consequence of producing a lens of variable refractive index. An approach consisting of the projection of fine laser beams through excised lenses in physiological solution has been used for in vitro study of lens optical quality. By varying the separation of the incident beams and/or the wavelength characteristics of the laser used, lens refractive properties and relative transparency may be examined. In the review provided, these optical properties are correlated to lens suture anatomy, lens mitochondrial morphology and function and the function of lens heat shock proteins. In addition, lens spherical aberration is evaluated as a function of accommodation. This work can be highlighted as follows: Mammalian lens suture morphology has a direct impact on lens optical function and, while suture structure of mammalian and avian lenses are very different, they both show an age-related deterioration in morphology and focusing ability. The distribution and appearance of mitochondria of the lens epithelium and superficial fiber cells are similar in all vertebrates. Lens mitochondrial integrity is correlated to lens focusing ability, suggesting a correlation between lens optical properties and lens metabolic function. The induction of cold cataract measured optically in cultured mammalian lenses is enhanced by thermal (heat) shock and this effect is prevented by inhibiting heat shock protein production. Finally, lens accommodative function can be studied by measuring lens refractive change using a physiological model involving an intact accommodative apparatus.
Collapse
Affiliation(s)
- Alice Banh
- School of Optometry, University of Waterloo, Waterloo, Ontario, Canada N2L 3G1
| | | | | | | | | |
Collapse
|
46
|
Affiliation(s)
- Alberto J L Macario
- Wadsworth Center, Division of Molecular Medicine, New York State Department of Health, Albany, NY 12201-0509, USA.
| | | |
Collapse
|
47
|
Mendes HF, van der Spuy J, Chapple JP, Cheetham ME. Mechanisms of cell death in rhodopsin retinitis pigmentosa: implications for therapy. Trends Mol Med 2005; 11:177-85. [PMID: 15823756 DOI: 10.1016/j.molmed.2005.02.007] [Citation(s) in RCA: 275] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Retinitis pigmentosa (RP) is a group of retinal degenerative diseases that are characterised primarily by the loss of rod photoreceptor cells. Mutations in rhodopsin are the most common cause of autosomal-dominant RP (ADRP). Here, we propose a new classification for rhodopsin mutations based on their biochemical and cellular properties. Several different potential gain-of-function mechanisms for rhodopsin ADRP are described and discussed. Possible dominant-negative mechanisms, which affect the processing, translocation or degradation of wild-type rhodopsin, are also considered. Understanding the molecular and cellular consequences of rod-opsin mutations and the underlying disease mechanisms in ADRP are essential to develop future therapies for this class of retinal dystrophies.
Collapse
Affiliation(s)
- Hugo F Mendes
- Division of Pathology, Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | | | | | | |
Collapse
|
48
|
Ben-Arie-Weintrob Y, Berson EL, Dryja TP. Histopathologic-genotypic correlations in retinitis pigmentosa and allied diseases. Ophthalmic Genet 2005; 26:91-100. [PMID: 16020312 DOI: 10.1080/13816810590968032] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
This paper reviews the published histopathologic findings of patients with retinitis pigmentosa (RP) or an allied disease in whom the responsible gene defect was identified, including 10 cases with dominant RP (cases with mutations in RHO, PRPC8, and RP1), three with dominant spinocerebellar ataxia (SCA7), three X-linked RP carrier females (RPGR), two with congenital retinal blindness (AIPL1 and RPE65), two with mitochondrial encephalomyopathy overlap syndrome (MTTL1), and one case each with dominant cone degeneration (GCAP1), X-linked cone degeneration (RCP), enhanced S-cone syndrome (NR2E3), and dominant late-onset retinal degeneration (CTRP5). No histopathologic descriptions were found of the vast majority of genetically defined forms of retinal degeneration.
Collapse
Affiliation(s)
- Yael Ben-Arie-Weintrob
- The Cogan Eye Pathology Laboratory and the Berman-Gund Laboratory for the Study of Retinal Degenerations, Harvard Medical School, Massachusetts Eye & Ear Infirmary, 243 Charles Street, Boston, MA 02114, U.S.A
| | | | | |
Collapse
|
49
|
Surgucheva I, Ninkina N, Buchman VL, Grasing K, Surguchov A. Protein aggregation in retinal cells and approaches to cell protection. Cell Mol Neurobiol 2005; 25:1051-66. [PMID: 16392036 DOI: 10.1007/s10571-005-8474-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Accepted: 04/21/2005] [Indexed: 10/25/2022]
Abstract
1. Retinal dystrophies (RD) comprise a group of clinically and genetically heterogeneous retinal disorders, which typically result in the degeneration of photoreceptors followed by the impairment or loss of vision. Although age-related macular degeneration (AMD) and retinitis pigmentosa (RP) are among the most common forms of RD, currently, there is no effective treatment for either disorder. 2. Recently, abnormal protein accumulation and aggregation due to protein misfolding and proteasome inhibition have been implicated in the pathogenesis of RD. In this paper we describe effects of several factors on protein aggregation and survival of photoreceptor cells. 3. Expression of rhodopsin carrying P23H mutation causes its accumulation in intracellular inclusion bodies in a perinuclear area of photoreceptor cells. beta- and gamma-synucleins and heat shock protein Hsp-70, but not alpha-synuclein, protect cultured ocular cells from mutant opsin accumulation. This effect might be explained by their chaperonic activity. 4. Knock-out of alpha- and gamma-synucleins does not affect gross retinal morphology, but induces tyrosine hydroxylase in the inner prexiform layer of the retina. Selegiline-a monoamine oxidase inhibitor used for the treatment of Parkinson's disease, reduces apoptosis and increases viability in cultured retinal pigment epithelium cells (APRE-19). 5. These results suggest that chaperones and selegiline may be considered promising candidates for the protection of ocular cells from the accumulation of misfolded and aggregated proteins.
Collapse
Affiliation(s)
- Irina Surgucheva
- Retinal Disease Research Laboratory, Veterans Administration Medical Center, Kansas City, Missouri 66148, USA
| | | | | | | | | |
Collapse
|
50
|
Macario AJL, Grippo TM, Conway de Macario E. Genetic disorders involving molecular-chaperone genes: a perspective. Genet Med 2005; 7:3-12. [PMID: 15654222 DOI: 10.1097/01.gim.0000151351.11876.c3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Molecular chaperones are important for maintaining a functional set of proteins in all cellular compartments. Together with protein degradation machineries (e.g., the ubiquitin-proteasome system), chaperones form the core of the cellular protein-quality control mechanism. Chaperones are proteins, and as such, they can be affected by mutations. At least 15 disorders have been identified that are associated with mutations in genes encoding chaperones, or molecules with features suggesting that they function as chaperones. These chaperonopathies and a few other candidates are presented in this article. In most cases, the mechanisms by which the defective genes contribute to the observed phenotypes are still uncharacterized. However, the reported observations definitely point to the possibility that abnormal chaperones participate in pathogenesis. The available data open novel perspectives and should encourage searches for new genetic chaperonopathies, as well as further analyses of the disorders discussed in this article, including detection of new cases.
Collapse
Affiliation(s)
- Alberto J L Macario
- Wadsworth Center, Division of Molecular Medicine, New York State Department of Health, Albany, New York, USA
| | | | | |
Collapse
|