1
|
Damodar K, Dubois G, Guillou L, Mamaeva D, Pequignot M, Erkilic N, Sanjurjo-Soriano C, Boukhaddaoui H, Bernex F, Bocquet B, Vialaret J, Arsenijevic Y, Redmond TM, Hirtz C, Meunier I, Brabet P, Kalatzis V. Dual CRALBP isoforms unveiled: iPSC-derived retinal modeling and AAV2/5-RLBP1 gene transfer raise considerations for effective therapy. Mol Ther 2024:S1525-0016(24)00664-6. [PMID: 39385467 DOI: 10.1016/j.ymthe.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/05/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Inherited retinal diseases (IRDs) are characterized by progressive vision loss. There are over 270 causative IRD genes, and variants within the same gene can cause clinically distinct disorders. One example is RLBP1, which encodes CRALBP. CRALBP is an essential protein in the rod and cone visual cycles that take place primarily in the retinal pigment epithelium (RPE) but also in Müller cells of the neuroretina. RLBP1 variants lead to three clinical subtypes: Bothnia dystrophy, retinitis punctata albescens, and Newfoundland rod-cone dystrophy. We modeled RLBP1-IRD subtypes using patient-specific induced pluripotent stem cell (iPSC)-derived RPE and identified pathophysiological markers that served as pertinent therapeutic read-outs. We developed an AAV2/5-mediated gene-supplementation strategy and performed a proof-of-concept study in the human models, which was validated in vivo in an Rlbp1-/- murine model. Most importantly, we identified a previously unsuspected smaller CRALBP isoform that is naturally and differentially expressed both in the human and murine retina. This previously unidentified isoform is produced from an alternative methionine initiation site. This work provides further insights into CRALBP expression and RLBP1-associated pathophysiology and raises important considerations for successful gene-supplementation therapy.
Collapse
Affiliation(s)
- Krishna Damodar
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France
| | - Gregor Dubois
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France
| | - Laurent Guillou
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France
| | - Daria Mamaeva
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France
| | - Marie Pequignot
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France
| | - Nejla Erkilic
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France; National Reference Centre for Inherited Sensory Diseases, University of Montpellier, Montpellier University Hospital, 34090 Montpellier, France
| | - Carla Sanjurjo-Soriano
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France
| | - Hassan Boukhaddaoui
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France
| | - Florence Bernex
- RHEM, Réseau d'Histologie Expérimentale de Montpellier, University of Montpellier, Biocampus, CNRS, Inserm, 34298 Montpellier, France
| | - Béatrice Bocquet
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France; National Reference Centre for Inherited Sensory Diseases, University of Montpellier, Montpellier University Hospital, 34090 Montpellier, France
| | - Jérôme Vialaret
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France; IRMB-PPC, INM, Montpellier University Hospital, Inserm, CNRS, University of Montpellier, 34295 Montpellier, France
| | - Yvan Arsenijevic
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, 1004 Lausanne, Switzerland
| | - T Michael Redmond
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christopher Hirtz
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France; IRMB-PPC, INM, Montpellier University Hospital, Inserm, CNRS, University of Montpellier, 34295 Montpellier, France
| | - Isabelle Meunier
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France; National Reference Centre for Inherited Sensory Diseases, University of Montpellier, Montpellier University Hospital, 34090 Montpellier, France
| | - Philippe Brabet
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France
| | - Vasiliki Kalatzis
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, Inserm, 34091 Montpellier, France; National Reference Centre for Inherited Sensory Diseases, University of Montpellier, Montpellier University Hospital, 34090 Montpellier, France.
| |
Collapse
|
2
|
Seah I, Goh D, Banerjee A, Su X. Modeling inherited retinal diseases using human induced pluripotent stem cell derived photoreceptor cells and retinal pigment epithelial cells. Front Med (Lausanne) 2024; 11:1328474. [PMID: 39011458 PMCID: PMC11246861 DOI: 10.3389/fmed.2024.1328474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 06/18/2024] [Indexed: 07/17/2024] Open
Abstract
Since the discovery of induced pluripotent stem cell (iPSC) technology, there have been many attempts to create cellular models of inherited retinal diseases (IRDs) for investigation of pathogenic processes to facilitate target discovery and validation activities. Consistency remains key in determining the utility of these findings. Despite the importance of consistency, quality control metrics are still not widely used. In this review, a toolkit for harnessing iPSC technology to generate photoreceptor, retinal pigment epithelial cell, and organoid disease models is provided. Considerations while developing iPSC-derived IRD models such as iPSC origin, reprogramming methods, quality control metrics, control strategies, and differentiation protocols are discussed. Various iPSC IRD models are dissected and the scientific hurdles of iPSC-based disease modeling are discussed to provide an overview of current methods and future directions in this field.
Collapse
Affiliation(s)
- Ivan Seah
- Translational Retinal Research Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Debbie Goh
- Department of Ophthalmology, National University Hospital (NUH), Singapore, Singapore
| | - Animesh Banerjee
- Translational Retinal Research Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xinyi Su
- Translational Retinal Research Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Ophthalmology, National University Hospital (NUH), Singapore, Singapore
- Singapore Eye Research Institute (SERI), Singapore, Singapore
| |
Collapse
|
3
|
Raeker MÖ, Perera ND, Karoukis AJ, Chen L, Feathers KL, Ali RR, Thompson DA, Fahim AT. Reduced Retinal Pigment Epithelial Autophagy Due to Loss of Rab12 Prenylation in a Human iPSC-RPE Model of Choroideremia. Cells 2024; 13:1068. [PMID: 38920696 PMCID: PMC11201631 DOI: 10.3390/cells13121068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/31/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
Choroideremia is an X-linked chorioretinal dystrophy caused by mutations in CHM, encoding Rab escort protein 1 (REP-1), leading to under-prenylation of Rab GTPases (Rabs). Despite ubiquitous expression of CHM, the phenotype is limited to degeneration of the retina, retinal pigment epithelium (RPE), and choroid, with evidence for primary pathology in RPE cells. However, the spectrum of under-prenylated Rabs in RPE cells and how they contribute to RPE dysfunction remain unknown. A CRISPR/Cas-9-edited CHM-/- iPSC-RPE model was generated with isogenic control cells. Unprenylated Rabs were biotinylated in vitro and identified by tandem mass tag (TMT) spectrometry. Rab12 was one of the least prenylated and has an established role in suppressing mTORC1 signaling and promoting autophagy. CHM-/- iPSC-RPE cells demonstrated increased mTORC1 signaling and reduced autophagic flux, consistent with Rab12 dysfunction. Autophagic flux was rescued in CHM-/- cells by transduction with gene replacement (ShH10-CMV-CHM) and was reduced in control cells by siRNA knockdown of Rab12. This study supports Rab12 under-prenylation as an important cause of RPE cell dysfunction in choroideremia and highlights increased mTORC1 and reduced autophagy as potential disease pathways for further investigation.
Collapse
Affiliation(s)
- Maide Ö. Raeker
- Department of Ophthalmology, University of Michigan, Ann Arbor, MI 48105, USA; (M.Ö.R.); (N.D.P.); (A.J.K.); (K.L.F.); (R.R.A.); (D.A.T.)
| | - Nirosha D. Perera
- Department of Ophthalmology, University of Michigan, Ann Arbor, MI 48105, USA; (M.Ö.R.); (N.D.P.); (A.J.K.); (K.L.F.); (R.R.A.); (D.A.T.)
| | - Athanasios J. Karoukis
- Department of Ophthalmology, University of Michigan, Ann Arbor, MI 48105, USA; (M.Ö.R.); (N.D.P.); (A.J.K.); (K.L.F.); (R.R.A.); (D.A.T.)
| | - Lisheng Chen
- Department of Orthopedic Surgery, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Kecia L. Feathers
- Department of Ophthalmology, University of Michigan, Ann Arbor, MI 48105, USA; (M.Ö.R.); (N.D.P.); (A.J.K.); (K.L.F.); (R.R.A.); (D.A.T.)
| | - Robin R. Ali
- Department of Ophthalmology, University of Michigan, Ann Arbor, MI 48105, USA; (M.Ö.R.); (N.D.P.); (A.J.K.); (K.L.F.); (R.R.A.); (D.A.T.)
- KCL Center for Cell and Gene Therapy, London WC2R 2LS, UK
| | - Debra A. Thompson
- Department of Ophthalmology, University of Michigan, Ann Arbor, MI 48105, USA; (M.Ö.R.); (N.D.P.); (A.J.K.); (K.L.F.); (R.R.A.); (D.A.T.)
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Abigail T. Fahim
- Department of Ophthalmology, University of Michigan, Ann Arbor, MI 48105, USA; (M.Ö.R.); (N.D.P.); (A.J.K.); (K.L.F.); (R.R.A.); (D.A.T.)
| |
Collapse
|
4
|
Kurzawa-Akanbi M, Tzoumas N, Corral-Serrano JC, Guarascio R, Steel DH, Cheetham ME, Armstrong L, Lako M. Pluripotent stem cell-derived models of retinal disease: Elucidating pathogenesis, evaluating novel treatments, and estimating toxicity. Prog Retin Eye Res 2024; 100:101248. [PMID: 38369182 DOI: 10.1016/j.preteyeres.2024.101248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/20/2024]
Abstract
Blindness poses a growing global challenge, with approximately 26% of cases attributed to degenerative retinal diseases. While gene therapy, optogenetic tools, photosensitive switches, and retinal prostheses offer hope for vision restoration, these high-cost therapies will benefit few patients. Understanding retinal diseases is therefore key to advance effective treatments, requiring in vitro models replicating pathology and allowing quantitative assessments for drug discovery. Pluripotent stem cells (PSCs) provide a unique solution given their limitless supply and ability to differentiate into light-responsive retinal tissues encompassing all cell types. This review focuses on the history and current state of photoreceptor and retinal pigment epithelium (RPE) cell generation from PSCs. We explore the applications of this technology in disease modelling, experimental therapy testing, biomarker identification, and toxicity studies. We consider challenges in scalability, standardisation, and reproducibility, and stress the importance of incorporating vasculature and immune cells into retinal organoids. We advocate for high-throughput automation in data acquisition and analyses and underscore the value of advanced micro-physiological systems that fully capture the interactions between the neural retina, RPE, and choriocapillaris.
Collapse
|
5
|
Sanjurjo-Soriano C, Jimenez-Medina C, Erkilic N, Cappellino L, Lefevre A, Nagel-Wolfrum K, Wolfrum U, Van Wijk E, Roux AF, Meunier I, Kalatzis V. USH2A variants causing retinitis pigmentosa or Usher syndrome provoke differential retinal phenotypes in disease-specific organoids. HGG ADVANCES 2023; 4:100229. [PMID: 37654703 PMCID: PMC10465966 DOI: 10.1016/j.xhgg.2023.100229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
There is an emblematic clinical and genetic heterogeneity associated with inherited retinal diseases (IRDs). The most common form is retinitis pigmentosa (RP), a rod-cone dystrophy caused by pathogenic variants in over 80 different genes. Further complexifying diagnosis, different variants in individual RP genes can also alter the clinical phenotype. USH2A is the most prevalent gene for autosomal-recessive RP and one of the most challenging because of its large size and, hence, large number of variants. Moreover, USH2A variants give rise to non-syndromic and syndromic RP, known as Usher syndrome (USH) type 2, which is associated with vision and hearing loss. The lack of a clear genotype-phenotype correlation or prognostic models renders diagnosis highly challenging. We report here a long-awaited differential non-syndromic RP and USH phenotype in three human disease-specific models: fibroblasts, induced pluripotent stem cells (iPSCs), and mature iPSC-derived retinal organoids. Moreover, we identified distinct retinal phenotypes in organoids from multiple RP and USH individuals, which were validated by isogenic-corrected controls. Non-syndromic RP organoids showed compromised photoreceptor differentiation, whereas USH organoids showed a striking and unexpected cone phenotype. Furthermore, complementary clinical investigations identified macular atrophy in a high proportion of USH compared with RP individuals, further validating our observations that USH2A variants differentially affect cones. Overall, identification of distinct non-syndromic RP and USH phenotypes in multiple models provides valuable and robust readouts for testing the pathogenicity of USH2A variants as well as the efficacy of therapeutic approaches in complementary cell types.
Collapse
Affiliation(s)
- Carla Sanjurjo-Soriano
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| | - Carla Jimenez-Medina
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| | - Nejla Erkilic
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| | - Luisina Cappellino
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| | - Arnaud Lefevre
- National Reference Centre for Inherited Sensory Diseases, University of Montpellier, CHU, Montpellier, France
| | - Kerstin Nagel-Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, and Photoreceptor Cell Biology, Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, and Photoreceptor Cell Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Erwin Van Wijk
- Department of Otorhinolaryngology, Hearing, & Genes, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition, and Behavior, Nijmegen, the Netherlands
| | - Anne-Françoise Roux
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
- Molecular Genetics Laboratory, University of Montpellier, CHU, Montpellier, France
| | - Isabelle Meunier
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
- National Reference Centre for Inherited Sensory Diseases, University of Montpellier, CHU, Montpellier, France
| | - Vasiliki Kalatzis
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| |
Collapse
|
6
|
Iwagawa T, Masumoto H, Tabuchi H, Tani K, Conklin BR, Watanabe S. Evaluation of CRISPR/Cas9 exon-skipping vector for choroideremia using human induced pluripotent stem cell-derived RPE. J Gene Med 2023; 25:e3464. [PMID: 36413603 PMCID: PMC9898118 DOI: 10.1002/jgm.3464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 10/21/2022] [Accepted: 11/13/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Exon-skipping is a powerful genetic tool, especially when delivering genes using an AAV-mediated full-length gene supplementation strategy is difficult owing to large length of genes. Here, we used engineered human induced pluripotent stem cells and artificial intelligence to evaluate clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9-based exon-skipping vectors targeting genes of the retinal pigment epithelium (RPE). The model system was choroideremia; this is an X-linked inherited retinal disease caused by mutation of the CHM gene. METHODS We explored whether artificial intelligence detected differentiation of human OTX2, PAX6 and MITF (hOPM) cells, in which OTX2, PAX6 and MITF expression was induced by doxycycline treatment, into RPE. Plasmid encoding CHM exon-skipping modules targeting the splice donor sites of exons 6 were constructed. A clonal hOPM cell line with a frameshift mutation in exon 6 was generated and differentiated into RPE. CHM exon 6-skipping was induced, and the effects of skipping on phagocytic activity, cell death and prenylation of Rab small GTPase (RAB) were evaluated using flow cytometry, an in vitro prenylation assay and western blotting. RESULTS Artificial intelligence-based evaluation of RPE differentiation was successful. Retinal pigment epithelium cells with a frameshift mutation in exon 6 showed increased cell death, reduced phagocytic activity and increased cytosolic unprenylated RABs only when oxidative stress was in play. The latter two phenotypes were partially rescued by exon 6-skipping of CHM. CONCLUSIONS CHM exon 6-skipping contributed to RPE phagocytosis probably by increasing RAB38 prenylation under oxidative stress.
Collapse
Affiliation(s)
- Toshiro Iwagawa
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Department of Retinal Biology and Pathology, University of Tokyo Hospital, University of Tokyo, Tokyo, Japan
| | - Hiroki Masumoto
- Xeno Hoc, inc
- Department of Ophthalmology, Tsukazaki Hospital, Hyogo, Japan
| | - Hitoshi Tabuchi
- Department of Ophthalmology, Tsukazaki Hospital, Hyogo, Japan
- Department of Technology and Design Thinking for Medicine, Hiroshima University, Hiroshima, Japan
| | - Kenzaburo Tani
- Laboratory of ALA Advanced Medical Research, Institute for quantitative Biosciences, University of Tokyo, Tokyo, Japan
| | - Bruce R. Conklin
- Gladstone Institutes, San Francisco, CA, USA
- Departments of Medicine, Ophthalmology & Cellular and Molecular Pharmacology University of California, San Francisco, CA, United States, USA
| | - Sumiko Watanabe
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Department of Retinal Biology and Pathology, University of Tokyo Hospital, University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Long P, Shi Y, Sun F, Wei Y, Wu B, Li Q, Jie Q, Ma Y. Establishment of a non‐integrated induced pluripotent stem cell line derived from human chorionic villi cells. J Clin Lab Anal 2022; 36:e24464. [PMID: 35527669 PMCID: PMC9169189 DOI: 10.1002/jcla.24464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background Few studies have investigated the generation of induced pluripotent stem cells (iPSCs) derived from human primary chorionic villi (CV) cells. The present study aimed to explore an optimal electroporation (EP) condition for generating non‐integrated iPSCs from CV cells (CV‐iPSCs). Methods The EGFP plasmid was transfected into CV cells under different EP conditions to evaluate cell adherence and the rate of EGFP positive cells. Subsequently, CV cells were transfected with the pEP4‐E02S‐ET2K and pCEP4‐miR‐302–367 plasmids under optimal EP conditions. Finally, CV‐iPSC pluripotency, karyotype analysis, and differentiation ability were investigated. Results Following EP for 48 h under different conditions, different confluency, and transfection efficiency were observed in CV cells. Higher cell density was observed in CV cells exposed to 200 V for 100 s, while higher transfection efficiency was obtained in cells electroporated at a pulse of 300 V for 300 s. To generate typical primitive iPSCs, CV cells were transfected with pEP4‐E02S‐ET2K and pCEP4‐miR‐302–367 plasmids using EP and were then cultured in induction medium for 20 days under selected conditions. Subsequently, monoclonal iPSCs were isolated and were evaluated pluripotency with AP positive staining, the expression of OCT4, SOX2, and NANOG in vitro and the formation of three germ layer teratomas in vivo. Conclusion CV‐iPSCs were successfully established under the conditions of 100 μl shock cup and EP pulse of 200 V for 300 s for two times. This may provide a novel strategy for investigating the pathogenesis of several diseases and gene therapy.
Collapse
Affiliation(s)
- Ping Long
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
- Guizhou Qiannan People's Hospital Guizhou China
| | - Yuechuan Shi
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
- Hainan Medical University Haikou Hainan China
| | - Fei Sun
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
- Department of Obstetrics and Gynecology of Nanfang Hospital Southern Medical University Guangzhou China
| | - Yunjian Wei
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
| | - Bangyong Wu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
| | - Qi Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
| | - Qiuling Jie
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research The First Affiliated Hospital of Hainan Medical University Haikou Haikou Hainan China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education Hainan Medical University Haikou Hainan China
- Haikou Key Laboratory of Human Genetic Resources Preservation of First Affiliated Hospital Hainan Medical University Haikou Hainan China
- Hainan Medical University Haikou Hainan China
| |
Collapse
|
8
|
Sarkar H, Moosajee M. Choroideremia: molecular mechanisms and therapies. Trends Mol Med 2022; 28:378-387. [PMID: 35341685 DOI: 10.1016/j.molmed.2022.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 10/18/2022]
Abstract
Choroideremia (CHM) is a monogenic X-linked chorioretinal dystrophy affecting the photoreceptors, retinal pigment epithelium (RPE), and choroid; it is caused by mutations involving the CHM gene. CHM is characterized by night blindness in early childhood, progressing to peripheral visual field loss and eventually to complete blindness from middle age. CHM encodes the ubiquitously expressed Rab escort protein 1 (REP1), which is responsible for prenylation of Rab proteins and is essential for intracellular trafficking of vesicles. In this review we explore the role of REP1 in the retina and its newly discovered systemic manifestations, and discuss the therapeutic strategies for tackling this disease, including the outcomes from recent clinical trials.
Collapse
Affiliation(s)
- Hajrah Sarkar
- Development, Ageing, and Disease, University College London (UCL) Institute of Ophthalmology, London, EC1V 9EL, UK; Ocular Genomics and Therapeutics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Mariya Moosajee
- Development, Ageing, and Disease, University College London (UCL) Institute of Ophthalmology, London, EC1V 9EL, UK; Ocular Genomics and Therapeutics Laboratory, The Francis Crick Institute, London, NW1 1AT, UK; Department of Genetics, Moorfields Eye Hospital National Health Service (NHS) Foundation Trust, London, EC1V 2PD, UK; Department of Ophthalmology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, WC1N 3JH, UK.
| |
Collapse
|
9
|
Bucher K, Rodriguez-Bocanegra E, Fischer MD. Benefits and Shortcomings of Laboratory Model Systems in the Development of Genetic Therapies. Klin Monbl Augenheilkd 2022; 239:263-269. [PMID: 35316853 DOI: 10.1055/a-1757-9879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Gene therapeutic approaches promise treatment or even a cure of diseases that were previously untreatable. Retinal gene therapies tested in clinical trials comprise a wide range of different strategies, including gene supplementation therapies, in vivo gene editing, modulation of splicing mechanisms, or the suppression of gene expression. To guarantee efficient transfer of genetic material into the respective target cells while avoiding major adverse effects, the development of genetic therapies requires appropriate in vitro model systems that allow tests of efficacy and safety of the gene therapeutic approach. In this review, we introduce various in vitro models of different levels of complexity used in the development of genetic therapies and discuss their respective benefits and shortcomings using the example of adeno-associated virus-based retinal gene therapy.
Collapse
Affiliation(s)
- Kirsten Bucher
- University Eye Hospital, University Hospital Tübingen Clinic of Ophthalmology, Tubingen, Germany.,Institute for Ophthalmic Research, University Hospital Tübingen Clinic of Ophthalmology, Tubingen, Germany
| | | | - M Dominik Fischer
- University Eye Hospital, University Hospital Tübingen Clinic of Ophthalmology, Tubingen, Germany.,Institute for Ophthalmic Research, University Hospital Tübingen Clinic of Ophthalmology, Tubingen, Germany.,Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom of Great Britain and Northern Ireland.,Department of Clinical Neurosciences, University of Oxford Nuffield Laboratory of Ophthalmology, Oxford, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
10
|
Kalatzis V, Roux AF, Meunier I. Molecular Therapy for Choroideremia: Pre-clinical and Clinical Progress to Date. Mol Diagn Ther 2021; 25:661-675. [PMID: 34661884 DOI: 10.1007/s40291-021-00558-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2021] [Indexed: 12/01/2022]
Abstract
Choroideremia is an inherited retinal disease characterised by a degeneration of the light-sensing photoreceptors, supporting retinal pigment epithelium and underlying choroid. Patients present with the same symptoms as those with classic rod-cone dystrophy: (1) night blindness early in life; (2) progressive peripheral visual field loss, and (3) central vision decline with a slow progression to legal blindness. Choroideremia is monogenic and caused by mutations in CHM. Eight clinical trials (three phase 1/2, four phase 2, and one phase 3) have started (four of which are already finished) to evaluate the therapeutic efficacy of gene supplementation mediated by subretinal delivery of an adeno-associated virus serotype 2 (AAV2/2) vector expressing CHM. Furthermore, one phase 1 clinical trial has been initiated to evaluate the efficiency of a novel AAV variant to deliver CHM to the outer retina following intravitreal delivery. Lastly, a non-viral-mediated CHM replacement strategy is currently under development, which could lead to a future clinical trial. Here, we summarise the rationale behind these various studies, as well as any results published to date. The diversity of these trials currently places choroideremia at the forefront of the retinal gene therapy field. As a consequence, the trial outcomes, regardless of the results, have the potential to change the landscape of gene supplementation for inherited retinal diseases.
Collapse
Affiliation(s)
- Vasiliki Kalatzis
- Institute for Neurosciences of Montpellier, Univ Montpellier, Inserm U1298, Hôpital St Eloi, 80 Avenue Augustin Fliche, 34091, Montpellier, France.
| | - Anne-Françoise Roux
- Institute for Neurosciences of Montpellier, Univ Montpellier, Inserm U1298, Hôpital St Eloi, 80 Avenue Augustin Fliche, 34091, Montpellier, France.,Molecular Genetics Laboratory, Univ Montpellier, CHU Montpellier, Montpellier, France
| | - Isabelle Meunier
- Institute for Neurosciences of Montpellier, Univ Montpellier, Inserm U1298, Hôpital St Eloi, 80 Avenue Augustin Fliche, 34091, Montpellier, France.,National Reference Centre for Inherited Sensory Diseases, University of Montpellier, CHU Montpellier, Montpellier, France
| |
Collapse
|
11
|
Huang Z, Zhang D, Chen SC, Jennings L, Carvalho LS, Fletcher S, Chen FK, McLenachan S. Gene replacement therapy restores RCBTB1 expression and cilium length in patient-derived retinal pigment epithelium. J Cell Mol Med 2021; 25:10020-10027. [PMID: 34617687 PMCID: PMC8572767 DOI: 10.1111/jcmm.16911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
Biallelic mutations in the RCBTB1 gene cause retinal dystrophy. Here, we characterized the effects of RCBTB1 gene deficiency in retinal pigment epithelial (RPE) cells derived from a patient with RCBTB1‐associated retinopathy and restored RCBTB1 expression in these cells using adeno‐associated viral (AAV) vectors. Induced pluripotent stem cells derived from a patient with compound heterozygous RCBTB1 mutations (c.170delG and c.707delA) and healthy control subjects were differentiated into RPE cells. RPE cells were treated with AAV vectors carrying a RCBTB1 transgene. Patient‐derived RPE cells showed reduced expression of RCBTB1. Expression of NFE2L2 showed a non‐significant reduction in patient RPE cells compared with controls, while expression of its target genes (RXRA, IDH1 and SLC25A25) was significantly reduced. Trans‐epithelial electrical resistance, surface microvillus densities and primary cilium lengths were reduced in patient‐derived RPE cells, compared with controls. Treatment of patient RPE with AAV vectors significantly increased RCBTB1, NFE2L2 and RXRA expression and cilium lengths. Our study provides the first report examining the phenotype of RPE cells derived from a patient with RCBTB1‐associated retinopathy. Furthermore, treatment of patient‐derived RPE with AAV‐RCBTB1 vectors corrected deficits in gene expression and RPE ultrastructure, supporting the use of gene replacement therapy for treating this inherited retinal disease.
Collapse
Affiliation(s)
- Zhiqin Huang
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, WA, Australia.,Lions Eye Institute, Nedlands, WA, Australia
| | - Dan Zhang
- Lions Eye Institute, Nedlands, WA, Australia
| | | | | | - Livia S Carvalho
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, WA, Australia.,Lions Eye Institute, Nedlands, WA, Australia
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Fred K Chen
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, WA, Australia.,Lions Eye Institute, Nedlands, WA, Australia.,Department of Ophthalmology, Royal Perth Hospital, Perth, WA, Australia.,Department of Ophthalmology, Perth Children's Hospital, Nedlands, WA, Australia
| | - Samuel McLenachan
- Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, WA, Australia.,Lions Eye Institute, Nedlands, WA, Australia
| |
Collapse
|
12
|
Coco-Martin RM, Pastor-Idoate S, Pastor JC. Cell Replacement Therapy for Retinal and Optic Nerve Diseases: Cell Sources, Clinical Trials and Challenges. Pharmaceutics 2021; 13:pharmaceutics13060865. [PMID: 34208272 PMCID: PMC8230855 DOI: 10.3390/pharmaceutics13060865] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/15/2022] Open
Abstract
The aim of this review was to provide an update on the potential of cell therapies to restore or replace damaged and/or lost cells in retinal degenerative and optic nerve diseases, describing the available cell sources and the challenges involved in such treatments when these techniques are applied in real clinical practice. Sources include human fetal retinal stem cells, allogenic cadaveric human cells, adult hippocampal neural stem cells, human CNS stem cells, ciliary pigmented epithelial cells, limbal stem cells, retinal progenitor cells (RPCs), human pluripotent stem cells (PSCs) (including both human embryonic stem cells (ESCs) and human induced pluripotent stem cells (iPSCs)) and mesenchymal stem cells (MSCs). Of these, RPCs, PSCs and MSCs have already entered early-stage clinical trials since they can all differentiate into RPE, photoreceptors or ganglion cells, and have demonstrated safety, while showing some indicators of efficacy. Stem/progenitor cell therapies for retinal diseases still have some drawbacks, such as the inhibition of proliferation and/or differentiation in vitro (with the exception of RPE) and the limited long-term survival and functioning of grafts in vivo. Some other issues remain to be solved concerning the clinical translation of cell-based therapy, including (1) the ability to enrich for specific retinal subtypes; (2) cell survival; (3) cell delivery, which may need to incorporate a scaffold to induce correct cell polarization, which increases the size of the retinotomy in surgery and, therefore, the chance of severe complications; (4) the need to induce a localized retinal detachment to perform the subretinal placement of the transplanted cell; (5) the evaluation of the risk of tumor formation caused by the undifferentiated stem cells and prolific progenitor cells. Despite these challenges, stem/progenitor cells represent the most promising strategy for retinal and optic nerve disease treatment in the near future, and therapeutics assisted by gene techniques, neuroprotective compounds and artificial devices can be applied to fulfil clinical needs.
Collapse
Affiliation(s)
- Rosa M. Coco-Martin
- Instituto de Oftalmobiologia Aplicada (IOBA), Medical School, Universidad de Valladolid, 47011 Valladolid, Spain; (S.P.-I.); (J.C.P.)
- National Institute of Health Carlos III (ISCIII), (RETICS) Cooperative Health Network for Research in Ophthalmology (Oftared), 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-983423559
| | - Salvador Pastor-Idoate
- Instituto de Oftalmobiologia Aplicada (IOBA), Medical School, Universidad de Valladolid, 47011 Valladolid, Spain; (S.P.-I.); (J.C.P.)
- National Institute of Health Carlos III (ISCIII), (RETICS) Cooperative Health Network for Research in Ophthalmology (Oftared), 28040 Madrid, Spain
- Department of Ophthalmology, Hospital Clinico Universitario of Valladolid, 47003 Valladolid, Spain
| | - Jose Carlos Pastor
- Instituto de Oftalmobiologia Aplicada (IOBA), Medical School, Universidad de Valladolid, 47011 Valladolid, Spain; (S.P.-I.); (J.C.P.)
- National Institute of Health Carlos III (ISCIII), (RETICS) Cooperative Health Network for Research in Ophthalmology (Oftared), 28040 Madrid, Spain
- Department of Ophthalmology, Hospital Clinico Universitario of Valladolid, 47003 Valladolid, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Fundacion del Instituto de Estudios de Ciencias de la Salud de Castilla y León (ICSCYL), 42002 Soria, Spain
| |
Collapse
|
13
|
Garita-Hernandez M, Chaffiol A, Guibbal L, Routet F, Khabou H, Riancho L, Toualbi L, Picaud S, Sahel JA, Goureau O, Duebel J, Dalkara D. Control of Microbial Opsin Expression in Stem Cell Derived Cones for Improved Outcomes in Cell Therapy. Front Cell Neurosci 2021; 15:648210. [PMID: 33815066 PMCID: PMC8012682 DOI: 10.3389/fncel.2021.648210] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Human-induced pluripotent stem cell (hiPSC) derived organoids have become increasingly used systems allowing 3D-modeling of human organ development, and disease. They are also a reliable source of cells for transplantation in cell therapy and an excellent model to validate gene therapies. To make full use of these systems, a toolkit of genetic modification techniques is necessary to control their activity in line with the downstream application. We have previously described adeno-associated viruse (AAV) vectors for efficient targeting of cells within human retinal organoids. Here, we describe biological restriction and enhanced gene expression in cone cells of such organoids thanks to the use of a 1.7-kb L-opsin promoter. We illustrate the usefulness of implementing such a promoter to enhance the expression of the red-shifted opsin Jaws in fusion with a fluorescent reporter gene, enabling cell sorting to enrich the desired cell population. Increased Jaws expression after transplantation improved light responses promising better therapeutic outcomes in a cell therapy setting. Our results point to the importance of promoter activity in restricting, improving, and controlling the kinetics of transgene expression during the maturation of hiPSC retinal derivatives. Differentiation requires mechanisms to initiate specific transcriptional changes and to reinforce those changes when mature cell states are reached. By employing a cell-type-specific promoter we put transgene expression under the new transcriptional program of mature cells.
Collapse
Affiliation(s)
| | | | - Laure Guibbal
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Fiona Routet
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Hanen Khabou
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Luisa Riancho
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Lyes Toualbi
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Serge Picaud
- Institut de la Vision, Sorbonne Université, Paris, France
| | - José-Alain Sahel
- Institut de la Vision, Sorbonne Université, Paris, France
- CHNO des Quinze−Vingts, DHU Sight Restore, Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Jens Duebel
- Institut de la Vision, Sorbonne Université, Paris, France
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany
| | - Deniz Dalkara
- Institut de la Vision, Sorbonne Université, Paris, France
| |
Collapse
|
14
|
Mamaeva D, Jazouli Z, DiFrancesco ML, Erkilic N, Dubois G, Hilaire C, Meunier I, Boukhaddaoui H, Kalatzis V. Novel roles for voltage-gated T-type Ca 2+ and ClC-2 channels in phagocytosis and angiogenic factor balance identified in human iPSC-derived RPE. FASEB J 2021; 35:e21406. [PMID: 33724552 DOI: 10.1096/fj.202002754r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 01/26/2023]
Abstract
Human-induced pluripotent stem cell (hiPSC)-derived retinal pigment epithelium (RPE) is a powerful tool for pathophysiological studies and preclinical therapeutic screening, as well as a source for clinical cell transplantation. Thus, it must be validated for maturity and functionality to ensure correct data readouts and clinical safety. Previous studies have validated hiPSC-derived RPE as morphologically characteristic of the tissue in the human eye. However, information concerning the expression and functionality of ion channels is still limited. We screened hiPSC-derived RPE for the polarized expression of a panel of L-type (CaV 1.1, CaV 1.3) and T-type (CaV 3.1, CaV 3.3) Ca2+ channels, K+ channels (Maxi-K, Kir4.1, Kir7.1), and the Cl- channel ClC-2 known to be expressed in native RPE. We also tested the roles of these channels in key RPE functions using specific inhibitors. In addition to confirming the native expression profiles and function of certain channels, such as L-type Ca2+ channels, we show for the first time that T-type Ca2+ channels play a role in both phagocytosis and vascular endothelial growth factor (VEGF) secretion. Moreover, we demonstrate that Maxi-K and Kir7.1 channels are involved in the polarized secretion of VEGF and pigment epithelium-derived factor (PEDF). Furthermore, we show a novel localization for ClC-2 channel on the apical side of hiPSC-derived RPE, with an overexpression at the level of fluid-filled domes, and demonstrate that it plays an important role in phagocytosis, as well as VEGF and PEDF secretion. Taken together, hiPSC-derived RPE is a powerful model for advancing fundamental knowledge of RPE functions.
Collapse
Affiliation(s)
- Daria Mamaeva
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Zhour Jazouli
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Mattia L DiFrancesco
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Nejla Erkilic
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France.,National Reference Centre for Inherited Sensory Diseases, Montpellier University, CHU, Montpellier, France
| | - Gregor Dubois
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Cecile Hilaire
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Isabelle Meunier
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France.,National Reference Centre for Inherited Sensory Diseases, Montpellier University, CHU, Montpellier, France
| | - Hassan Boukhaddaoui
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| | - Vasiliki Kalatzis
- Institute for Neurosciences of Montpellier, Inserm, Montpellier University, Montpellier, France
| |
Collapse
|
15
|
Zeitz C, Nassisi M, Laurent-Coriat C, Andrieu C, Boyard F, Condroyer C, Démontant V, Antonio A, Lancelot ME, Frederiksen H, Kloeckener-Gruissem B, El-Shamieh S, Zanlonghi X, Meunier I, Roux AF, Mohand-Saïd S, Sahel JA, Audo I. CHM mutation spectrum and disease: An update at the time of human therapeutic trials. Hum Mutat 2021; 42:323-341. [PMID: 33538369 DOI: 10.1002/humu.24174] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/21/2020] [Accepted: 01/20/2021] [Indexed: 12/15/2022]
Abstract
Choroideremia is an X-linked inherited retinal disorder (IRD) characterized by the degeneration of retinal pigment epithelium, photoreceptors, choriocapillaris and choroid affecting males with variable phenotypes in female carriers. Unlike other IRD, characterized by a large clinical and genetic heterogeneity, choroideremia shows a specific phenotype with causative mutations in only one gene, CHM. Ongoing gene replacement trials raise further interests in this disorder. We describe here the clinical and genetic data from a French cohort of 45 families, 25 of which carry novel variants, in the context of 822 previously reported choroideremia families. Most of the variants represent loss-of-function mutations with eleven families having large (i.e. ≥6 kb) genomic deletions, 18 small insertions, deletions or insertion deletions, six showing nonsense variants, eight splice site variants and two missense variants likely to affect splicing. Similarly, 822 previously published families carry mostly loss-of-function variants. Recurrent variants are observed worldwide, some of which linked to a common ancestor, others arisen independently in specific CHM regions prone to mutations. Since all exons of CHM may harbor variants, Sanger sequencing combined with quantitative polymerase chain reaction or multiplex ligation-dependent probe amplification experiments are efficient to achieve the molecular diagnosis in patients with typical choroideremia features.
Collapse
Affiliation(s)
- Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Marco Nassisi
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Camille Andrieu
- CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France
| | - Fiona Boyard
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Vanessa Démontant
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Aline Antonio
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Helen Frederiksen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Barbara Kloeckener-Gruissem
- Institute of Medical Molecular Genetics, University of Zurich, Schlieren, Switzerland.,Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Said El-Shamieh
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| | - Xavier Zanlonghi
- Clinique Pluridisciplinaire Jules Verne, Institut Ophtalmologique de l'Ouest, Nantes, France
| | - Isabelle Meunier
- National Reference Centre for Inherited Sensory Diseases, University of Montpellier, Montpellier University Hospital, Montpellier, France.,Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| | - Anne-Françoise Roux
- Laboratoire de Génétique Moléculaire, CHU de Montpellier, Université de Montpellier, Montpellier, France
| | - Saddek Mohand-Saïd
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France.,Fondation Ophtalmologique Adolphe de Rothschild, Paris, France.,Académie des Sciences-Institut de France, Paris, France.,Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France.,Department of Genetics, UCL-Institute of Ophthalmology, London, UK
| |
Collapse
|
16
|
Buck TM, Wijnholds J. Recombinant Adeno-Associated Viral Vectors (rAAV)-Vector Elements in Ocular Gene Therapy Clinical Trials and Transgene Expression and Bioactivity Assays. Int J Mol Sci 2020; 21:E4197. [PMID: 32545533 PMCID: PMC7352801 DOI: 10.3390/ijms21124197] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Inherited retinal dystrophies and optic neuropathies cause chronic disabling loss of visual function. The development of recombinant adeno-associated viral vectors (rAAV) gene therapies in all disease fields have been promising, but the translation to the clinic has been slow. The safety and efficacy profiles of rAAV are linked to the dose of applied vectors. DNA changes in the rAAV gene cassette affect potency, the expression pattern (cell-specificity), and the production yield. Here, we present a library of rAAV vectors and elements that provide a workflow to design novel vectors. We first performed a meta-analysis on recombinant rAAV elements in clinical trials (2007-2020) for ocular gene therapies. We analyzed 33 unique rAAV gene cassettes used in 57 ocular clinical trials. The rAAV gene therapy vectors used six unique capsid variants, 16 different promoters, and six unique polyadenylation sequences. Further, we compiled a list of promoters, enhancers, and other sequences used in current rAAV gene cassettes in preclinical studies. Then, we give an update on pro-viral plasmid backbones used to produce the gene therapy vectors, inverted terminal repeats, production yield, and rAAV safety considerations. Finally, we assess rAAV transgene and bioactivity assays applied to cells or organoids in vitro, explants ex vivo, and clinical studies.
Collapse
Affiliation(s)
- Thilo M. Buck
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
- Netherlands Institute of Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
17
|
Udry F, Decembrini S, Gamm DM, Déglon N, Kostic C, Arsenijevic Y. Lentiviral mediated RPE65 gene transfer in healthy hiPSCs-derived retinal pigment epithelial cells markedly increased RPE65 mRNA, but modestly protein level. Sci Rep 2020; 10:8890. [PMID: 32483256 PMCID: PMC7264209 DOI: 10.1038/s41598-020-65657-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 05/08/2020] [Indexed: 12/15/2022] Open
Abstract
The retinal pigment epithelium (RPE) is a monolayer of cobblestone-like epithelial cells that accomplishes critical functions for the retina. Several protocols have been published to differentiate pluripotent stem cells into RPE cells suitable for disease modelling and therapy development. In our study, the RPE identity of human induced pluripotent stem cell (hiPSC)-derived RPE (iRPE) was extensively characterized, and then used to test a lentiviral-mediated RPE65 gene augmentation therapy. A dose study of the lentiviral vector revealed a dose-dependent effect of the vector on RPE65 mRNA levels. A marked increase of the RPE65 mRNA was also observed in the iRPE (100-fold) as well as in an experimental set with RPE derived from another hiPSC source and from foetal human RPE. Although iRPE displayed features close to bona fide RPE, no or a modest increase of the RPE65 protein level was observed depending on the protein detection method. Similar results were observed with the two other cell lines. The mechanism of RPE65 protein regulation remains to be elucidated, but the current work suggests that high vector expression will not produce an excess of the normal RPE65 protein level.
Collapse
Affiliation(s)
- Florian Udry
- Department of ophthalmology, Unit of Retinal Degeneration and Regeneration, University of Lausanne, Hôpital ophtalmique Jules-Gonin, 1004, Lausanne, Switzerland
| | - Sarah Decembrini
- Department of ophthalmology, Unit of Retinal Degeneration and Regeneration, University of Lausanne, Hôpital ophtalmique Jules-Gonin, 1004, Lausanne, Switzerland
- Department of Biomedicine, University Hospital Basel & University Basel, Hebelstr. 20, 4031, Basel, Switzerland
| | - David M Gamm
- McPherson Eye Research Institute, Waisman Center and Department of Ophthalmology and Visual Sciences, and University of Wisconsin-Madison, Madison, USA
| | - Nicole Déglon
- Neuroscience Research Center, Laboratory of Neurotherapies and Neuromodulation, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Corinne Kostic
- Department of ophthalmology, Unit of Retinal Degeneration and Regeneration, University of Lausanne, Hôpital ophtalmique Jules-Gonin, 1004, Lausanne, Switzerland
| | - Yvan Arsenijevic
- Department of ophthalmology, Unit of Retinal Degeneration and Regeneration, University of Lausanne, Hôpital ophtalmique Jules-Gonin, 1004, Lausanne, Switzerland.
| |
Collapse
|
18
|
Study of Usutu virus neuropathogenicity in mice and human cellular models. PLoS Negl Trop Dis 2020; 14:e0008223. [PMID: 32324736 PMCID: PMC7179837 DOI: 10.1371/journal.pntd.0008223] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/16/2020] [Indexed: 01/20/2023] Open
Abstract
Usutu virus (USUV), an African mosquito-borne flavivirus closely related to West Nile virus, was first isolated in South Africa in 1959. USUV emerged in Europe two decades ago, causing notably massive mortality in Eurasian blackbirds. USUV is attracting increasing attention due to its potential for emergence and its rapid spread in Europe in recent years. Although mainly asymptomatic or responsible for mild clinical signs, USUV was recently described as being associated with neurological disorders in humans such as encephalitis and meningoencephalitis, highlighting the potential health threat posed by the virus. Despite this, USUV pathogenesis remains largely unexplored. The aim of this study was to evaluate USUV neuropathogenicity using in vivo and in vitro approaches. Our results indicate that USUV efficiently replicates in the murine central nervous system. Replication in the spinal cord and brain is associated with recruitment of inflammatory cells and the release of inflammatory molecules as well as induction of antiviral-responses without major modulation of blood-brain barrier integrity. Endothelial cells integrity is also maintained in a human model of the blood-brain barrier despite USUV replication and release of pro-inflammatory cytokines. Furthermore, USUV-inoculated mice developed major ocular defects associated with inflammation. Moreover, USUV efficiently replicates in human retinal pigment epithelium. Our results will help to better characterize the physiopathology related to USUV infection in order to anticipate the potential threat of USUV emergence. Number of emerging arboviruses involved in human infections has increased considerably in the past years. Among them, Usutu virus (USUV) is an African mosquito-borne virus first isolated in South Africa that recently emerged. USUV infection in humans is considered to be most often asymptomatic or to cause mild clinical signs. Nonetheless, increased cases of neurological complications such as encephalitis or meningoencephalitis have been reported in Europe but the mechanisms behind this neuropathogenesis remain largely unclear. In this study we showed that USUV can infect efficiently several organs and cells of the central nervous system associated with a drastic inflammation and various deleterious effects. Our results contribute to the characterization of the neurotropism related to USUV infection.
Collapse
|
19
|
Erkilic N, Gatinois V, Torriano S, Bouret P, Sanjurjo-Soriano C, Luca VD, Damodar K, Cereso N, Puechberty J, Sanchez-Alcudia R, Hamel CP, Ayuso C, Meunier I, Pellestor F, Kalatzis V. A Novel Chromosomal Translocation Identified due to Complex Genetic Instability in iPSC Generated for Choroideremia. Cells 2019; 8:cells8091068. [PMID: 31514470 PMCID: PMC6770680 DOI: 10.3390/cells8091068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 08/28/2019] [Accepted: 09/07/2019] [Indexed: 12/19/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) have revolutionized the study of human diseases as they can renew indefinitely, undergo multi-lineage differentiation, and generate disease-specific models. However, the difficulty of working with iPSCs is that they are prone to genetic instability. Furthermore, genetically unstable iPSCs are often discarded, as they can have unforeseen consequences on pathophysiological or therapeutic read-outs. We generated iPSCs from two brothers of a previously unstudied family affected with the inherited retinal dystrophy choroideremia. We detected complex rearrangements involving chromosomes 12, 20 and/or 5 in the generated iPSCs. Suspecting an underlying chromosomal aberration, we performed karyotype analysis of the original fibroblasts, and of blood cells from additional family members. We identified a novel chromosomal translocation t(12;20)(q24.3;q11.2) segregating in this family. We determined that the translocation was balanced and did not impact subsequent retinal differentiation. We show for the first time that an undetected genetic instability in somatic cells can breed further instability upon reprogramming. Therefore, the detection of chromosomal aberrations in iPSCs should not be disregarded, as they may reveal rearrangements segregating in families. Furthermore, as such rearrangements are often associated with reproductive failure or birth defects, this in turn has important consequences for genetic counseling of family members.
Collapse
Affiliation(s)
- Nejla Erkilic
- Inserm U1051, Institute for Neurosciences of Montpellier, 34091 Montpellier CEDEX 5, France
- University of Montpellier, 34090 Montpellier, France
| | - Vincent Gatinois
- Chromosomal Genetics Unit, Chromostem Platform, CHU, Montpellier, France
| | - Simona Torriano
- Inserm U1051, Institute for Neurosciences of Montpellier, 34091 Montpellier CEDEX 5, France
- University of Montpellier, 34090 Montpellier, France
| | - Pauline Bouret
- Chromosomal Genetics Unit, Chromostem Platform, CHU, Montpellier, France
| | - Carla Sanjurjo-Soriano
- Inserm U1051, Institute for Neurosciences of Montpellier, 34091 Montpellier CEDEX 5, France
- University of Montpellier, 34090 Montpellier, France
| | - Valerie De Luca
- Inserm U1051, Institute for Neurosciences of Montpellier, 34091 Montpellier CEDEX 5, France
- University of Montpellier, 34090 Montpellier, France
| | - Krishna Damodar
- Inserm U1051, Institute for Neurosciences of Montpellier, 34091 Montpellier CEDEX 5, France
- University of Montpellier, 34090 Montpellier, France
| | - Nicolas Cereso
- Inserm U1051, Institute for Neurosciences of Montpellier, 34091 Montpellier CEDEX 5, France
- University of Montpellier, 34090 Montpellier, France
| | - Jacques Puechberty
- Service of Clinical Genetics, Department of Medical Genetics, Rare Diseases and Personalized Medicine, CHU, Montpellier, France
| | - Rocio Sanchez-Alcudia
- Department of Genetics, Institute for Sanitary Investigation, Foundation Jimenez Diaz, 28040 Madrid, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 28029 Madrid, Spain
| | - Christian P Hamel
- Inserm U1051, Institute for Neurosciences of Montpellier, 34091 Montpellier CEDEX 5, France
- University of Montpellier, 34090 Montpellier, France
- National Reference Centre for Inherited Sensory Diseases, CHU, 34295 Montpellier, France
| | - Carmen Ayuso
- Department of Genetics, Institute for Sanitary Investigation, Foundation Jimenez Diaz, 28040 Madrid, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), 28029 Madrid, Spain
| | - Isabelle Meunier
- Inserm U1051, Institute for Neurosciences of Montpellier, 34091 Montpellier CEDEX 5, France
- University of Montpellier, 34090 Montpellier, France
- National Reference Centre for Inherited Sensory Diseases, CHU, 34295 Montpellier, France
| | - Franck Pellestor
- Chromosomal Genetics Unit, Chromostem Platform, CHU, Montpellier, France
| | - Vasiliki Kalatzis
- Inserm U1051, Institute for Neurosciences of Montpellier, 34091 Montpellier CEDEX 5, France.
- University of Montpellier, 34090 Montpellier, France.
| |
Collapse
|
20
|
Mishra B, Wilson DR, Sripathi SR, Suprenant MP, Rui Y, Wahlin KJ, Berlinicke CA, Green JJ, Zack DJ. A combinatorial library of biodegradable polyesters enables non-viral gene delivery to post-mitotic human stem cell-derived polarized RPE monolayers. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019; 6:273-285. [PMID: 33732871 PMCID: PMC7962803 DOI: 10.1007/s40883-019-00118-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 04/16/2019] [Accepted: 07/01/2019] [Indexed: 12/28/2022]
Abstract
Safe and effective delivery of DNA to post-mitotic cells, especially highly differentiated cells, remains a challenge despite significant progress in the development of gene delivery tools. Biodegradable polymeric nanoparticles (NPs) offer an array of advantages for gene delivery over viral vectors due to improved safety, carrying capacity, ease of manufacture, and cell-type specificity. Here we demonstrate the use of a high-throughput screening (HTS) platform to synthesize and screen a library of 148 biodegradable polymeric nanoparticles, successfully identifying structures that enable efficient transfection of human pluripotent stem cell differentiated human retinal pigment epithelial (RPE) cells with minimal toxicity. These NPs can deliver plasmid DNA (pDNA) to RPE monolayers more efficiently than leading commercially available transfection reagents. Novel synthetic polymers are described that enable high efficacy non-viral gene delivery to hard-to-transfect polarized human RPE monolayers, enabling gene loss- and gain-of-function studies of cell signaling, developmental, and disease-related pathways. One new synthetic polymer in particular, 3,3'-iminobis(N,N-dimethylpropylamine)-end terminated poly(1,5-pentanediol diacrylate-co-3 amino-1-propanol) (5-3-J12), was found to form self-assembled nanoparticles when mixed with plasmid DNA that transfect a majority of these human post-mitotic cells with minimal cytotoxicity. The platform described here can be utilized as an enabling technology for gene transfer to human primary and stem cell-derived cells, which are often fragile and resistant to conventional gene transfer approaches.
Collapse
Affiliation(s)
- Bibhudatta Mishra
- Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - David R. Wilson
- Biomedical Engineering, Johns Hopkins University, Baltimore, 21231, United States
- Translational Tissue Engineering Center, Johns Hopkins of Medicine, Baltimore, MD 21231, United States
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21231, United States
| | - Srinivas R. Sripathi
- Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Mark P. Suprenant
- Biomedical Engineering, Johns Hopkins University, Baltimore, 21231, United States
- Translational Tissue Engineering Center, Johns Hopkins of Medicine, Baltimore, MD 21231, United States
| | - Yuan Rui
- Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
- Biomedical Engineering, Johns Hopkins University, Baltimore, 21231, United States
- Translational Tissue Engineering Center, Johns Hopkins of Medicine, Baltimore, MD 21231, United States
| | - Karl J. Wahlin
- Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Cynthia A. Berlinicke
- Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Jordan J. Green
- Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
- Biomedical Engineering, Johns Hopkins University, Baltimore, 21231, United States
- Translational Tissue Engineering Center, Johns Hopkins of Medicine, Baltimore, MD 21231, United States
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21231, United States
- Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
- Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21231, United States
- Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Donald J. Zack
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21231, United States
- Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
- Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
- Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
- Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| |
Collapse
|
21
|
Comparative AAV-eGFP Transgene Expression Using Vector Serotypes 1-9, 7m8, and 8b in Human Pluripotent Stem Cells, RPEs, and Human and Rat Cortical Neurons. Stem Cells Int 2019; 2019:7281912. [PMID: 30800164 PMCID: PMC6360060 DOI: 10.1155/2019/7281912] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 10/30/2018] [Accepted: 11/16/2018] [Indexed: 01/03/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV), produced from a nonpathogenic parvovirus, has become an increasing popular vector for gene therapy applications in human clinical trials. However, transduction and transgene expression of rAAVs can differ across in vitro and ex vivo cellular transduction strategies. This study compared 11 rAAV serotypes, carrying one reporter transgene cassette containing a cytomegalovirus immediate-early enhancer (eCMV) and chicken beta actin (CBA) promoter driving the expression of an enhanced green-fluorescent protein (eGFP) gene, which was transduced into four different cell types: human iPSC, iPSC-derived RPE, iPSC-derived cortical, and dissociated embryonic day 18 rat cortical neurons. Each cell type was exposed to three multiplicity of infections (MOI: 1E4, 1E5, and 1E6 vg/cell). After 24, 48, 72, and 96 h posttransduction, GFP-expressing cells were examined and compared across dosage, time, and cell type. Retinal pigmented epithelium showed highest AAV-eGFP expression and iPSC cortical the lowest. At an MOI of 1E6 vg/cell, all serotypes show measurable levels of AAV-eGFP expression; moreover, AAV7m8 and AAV6 perform best across MOI and cell type. We conclude that serotype tropism is not only capsid dependent but also cell type plays a significant role in transgene expression dynamics.
Collapse
|
22
|
Utility of Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelium for an In Vitro Model of Proliferative Vitreoretinopathy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1186:33-53. [PMID: 31654385 DOI: 10.1007/978-3-030-28471-8_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The advent of stem cell technology, including the technology to induce pluripotency in somatic cells, and direct differentiation of stem cells into specific somatic cell types, has created an exciting new field of scientific research. Much of the work with pluripotent stem (PS) cells has been focused on the exploration and exploitation of their potential as cells/tissue replacement therapies for personalized medicine. However, PS and stem cell-derived somatic cells are also proving to be valuable tools to study disease pathology and tissue-specific responses to injury. High-throughput drug screening assays using tissue-specific injury models have the potential to identify specific and effective treatments that will promote wound healing. Retinal pigment epithelium (RPE) derived from induced pluripotent stem cells (iPS-RPE) are well characterized cells that exhibit the phenotype and functions of in vivo RPE. In addition to their role as a source of cells to replace damaged or diseased RPE, iPS-RPE provide a robust platform for in vitro drug screening to identify novel therapeutics to promote healing and repair of ocular tissues after injury. Proliferative vitreoretinopathy (PVR) is an abnormal wound healing process that occurs after retinal tears or detachments. In this chapter, the role of iPS-RPE in the development of an in vitro model of PVR is described. Comprehensive analyses of the iPS-RPE response to injury suggests that these cells provide a physiologically relevant tool to investigate the cellular mechanisms of the three phases of PVR pathology: migration, proliferation, and contraction. This in vitro model will provide valuable information regarding cellular wound healing responses specific to RPE and enable the identification of effective therapeutics.
Collapse
|
23
|
Cehajic Kapetanovic J, Patrício MI, MacLaren RE. Progress in the development of novel therapies for choroideremia. EXPERT REVIEW OF OPHTHALMOLOGY 2019; 14:277-285. [PMID: 32002021 PMCID: PMC6992425 DOI: 10.1080/17469899.2019.1699406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
INTRODUCTION There are no currently approved treatments for choroideremia, an X-linked progressive inherited retinal degeneration that leads to blindness by middle age. Several treatment options are being explored, but with major advances in adeno-associated vector (AAV) gene replacement therapy that has reached phase III clinical trials. AREAS COVERED In this review we discuss new insights into the clinical phenotyping and genetic testing of choroideremia patients, that aid disease characterisation, progression and patient inclusion into clinical trials. Recent advances in in-vitro studies have resulted in the development of functional assays that can be used to confirm the diagnosis in challenging cases and to quantify vector potency for use in clinical trials. We review the progress in current gene therapy trials and some considerations towards gene therapy approval for the treatment of choroideremia. Lastly, we discuss developments in alternative therapies including optogenetics. EXPERT COMMENTARY AAV gene replacement therapy is the most promising treatment strategy for choroideremia, that has developed exponentially over the last few years with a phase III clinical trial now underway. Optogenetics is a promising alternative strategy that might be applicable in late stages of degeneration.
Collapse
Affiliation(s)
- Jasmina Cehajic Kapetanovic
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Maria I Patrício
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK
- Nuffield Laboratory of Ophthalmology, Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
24
|
Simonin Y, Erkilic N, Damodar K, Clé M, Desmetz C, Bolloré K, Taleb M, Torriano S, Barthelemy J, Dubois G, Lajoix AD, Foulongne V, Tuaillon E, Van de Perre P, Kalatzis V, Salinas S. Zika virus induces strong inflammatory responses and impairs homeostasis and function of the human retinal pigment epithelium. EBioMedicine 2019; 39:315-331. [PMID: 30579862 PMCID: PMC6354710 DOI: 10.1016/j.ebiom.2018.12.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/19/2018] [Accepted: 12/06/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Zika virus (ZIKV) has recently re-emerged as a pathogenic agent with epidemic capacities as was well illustrated in South America. Because of the extent of this health crisis, a number of more serious symptoms have become associated with ZIKV infection than what was initially described. In particular, neuronal and ocular disorders have been characterized, both in infants and in adults. Notably, the macula and the retina can be strongly affected by ZIKV, possibly by a direct effect of the virus. This is supported by the detection of replicative and infectious virus in lachrimal fluid in human patients and mouse models. METHODS Here, we used an innovative, state-of-the-art iPSC-derived human retinal pigment epithelium (RPE) model to study ZIKV retinal impairment. FINDINGS We showed that the human RPE is highly susceptible to ZIKV infection and that a ZIKV African strain was more virulent and led to a more potent epithelium disruption and stronger anti-viral response than an Asian strain, suggesting lineage differences. Moreover, ZIKV infection led to impaired membrane dynamics involved in endocytosis, organelle biogenesis and potentially secretion, key mechanisms of RPE homeostasis and function. INTERPRETATION Taken together, our results suggest that ZIKV has a highly efficient ocular tropism, which creates a strong inflammatory environment that could have acute or chronic adverse effects. FUND: This work was funded by Retina France, REACTing and La Région Languedoc-Roussillon.
Collapse
Affiliation(s)
- Yannick Simonin
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, University of Montpellier, Montpellier, France
| | - Nejla Erkilic
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
| | - Krishna Damodar
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
| | - Marion Clé
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, University of Montpellier, Montpellier, France
| | - Caroline Desmetz
- BioCommunication en CardioMétabolique, University of Montpellier, Montpellier, France
| | - Karine Bolloré
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, University of Montpellier, Montpellier, France
| | - Mehdi Taleb
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, University of Montpellier, Montpellier, France
| | - Simona Torriano
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
| | - Jonathan Barthelemy
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, University of Montpellier, Montpellier, France
| | - Grégor Dubois
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
| | - Anne Dominique Lajoix
- BioCommunication en CardioMétabolique, University of Montpellier, Montpellier, France
| | - Vincent Foulongne
- Pathogenesis and Control of Chronic Infections. INSERM, University of Montpellier, Etablissement Français du Sang, CHU Montpellier, Montpellier, France
| | - Edouard Tuaillon
- Pathogenesis and Control of Chronic Infections. INSERM, University of Montpellier, Etablissement Français du Sang, CHU Montpellier, Montpellier, France
| | - Philippe Van de Perre
- Pathogenesis and Control of Chronic Infections. INSERM, University of Montpellier, Etablissement Français du Sang, CHU Montpellier, Montpellier, France
| | - Vasiliki Kalatzis
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France.
| | - Sara Salinas
- Pathogenesis and Control of Chronic Infections, INSERM, Etablissement Français du Sang, University of Montpellier, Montpellier, France.
| |
Collapse
|
25
|
Mitsios A, Dubis AM, Moosajee M. Choroideremia: from genetic and clinical phenotyping to gene therapy and future treatments. Ther Adv Ophthalmol 2018; 10:2515841418817490. [PMID: 30627697 PMCID: PMC6311551 DOI: 10.1177/2515841418817490] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/05/2018] [Indexed: 11/15/2022] Open
Abstract
Choroideremia is an X-linked inherited chorioretinal dystrophy leading to blindness by late adulthood. Choroideremia is caused by mutations in the CHM gene which encodes Rab escort protein 1 (REP1), an ubiquitously expressed protein involved in intracellular trafficking and prenylation activity. The exact site of pathogenesis remains unclear but results in degeneration of the photoreceptors, retinal pigment epithelium and choroid. Animal and stem cell models have been used to study the molecular defects in choroideremia and test effectiveness of treatment interventions. Natural history studies of choroideremia have provided additional insight into the clinical phenotype of the condition and prepared the way for clinical trials aiming to investigate the safety and efficacy of suitable therapies. In this review, we provide a summary of the current knowledge on the genetics, pathophysiology, clinical features and therapeutic strategies that might become available for choroideremia in the future, including gene therapy, stem cell treatment and small-molecule drugs with nonsense suppression action.
Collapse
Affiliation(s)
- Andreas Mitsios
- Institute of Ophthalmology, University College London, London, UK
| | - Adam M Dubis
- Institute of Ophthalmology, University College London, London, UK
| | - Mariya Moosajee
- Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
26
|
Bayó-Puxan N, Terrasso AP, Creyssels S, Simão D, Begon-Pescia C, Lavigne M, Salinas S, Bernex F, Bosch A, Kalatzis V, Levade T, Cuervo AM, Lory P, Consiglio A, Brito C, Kremer EJ. Lysosomal and network alterations in human mucopolysaccharidosis type VII iPSC-derived neurons. Sci Rep 2018; 8:16644. [PMID: 30413728 PMCID: PMC6226539 DOI: 10.1038/s41598-018-34523-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/19/2018] [Indexed: 01/04/2023] Open
Abstract
Mucopolysaccharidosis type VII (MPS VII) is a lysosomal storage disease caused by deficient β-glucuronidase (β-gluc) activity. Significantly reduced β-gluc activity leads to accumulation of glycosaminoglycans (GAGs) in many tissues, including the brain. Numerous combinations of mutations in GUSB (the gene that codes for β-gluc) cause a range of neurological features that make disease prognosis and treatment challenging. Currently, there is little understanding of the molecular basis for MPS VII brain anomalies. To identify a neuronal phenotype that could be used to complement genetic analyses, we generated two iPSC clones derived from skin fibroblasts of an MPS VII patient. We found that MPS VII neurons exhibited reduced β-gluc activity and showed previously established disease-associated phenotypes, including GAGs accumulation, expanded endocytic compartments, accumulation of lipofuscin granules, more autophagosomes, and altered lysosome function. Addition of recombinant β-gluc to MPS VII neurons, which mimics enzyme replacement therapy, restored disease-associated phenotypes to levels similar to the healthy control. MPS VII neural cells cultured as 3D neurospheroids showed upregulated GFAP gene expression, which was associated with astrocyte reactivity, and downregulation of GABAergic neuron markers. Spontaneous calcium imaging analysis of MPS VII neurospheroids showed reduced neuronal activity and altered network connectivity in patient-derived neurospheroids compared to a healthy control. These results demonstrate the interplay between reduced β-gluc activity, GAG accumulation and alterations in neuronal activity, and provide a human experimental model for elucidating the bases of MPS VII-associated cognitive defects.
Collapse
Affiliation(s)
- Neus Bayó-Puxan
- Institute de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
| | - Ana Paula Terrasso
- iBET - Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Sophie Creyssels
- Institute de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Daniel Simão
- iBET - Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Christina Begon-Pescia
- Institute de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Marina Lavigne
- Institute de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Sara Salinas
- Institute de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | | | - Assumpció Bosch
- Departament Bioquímica i Biologia Molecular, and Center of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma Barcelona, Bellaterra, Spain
| | | | - Thierry Levade
- Laboratoire de Biochimie Métabolique, IFB, CHU Purpan, and Inserm 1037, CRCT, University Paul Sabatier Toulouse-III, Toulouse, France
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology and Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Philippe Lory
- IGF, CNRS, Inserm, University Montpellier, Montpellier, France
| | - Antonella Consiglio
- Department of Pathology and Experimental Therapeutics, Bellvitge University Hospital-IDIBELL, Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, BS, Italy
| | - Catarina Brito
- iBET - Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
- The Discoveries Centre for Regenerative and Precision Medicine, NOVA University Lisbon, Av da República, 2780-157 Oeiras, Portugal.
| | - Eric J Kremer
- Institute de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| |
Collapse
|
27
|
Vaché C, Torriano S, Faugère V, Erkilic N, Baux D, Garcia-Garcia G, Hamel CP, Meunier I, Zanlonghi X, Koenig M, Kalatzis V, Roux AF. Pathogenicity of novel atypical variants leading to choroideremia as determined by functional analyses. Hum Mutat 2018; 40:31-35. [DOI: 10.1002/humu.23671] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Christel Vaché
- Laboratoire de Génétique Moléculaire; CHU de Montpellier; Université de Montpellier; Montpellier France
| | - Simona Torriano
- Institut des Neurosciences de Montpellier; INSERM; Université de Montpellier; Montpellier France
| | - Valérie Faugère
- Laboratoire de Génétique Moléculaire; CHU de Montpellier; Université de Montpellier; Montpellier France
| | - Nejla Erkilic
- Institut des Neurosciences de Montpellier; INSERM; Université de Montpellier; Montpellier France
| | - David Baux
- Laboratoire de Génétique Moléculaire; CHU de Montpellier; Université de Montpellier; Montpellier France
| | - Gema Garcia-Garcia
- Laboratoire de Génétique Moléculaire; CHU de Montpellier; Université de Montpellier; Montpellier France
| | - Christian P. Hamel
- Institut des Neurosciences de Montpellier; INSERM; Université de Montpellier; Montpellier France
- Centre de Référence Maladies Sensorielles Génétiques; CHU de Montpellier; Université de Montpellier; Montpellier France
| | - Isabelle Meunier
- Institut des Neurosciences de Montpellier; INSERM; Université de Montpellier; Montpellier France
- Centre de Référence Maladies Sensorielles Génétiques; CHU de Montpellier; Université de Montpellier; Montpellier France
| | - Xavier Zanlonghi
- Centre de Compétence Maladie Rares; Clinique Pluridisciplinaire Jules Verne; Nantes France
| | - Michel Koenig
- Laboratoire de Génétique Moléculaire; CHU de Montpellier; Université de Montpellier; Montpellier France
| | - Vasiliki Kalatzis
- Institut des Neurosciences de Montpellier; INSERM; Université de Montpellier; Montpellier France
| | - Anne-Françoise Roux
- Laboratoire de Génétique Moléculaire; CHU de Montpellier; Université de Montpellier; Montpellier France
| |
Collapse
|
28
|
Foltz LP, Clegg DO. Patient-derived induced pluripotent stem cells for modelling genetic retinal dystrophies. Prog Retin Eye Res 2018; 68:54-66. [PMID: 30217765 DOI: 10.1016/j.preteyeres.2018.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 09/05/2018] [Accepted: 09/07/2018] [Indexed: 12/22/2022]
Abstract
The human retina is a highly complex tissue that makes up an integral part of our central nervous system. It is astonishing that our retina works seamlessly to provide one of our most critical senses, and it is equally devastating when a disease destroys a portion of the retina and robs people of their vision. After decades of research, scientists are beginning to understand retinal cells in a way that can benefit the millions of individuals suffering from inherited blindness. This understanding has come about in part with the ability to culture human embryonic stem cells and the innovation of induced pluripotent stem cells, which can be cultured from patients and used to model their disease. In this review, we highlight the successes of specific disease modelling studies and resulting molecular discoveries. The greatest strides in cellular modelling have come from mutations in genes with established and well-understood cellular functions in the context of the retina. We believe that the future of cellular modelling depends on emphasising reproducible production of retinal cell types, demonstrating functional rescue using site-specific programmable nucleases, and shifting towards unbiased screening using next generation sequencing.
Collapse
Affiliation(s)
- Leah P Foltz
- Biochemistry and Molecular Biology, University of California, Santa Barbara, CA, USA; Center for Stem Cell Biology and Engineering, University of California, Santa Barbara, CA, USA.
| | - Dennis O Clegg
- Biochemistry and Molecular Biology, University of California, Santa Barbara, CA, USA; Center for Stem Cell Biology and Engineering, University of California, Santa Barbara, CA, USA
| |
Collapse
|
29
|
Application of CRISPR/Cas9 technologies combined with iPSCs in the study and treatment of retinal degenerative diseases. Hum Genet 2018; 137:679-688. [DOI: 10.1007/s00439-018-1933-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 09/01/2018] [Indexed: 12/27/2022]
|
30
|
Patrício MI, Barnard AR, Xue K, MacLaren RE. Choroideremia: molecular mechanisms and development of AAV gene therapy. Expert Opin Biol Ther 2018; 18:807-820. [DOI: 10.1080/14712598.2018.1484448] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Maria I Patrício
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Alun R Barnard
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
31
|
Song JY, Aravand P, Nikonov S, Leo L, Lyubarsky A, Bennicelli JL, Pan J, Wei Z, Shpylchak I, Herrera P, Bennett DJ, Commins N, Maguire AM, Pham J, den Hollander AI, Cremers FPM, Koenekoop RK, Roepman R, Nishina P, Zhou S, Pan W, Ying GS, Aleman TS, de Melo J, McNamara I, Sun J, Mills J, Bennett J. Amelioration of Neurosensory Structure and Function in Animal and Cellular Models of a Congenital Blindness. Mol Ther 2018; 26:1581-1593. [PMID: 29673930 PMCID: PMC5986734 DOI: 10.1016/j.ymthe.2018.03.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 03/14/2018] [Accepted: 03/18/2018] [Indexed: 02/08/2023] Open
Abstract
Most genetically distinct inherited retinal degenerations are primary photoreceptor degenerations. We selected a severe early onset form of Leber congenital amaurosis (LCA), caused by mutations in the gene LCA5, in order to test the efficacy of gene augmentation therapy for a ciliopathy. The LCA5-encoded protein, Lebercilin, is essential for the trafficking of proteins and vesicles to the photoreceptor outer segment. Using the AAV serotype AAV7m8 to deliver a human LCA5 cDNA into an Lca5 null mouse model of LCA5, we show partial rescue of retinal structure and visual function. Specifically, we observed restoration of rod-and-cone-driven electroretinograms in about 25% of injected eyes, restoration of pupillary light responses in the majority of treated eyes, an ∼20-fold decrease in target luminance necessary for visually guided behavior, and improved retinal architecture following gene transfer. Using LCA5 patient-derived iPSC-RPEs, we show that delivery of the LCA5 cDNA restores lebercilin protein and rescues cilia quantity. The results presented in this study support a path forward aiming to develop safety and efficacy trials for gene augmentation therapy in human subjects with LCA5 mutations. They also provide the framework for measuring the effects of intervention in ciliopathies and other severe, early-onset blinding conditions.
Collapse
Affiliation(s)
- Ji Yun Song
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Puya Aravand
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sergei Nikonov
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Penn Vision Research Center, University of Pennsylvania Perelman, Philadelphia, PA, USA
| | - Lanfranco Leo
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arkady Lyubarsky
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Penn Vision Research Center, University of Pennsylvania Perelman, Philadelphia, PA, USA
| | - Jeannette L Bennicelli
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jieyan Pan
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zhangyong Wei
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ivan Shpylchak
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Pamela Herrera
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Bennett
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nicoletta Commins
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Albert M Maguire
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jennifer Pham
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Anneke I den Hollander
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Frans P M Cremers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Robert K Koenekoop
- McGill Ocular Genetics Center, McGill University Health Center, Montreal, QC, Canada
| | - Ronald Roepman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Shangzhen Zhou
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Wei Pan
- Penn Vision Research Center, University of Pennsylvania Perelman, Philadelphia, PA, USA; The Jackson Laboratory, Bar Harbor, ME, USA
| | - Gui-Shuang Ying
- Penn Vision Research Center, University of Pennsylvania Perelman, Philadelphia, PA, USA; Center for Preventive Ophthalmology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Tomas S Aleman
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jimmy de Melo
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ilan McNamara
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Junwei Sun
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jason Mills
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jean Bennett
- Center for Advanced Retinal and Ocular Therapeutics (CAROT) and F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Penn Vision Research Center, University of Pennsylvania Perelman, Philadelphia, PA, USA.
| |
Collapse
|
32
|
The effect of PTC124 on choroideremia fibroblasts and iPSC-derived RPE raises considerations for therapy. Sci Rep 2018; 8:8234. [PMID: 29844446 PMCID: PMC5974348 DOI: 10.1038/s41598-018-26481-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 05/04/2018] [Indexed: 11/24/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are caused by mutations in over 200 genes, resulting in a range of therapeutic options. Translational read-through inducing drugs (TRIDs) offer the possibility of treating multiple IRDs regardless of the causative gene. TRIDs promote ribosomal misreading of premature stop codons, which results in the incorporation of a near-cognate amino acid to produce a full-length protein. The IRD choroideremia (CHM) is a pertinent candidate for TRID therapy, as nonsense variants cause 30% of cases. Recently, treatment of the UAA nonsense-carrying CHM zebrafish model with the TRID PTC124 corrected the underlying biochemical defect and improved retinal phenotype. To be clinically relevant, we studied PTC124 efficiency in UAA nonsense-carrying human fibroblasts and induced pluripotent stem cell-derived retinal pigment epithelium, as well as in a UAA-mutated CHM overexpression system. We showed that PTC124 treatment induces a non-significant trend for functional rescue, which could not be improved by nonsense-mediated decay inhibition. Furthermore, it does not produce a detectable CHM-encoded protein even when coupled with a proteasome inhibitor. We suggest that drug efficiency may depend upon on the target amino acid and its evolutionary conservation, and argue that patient cells should be screened in vitro prior to inclusion in a clinical trial.
Collapse
|
33
|
Xue K, MacLaren RE. Ocular gene therapy for choroideremia: clinical trials and future perspectives. EXPERT REVIEW OF OPHTHALMOLOGY 2018; 13:129-138. [PMID: 31105764 DOI: 10.1080/17469899.2018.1475232] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Introduction Gene therapy offers the potential for targeted replacement of single gene defects in inherited retinal degenerations. Areas covered Choroideremia is an X-linked blinding retinal disease resulting from deficiency of the CHM gene product, REP1. The disease represents an ideal target for retinal gene therapy, as it is readily diagnosed in the clinic, relatively homogenous in phenotype and slow progressing, thereby providing a wide therapeutic window for intervention. Ongoing clinical trials of retinal gene therapy for choroideremia using an adeno-associated viral vector have demonstrated safety and early efficacy. We review the clinical characteristics of the disease with a view to interpreting the findings of gene therapy clinical trials and discuss future directions. Expert commentary Choroideremia gene therapy has so far demonstrated good safety profile and early functional visual acuity gains in a proportion of trial participants, which appear to be sustained.
Collapse
Affiliation(s)
- Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford & Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford & Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
34
|
Guiding Lights in Genome Editing for Inherited Retinal Disorders: Implications for Gene and Cell Therapy. Neural Plast 2018; 2018:5056279. [PMID: 29853845 PMCID: PMC5964415 DOI: 10.1155/2018/5056279] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/18/2018] [Indexed: 12/26/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are a leading cause of visual impairment in the developing world. These conditions present an irreversible dysfunction or loss of neural retinal cells, which significantly impacts quality of life. Due to the anatomical accessibility and immunoprivileged status of the eye, ophthalmological research has been at the forefront of innovative and advanced gene- and cell-based therapies, both of which represent great potential as therapeutic treatments for IRD patients. However, due to a genetic and clinical heterogeneity, certain IRDs are not candidates for these approaches. New advances in the field of genome editing using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR-associated protein (Cas) have provided an accurate and efficient way to edit the human genome and represent an appealing alternative for treating IRDs. We provide a brief update on current gene augmentation therapies for retinal dystrophies. Furthermore, we discuss recent advances in the field of genome editing and stem cell technologies, which together enable precise and personalized therapies for patients. Lastly, we highlight current technological limitations and barriers that need to be overcome before this technology can become a viable treatment option for patients.
Collapse
|
35
|
Patrício MI, Barnard AR, Cox CI, Blue C, MacLaren RE. The Biological Activity of AAV Vectors for Choroideremia Gene Therapy Can Be Measured by In Vitro Prenylation of RAB6A. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 9:288-295. [PMID: 29707603 PMCID: PMC5918179 DOI: 10.1016/j.omtm.2018.03.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/25/2018] [Indexed: 11/23/2022]
Abstract
Choroideremia (CHM) is a rare, X-linked recessive retinal dystrophy caused by mutations in the CHM gene. CHM is ubiquitously expressed in human cells and encodes Rab escort protein 1 (REP1). REP1 plays a key role in intracellular trafficking through the prenylation of Rab GTPases, a reaction that can be reproduced in vitro. With recent advances in adeno-associated virus (AAV) gene therapy for CHM showing gene replacement to be a promising approach, an assay to assess the biological activity of the vectors is of the uttermost importance. Here we sought to compare the response of two Rab proteins, RAB27A and RAB6A, to the incorporation of a biotinylated lipid donor in a prenylation reaction in vitro. First, we found the expression of REP1 to be proportional to the amount of recombinant AAV (rAAV)2/2-REP1 used to transduce the cells. Second, prenylation of RAB6A appeared to be more sensitive to REP1 protein expression than prenylation of RAB27A. Moreover, the method was reproducible in other cell lines. These results support the further development of a prenylation reaction using a biotinylated lipid donor and RAB6A to assess the biological activity of AAV vectors for CHM gene therapy.
Collapse
Affiliation(s)
- Maria I Patrício
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Oxford, UK.,Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Alun R Barnard
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Oxford, UK.,Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Christopher I Cox
- Nightstar Therapeutics, Wellcome Gibbs Building, 215 Euston Road, London, UK
| | - Clare Blue
- Nightstar Therapeutics, Wellcome Gibbs Building, 215 Euston Road, London, UK
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Oxford, UK.,Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
36
|
Duong TT, Vasireddy V, Ramachandran P, Herrera PS, Leo L, Merkel C, Bennett J, Mills JA. Use of induced pluripotent stem cell models to probe the pathogenesis of Choroideremia and to develop a potential treatment. Stem Cell Res 2018; 27:140-150. [PMID: 29414605 DOI: 10.1016/j.scr.2018.01.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/04/2018] [Accepted: 01/06/2018] [Indexed: 12/17/2022] Open
Abstract
Choroideremia (CHM) is a rare monogenic, X-linked recessive inherited retinal degeneration resulting from mutations in the Rab Escort Protein-1 (REP1) encoding CHM gene. The primary retinal cell type leading to CHM is unknown. In this study, we explored the utility of induced pluripotent stem cell-derived models of retinal pigmented epithelium (iPSC-RPE) to study disease pathogenesis and a potential gene-based intervention in four different genetically distinct forms of CHM. A number of abnormal cell biologic, biochemical, and physiologic functions were identified in the CHM mutant cells. We then identified a recombinant adeno-associated virus (AAV) serotype, AAV7m8, that is optimal for both delivering transgenes to iPSC-RPEs as well as to appropriate target cells (RPE cells and rod photoreceptors) in the primate retina. To establish the proof of concept of AAV7m8 mediated CHM gene therapy, we developed AAV7m8.hCHM, which delivers the human CHM cDNA under control of CMV-enhanced chicken β-actin promoter (CßA). Delivery of AAV7m8.hCHM to CHM iPSC-RPEs restored protein prenylation, trafficking and phagocytosis. The results confirm that AAV-mediated delivery of the REP1-encoding gene can rescue defects in CHM iPSC-RPE regardless of the type of disease-causing mutation. The results also extend our understanding of mechanisms involved in the pathophysiology of choroideremia.
Collapse
Affiliation(s)
- Thu T Duong
- F.M. Kirby Center for Molecular Ophthalmology and Center for Advanced Retinal and Ocular Therapeutics (CAROT), Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, PA 19104, USA
| | - Vidyullatha Vasireddy
- F.M. Kirby Center for Molecular Ophthalmology and Center for Advanced Retinal and Ocular Therapeutics (CAROT), Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, PA 19104, USA
| | - Pavitra Ramachandran
- F.M. Kirby Center for Molecular Ophthalmology and Center for Advanced Retinal and Ocular Therapeutics (CAROT), Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, PA 19104, USA
| | - Pamela S Herrera
- F.M. Kirby Center for Molecular Ophthalmology and Center for Advanced Retinal and Ocular Therapeutics (CAROT), Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, PA 19104, USA
| | - Lanfranco Leo
- F.M. Kirby Center for Molecular Ophthalmology and Center for Advanced Retinal and Ocular Therapeutics (CAROT), Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, PA 19104, USA
| | - Carrie Merkel
- F.M. Kirby Center for Molecular Ophthalmology and Center for Advanced Retinal and Ocular Therapeutics (CAROT), Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, PA 19104, USA
| | - Jean Bennett
- F.M. Kirby Center for Molecular Ophthalmology and Center for Advanced Retinal and Ocular Therapeutics (CAROT), Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, PA 19104, USA
| | - Jason A Mills
- F.M. Kirby Center for Molecular Ophthalmology and Center for Advanced Retinal and Ocular Therapeutics (CAROT), Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, PA 19104, USA.
| |
Collapse
|
37
|
Torriano S, Erkilic N, Faugère V, Damodar K, Hamel CP, Roux AF, Kalatzis V. Pathogenicity of a novel missense variant associated with choroideremia and its impact on gene replacement therapy. Hum Mol Genet 2018; 26:3573-3584. [PMID: 28911202 DOI: 10.1093/hmg/ddx244] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/19/2017] [Indexed: 12/15/2022] Open
Abstract
Choroideremia (CHM) is an inherited retinal dystrophy characterised by progressive degeneration of photoreceptors, retinal pigment epithelium (RPE) and underlying choroid. It is caused by loss-of-function mutations in CHM, which has an X-linked inheritance, and is thus an ideal candidate for gene replacement strategies. CHM encodes REP1, which plays a key role in the prenylation of Rab GTPases. We recently showed that an induced pluripotent stem cell (iPSc)-derived RPE model for CHM is fully functional and reproduces the underlying prenylation defect. This criterion can thus be used for testing the pathogenic nature of novel variants. Until recently, missense variants were not associated with CHM. Currently, at least nine such variants have been reported but only two have been shown to be pathogenic. We report here the characterisation of the third pathogenic missense CHM variant, p.Leu457Pro. Clinically, the associated phenotype is indistinguishable from that of loss-of-function mutations. By contrast, this missense variant results in wild type CHM expression levels and detectable levels of mutant protein. The prenylation status of patient-specific fibroblasts and iPSc-derived RPE is within the range observed for loss-of-function mutations, consistent with the clinical phenotype. Lastly, considering the current climate of CHM gene therapy, we assayed whether the presence of mutant REP1 could interfere with a gene replacement strategy by testing the prenylation status of patient-specific iPSc-derived RPE following AAV-mediated gene transfer. Our results show that correction of the functional defect is possible and highlight the predictive value of these models for therapy screening prior to inclusion in clinical trials.
Collapse
Affiliation(s)
- Simona Torriano
- Inserm U1051, Institute for Neurosciences of Montpellier, Montpellier, France.,University of Montpellier, Montpellier, France
| | - Nejla Erkilic
- Inserm U1051, Institute for Neurosciences of Montpellier, Montpellier, France.,University of Montpellier, Montpellier, France
| | | | - Krishna Damodar
- Inserm U1051, Institute for Neurosciences of Montpellier, Montpellier, France.,University of Montpellier, Montpellier, France
| | - Christian P Hamel
- Inserm U1051, Institute for Neurosciences of Montpellier, Montpellier, France.,University of Montpellier, Montpellier, France.,Department of Ophthalmology, CHRU, Montpellier, France.,Centre of Reference for Genetic Sensory Diseases, CHRU, Montpellier, France
| | - Anne-Francoise Roux
- Laboratory of Molecular Genetics, CHRU, Montpellier, France.,Laboratory of Rare Genetic Diseases, EA 7402, University of Montpellier, Montpellier, France
| | - Vasiliki Kalatzis
- Inserm U1051, Institute for Neurosciences of Montpellier, Montpellier, France.,University of Montpellier, Montpellier, France
| |
Collapse
|
38
|
Quinn PM, Buck TM, Ohonin C, Mikkers HMM, Wijnholds J. Production of iPS-Derived Human Retinal Organoids for Use in Transgene Expression Assays. Methods Mol Biol 2018; 1715:261-273. [PMID: 29188520 DOI: 10.1007/978-1-4939-7522-8_19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In vitro retinal organoid modeling from human pluripotent stem cells is becoming more common place in many ophthalmic laboratories worldwide. These organoids mimic human retinogenesis through formation of organized layered retinal structures that display markers for typical retinal cell types. Pivotally these humanized retinal models provide a stepping stone to the clinic as therapeutic tools and are expected to provide a promising alternative to current animal models. Thus pluripotent stem cell based healthy as well as diseased human retinal organoids are attractive for use in drug potency assays and gene augmentation therapeutics. Here we outline an established protocol for generation of these retinal organoids and how they can be used in conjunction with adeno-associated virus vectors for transgene expression assays.
Collapse
Affiliation(s)
- Peter M Quinn
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Thilo M Buck
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Charlotte Ohonin
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Harald M M Mikkers
- Department of Molecular Cell Biology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Leiden, The Netherlands. .,Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands.
| |
Collapse
|
39
|
Retinal Gene Therapy for Choroideremia: In Vitro Testing for Gene Augmentation Using an Adeno-Associated Viral (AAV) Vector. Methods Mol Biol 2017. [PMID: 29188508 DOI: 10.1007/978-1-4939-7522-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
As gene therapy of choroideremia is becoming a clinical reality, there is a need for reliable and sensitive assays to determine the expression of exogenously delivered Rab Escort Protein-1 (REP1), in particular to test new gene therapy vectors and as a quality control screen for clinical vector stocks. Here we describe an in vitro protocol to test transgene expression following AAV2/2-REP1 transduction of a human cell line. Gene augmentation can be confirmed by western blot and quantification of the fold-increase of human REP1 levels over untransduced controls.
Collapse
|
40
|
Gupta PR, Huckfeldt RM. Gene therapy for inherited retinal degenerations: initial successes and future challenges. J Neural Eng 2017; 14:051002. [DOI: 10.1088/1741-2552/aa7a27] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
41
|
Weed LS, Mills JA. Strategies for retinal cell generation from human pluripotent stem cells. Stem Cell Investig 2017; 4:65. [PMID: 28815176 DOI: 10.21037/sci.2017.07.02] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/24/2017] [Indexed: 12/22/2022]
Abstract
Induced pluripotent stem cells (iPSCs) are specialized self-renewing cells that are generated by exogenously expressing pluripotency-associated transcription factors in somatic cells such as fibroblasts, peripheral blood mononuclear cells, or lymphoblastoid cell lines (LCLs). iPSCs are functionally similar to naturally pluripotent embryonic stem cells (ESCs) in their capacity to propagate indefinitely and potential to differentiate into all human cell types, and are devoid of the associated ethical complications of origin. iPSCs are useful for studying embryonic development, disease modeling, and drug screening. Additionally, iPSCs provide a personalized approach for pathological studies, particularly for diseases that lack appropriate animal models. Retinal cell differentiations using iPSCs have been successful in this regard. Several protocols to generate various retinal cells have been developed to maximize a specific cell type or, most recently, to mimic in vivo retinal structure and cellular environment. As differentiation protocols continue to improve we are likely to see an increase in our basic understanding of various retinal degenerative diseases and the utilization of iPSCs in clinical trials.
Collapse
Affiliation(s)
- Lindsey S Weed
- Center for Advanced Retinal and Ocular Therapeutics, F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jason A Mills
- Center for Advanced Retinal and Ocular Therapeutics, F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
42
|
Peng YQ, Tang LS, Yoshida S, Zhou YD. Applications of CRISPR/Cas9 in retinal degenerative diseases. Int J Ophthalmol 2017; 10:646-651. [PMID: 28503441 DOI: 10.18240/ijo.2017.04.23] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/09/2017] [Indexed: 02/06/2023] Open
Abstract
Gene therapy is a potentially effective treatment for retinal degenerative diseases. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system has been developed as a new genome-editing tool in ophthalmic studies. Recent advances in researches showed that CRISPR/Cas9 has been applied in generating animal models as well as gene therapy in vivo of retinitis pigmentosa (RP) and leber congenital amaurosis (LCA). It has also been shown as a potential attempt for clinic by combining with other technologies such as adeno-associated virus (AAV) and induced pluripotent stem cells (iPSCs). In this review, we highlight the main points of further prospect of using CRISPR/Cas9 in targeting retinal degeneration. We also emphasize the potential applications of this technique in treating retinal degenerative diseases.
Collapse
Affiliation(s)
- Ying-Qian Peng
- Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Luo-Sheng Tang
- Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Ye-Di Zhou
- Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
43
|
Zika Virus Efficiently Replicates in Human Retinal Epithelium and Disturbs Its Permeability. J Virol 2017; 91:JVI.02144-16. [PMID: 27852856 DOI: 10.1128/jvi.02144-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
44
|
Ertl HCJ, High KA. Impact of AAV Capsid-Specific T-Cell Responses on Design and Outcome of Clinical Gene Transfer Trials with Recombinant Adeno-Associated Viral Vectors: An Evolving Controversy. Hum Gene Ther 2016; 28:328-337. [PMID: 28042943 DOI: 10.1089/hum.2016.172] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Recombinant adenovirus-associated (rAAV) vectors due to their ease of construction, wide tissue tropism, and lack of pathogenicity remain at the forefront for long-term gene replacement therapy. In spite of very encouraging preclinical results, clinical trials were initially unsuccessful; expression of the rAAV vector-delivered therapeutic protein was transient. Loss of expression was linked to an expansion of AAV capsid-specific T-cell responses, leading to the hypothesis that rAAV vectors recall pre-existing memory T cells that had been induced by natural infections with AAV together with a helper virus. Although this was hotly debated at first, AAV capsid-specific T-cell responses were observed in several gene transfer trials that used high doses of rAAV vectors. Subsequent trials designed to circumvent these T-cell responses through the use of immunosuppressive drugs, rAAV vectors based on rare serotypes, or modified to allow for therapeutic levels of the transgene product at low, non-immunogenic vector doses are now successful in correcting debilitating diseases.
Collapse
|
45
|
Wiley LA, Burnight ER, Drack AV, Banach BB, Ochoa D, Cranston CM, Madumba RA, East JS, Mullins RF, Stone EM, Tucker BA. Using Patient-Specific Induced Pluripotent Stem Cells and Wild-Type Mice to Develop a Gene Augmentation-Based Strategy to Treat CLN3-Associated Retinal Degeneration. Hum Gene Ther 2016; 27:835-846. [PMID: 27400765 DOI: 10.1089/hum.2016.049] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Juvenile neuronal ceroid lipofuscinosis (JNCL) is a childhood neurodegenerative disease with early-onset, severe central vision loss. Affected children develop seizures and CNS degeneration accompanied by severe motor and cognitive deficits. There is no cure for JNCL, and patients usually die during the second or third decade of life. In this study, independent lines of induced pluripotent stem cells (iPSCs) were generated from two patients with molecularly confirmed mutations in CLN3, the gene mutated in JNCL. Clinical-grade adeno-associated adenovirus serotype 2 (AAV2) carrying the full-length coding sequence of human CLN3 was generated in a U.S. Food and Drug Administration-registered cGMP facility. AAV2-CLN3 was efficacious in restoring full-length CLN3 transcript and protein in patient-specific fibroblasts and iPSC-derived retinal neurons. When injected into the subretinal space of wild-type mice, purified AAV2-CLN3 did not show any evidence of retinal toxicity. This study provides proof-of-principle for initiation of a clinical trial using AAV-mediated gene augmentation for the treatment of children with CLN3-associated retinal degeneration.
Collapse
Affiliation(s)
- Luke A Wiley
- Department of Ophthalmology and Visual Sciences, Stephen A. Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Erin R Burnight
- Department of Ophthalmology and Visual Sciences, Stephen A. Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Arlene V Drack
- Department of Ophthalmology and Visual Sciences, Stephen A. Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Bailey B Banach
- Department of Ophthalmology and Visual Sciences, Stephen A. Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Dalyz Ochoa
- Department of Ophthalmology and Visual Sciences, Stephen A. Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Cathryn M Cranston
- Department of Ophthalmology and Visual Sciences, Stephen A. Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Robert A Madumba
- Department of Ophthalmology and Visual Sciences, Stephen A. Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Jade S East
- Department of Ophthalmology and Visual Sciences, Stephen A. Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Robert F Mullins
- Department of Ophthalmology and Visual Sciences, Stephen A. Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Edwin M Stone
- Department of Ophthalmology and Visual Sciences, Stephen A. Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Budd A Tucker
- Department of Ophthalmology and Visual Sciences, Stephen A. Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| |
Collapse
|
46
|
Moosajee M, Tracey-White D, Smart M, Weetall M, Torriano S, Kalatzis V, da Cruz L, Coffey P, Webster AR, Welch E. Functional rescue of REP1 following treatment with PTC124 and novel derivative PTC-414 in human choroideremia fibroblasts and the nonsense-mediated zebrafish model. Hum Mol Genet 2016; 25:3416-3431. [DOI: 10.1093/hmg/ddw184] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 06/03/2016] [Accepted: 06/06/2016] [Indexed: 01/15/2023] Open
|
47
|
De Vos J, Bouckenheimer J, Sansac C, Lemaître JM, Assou S. Human induced pluripotent stem cells: A disruptive innovation. Curr Res Transl Med 2016; 64:91-6. [PMID: 27316392 DOI: 10.1016/j.retram.2016.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/04/2016] [Accepted: 04/08/2016] [Indexed: 12/31/2022]
Abstract
This year (2016) will mark the 10th anniversary of the discovery of induced pluripotent stem cells (iPSCs). The finding that the transient expression of four transcription factors can radically remodel the epigenome, transcriptome and metabolome of differentiated cells and reprogram them into pluripotent stem cells has been a major and groundbreaking technological innovation. In this review, we discuss the major applications of this technology that we have grouped in nine categories: a model to study cell fate control; a model to study pluripotency; a model to study human development; a model to study human tissue and organ physiology; a model to study genetic diseases in a dish; a tool for cell rejuvenation; a source of cells for drug screening; a source of cells for regenerative medicine; a tool for the production of human organs in animals.
Collapse
Affiliation(s)
- J De Vos
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, 34000 Montpellier, France; INSERM, U1183, 34000 Montpellier, France; Université de Montpellier, UFR de Médecine, 34000 Montpellier, France; Institut de Biologie Computationnelle, 34000 Montpellier, France; CHU Montpellier, SAFE-IPS Reprogramming Platform, Institute of Research in Biotherapy, 34000 Montpellier, France; CHU Montpellier, Unit for Cellular Therapy, Hospital Saint-Eloi, 34000 Montpellier, France.
| | - J Bouckenheimer
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, 34000 Montpellier, France; INSERM, U1183, 34000 Montpellier, France; Université de Montpellier, UFR de Pharmacie, 34000 Montpellier, France
| | - C Sansac
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, 34000 Montpellier, France; INSERM, U1183, 34000 Montpellier, France; Université de Montpellier, UFR de Pharmacie, 34000 Montpellier, France
| | - J-M Lemaître
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, 34000 Montpellier, France; INSERM, U1183, 34000 Montpellier, France
| | - S Assou
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, 34000 Montpellier, France; INSERM, U1183, 34000 Montpellier, France; Université de Montpellier, UFR de Médecine, 34000 Montpellier, France.
| |
Collapse
|
48
|
Update on ocular gene therapy and advances in treatment of inherited retinal diseases and exudative macular degeneration. Curr Opin Ophthalmol 2016; 27:268-73. [DOI: 10.1097/icu.0000000000000256] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
49
|
Song MJ, Bharti K. Looking into the future: Using induced pluripotent stem cells to build two and three dimensional ocular tissue for cell therapy and disease modeling. Brain Res 2016; 1638:2-14. [PMID: 26706569 PMCID: PMC4837038 DOI: 10.1016/j.brainres.2015.12.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/24/2015] [Accepted: 12/08/2015] [Indexed: 01/02/2023]
Abstract
Retinal degenerative diseases are the leading cause of irreversible vision loss in developed countries. In many cases the diseases originate in the homeostatic unit in the back of the eye that contains the retina, retinal pigment epithelium (RPE) and the choriocapillaris. RPE is a central and a critical component of this homeostatic unit, maintaining photoreceptor function and survival on the apical side and choriocapillaris health on the basal side. In diseases like age-related macular degeneration (AMD), it is thought that RPE dysfunctions cause disease-initiating events and as the RPE degenerates photoreceptors begin to die and patients start loosing vision. Patient-specific induced pluripotent stem (iPS) cell-derived RPE provides direct access to a patient's genetics and allow the possibility of identifying the initiating events of RPE-associated degenerative diseases. Furthermore, iPS cell-derived RPE cells are being tested as a potential cell replacement in disease stages with RPE atrophy. In this article we summarize the recent progress in the field of iPS cell-derived RPE "disease modeling" and cell therapies and also discuss the possibilities of developing a model of the entire homeostatic unit to aid in studying disease processes in the future. This article is part of a Special Issue entitled SI: PSC and the brain.
Collapse
Affiliation(s)
- Min Jae Song
- Unit on Ocular and Stem Cell Translational Research National Eye Institute, 10 Center Drive, Room 10B10, Bethesda, MD 20892, United States
| | - Kapil Bharti
- Unit on Ocular and Stem Cell Translational Research National Eye Institute, 10 Center Drive, Room 10B10, Bethesda, MD 20892, United States.
| |
Collapse
|
50
|
Personalized Medicine: Cell and Gene Therapy Based on Patient-Specific iPSC-Derived Retinal Pigment Epithelium Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 854:549-55. [PMID: 26427458 DOI: 10.1007/978-3-319-17121-0_73] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interest in generating human induced pluripotent stem (iPS) cells for stem cell modeling of diseases has overtaken that of patient-specific human embryonic stem cells due to the ethical, technical, and political concerns associated with the latter. In ophthalmology, researchers are currently using iPS cells to explore various applications, including: (1) modeling of retinal diseases using patient-specific iPS cells; (2) autologous transplantation of differentiated retinal cells that undergo gene correction at the iPS cell stage via gene editing tools (e.g., CRISPR/Cas9, TALENs and ZFNs); and (3) autologous transplantation of patient-specific iPS-derived retinal cells treated with gene therapy. In this review, we will discuss the uses of patient-specific iPS cells for differentiating into retinal pigment epithelium (RPE) cells, uncovering disease pathophysiology, and developing new treatments such as gene therapy and cell replacement therapy via autologous transplantation.
Collapse
|