1
|
Beryozkin A, Byrne LC. In Vivo Imaging of Rodent Retina in Retinal Disease. Methods Mol Biol 2025; 2848:151-167. [PMID: 39240522 DOI: 10.1007/978-1-0716-4087-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
High-quality imaging of the retina is crucial to the diagnosis and monitoring of disease, as well as for evaluating the success of therapeutics in human patients and in preclinical animal models. Here, we describe the basic principles and methods for in vivo retinal imaging in rodents, including fundus imaging, fluorescein angiography, optical coherence tomography, fundus autofluorescence, and infrared imaging. After providing a concise overview of each method and detailing the retinal diseases and conditions that can be visualized through them, we will proceed to discuss the advantages and disadvantages of each approach. These protocols will facilitate the acquisition of optimal images for subsequent quantification and analysis. Additionally, a brief explanation will be given regarding the potential results and the clinical significance of the detected abnormalities.
Collapse
Affiliation(s)
- Avigail Beryozkin
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leah C Byrne
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Pincay J, Rodriguez M, Kaushal D, Tsang SH. Rod-sparing in a bardet-biedl syndrome patient with mutations in the ARL6 gene. Doc Ophthalmol 2024; 149:133-138. [PMID: 39078565 DOI: 10.1007/s10633-024-09985-8] [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: 06/04/2024] [Accepted: 07/16/2024] [Indexed: 07/31/2024]
Abstract
PURPOSE Bardet-Biedl Syndrome (BBS) is an autosomal recessive disorder characterized by pleiotropism that affects multiple organ systems. The primary features of BBS include rod-cone dystrophy, renal anomalies, post axial polydactyly, and neurologic deficits. The clinical picture of BBS is extensively heterogenous, with inter and intra familial patients varying in levels of syndromic manifestations and severity of symptoms. METHODS In this study we examined a monocular BBS patient who was compound heterozygous for mutations in the ARL6 (BBS3) gene. RESULTS The patient reported visual complaints consistent with a clinical picture of cone or cone-rod dystrophy. Fundus imaging showed retinal mottling on color photos and a parafoveal hyperfluorescent ring on short wave autofluorescence (SW-AF). Full field electroretinogram (ffERG) revealed normal scotopic step tracings and diminished amplitudes in the photopic steps. CONCLUSION This rod-sparing result was consistent with cone-dystrophy and is the first known case of a rod-sparing ffERG phenotype in a BBS patient with mutations in the ARL6 gene. This contributes to the existing phenotype and may potentially contribute to furthering our understanding of BBS pathophysiology.
Collapse
Affiliation(s)
- Jorge Pincay
- Department of Ophthalmology, Columbia University Irving Medical Center, Vanderbilt Clinic 622 W 168th St 3rd Floor, New York, NY, 10032, USA
- State University of New York at Downstate Medical Center, Brooklyn, NY, USA
| | - Marilyn Rodriguez
- Department of Ophthalmology, Columbia University Irving Medical Center, Vanderbilt Clinic 622 W 168th St 3rd Floor, New York, NY, 10032, USA
| | - Divya Kaushal
- Department of Ophthalmology, Columbia University Irving Medical Center, Vanderbilt Clinic 622 W 168th St 3rd Floor, New York, NY, 10032, USA
| | - Stephen H Tsang
- Department of Ophthalmology, Columbia University Irving Medical Center, Vanderbilt Clinic 622 W 168th St 3rd Floor, New York, NY, 10032, USA.
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA.
| |
Collapse
|
3
|
Arsenijevic Y, Chang N, Mercey O, El Fersioui Y, Koskiniemi-Kuendig H, Joubert C, Bemelmans AP, Rivolta C, Banin E, Sharon D, Guichard P, Hamel V, Kostic C. Fine-tuning FAM161A gene augmentation therapy to restore retinal function. EMBO Mol Med 2024; 16:805-822. [PMID: 38504136 PMCID: PMC11018783 DOI: 10.1038/s44321-024-00053-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/21/2024] Open
Abstract
For 15 years, gene therapy has been viewed as a beacon of hope for inherited retinal diseases. Many preclinical investigations have centered around vectors with maximal gene expression capabilities, yet despite efficient gene transfer, minimal physiological improvements have been observed in various ciliopathies. Retinitis pigmentosa-type 28 (RP28) is the consequence of bi-allelic null mutations in the FAM161A, an essential protein for the structure of the photoreceptor connecting cilium (CC). In its absence, cilia become disorganized, leading to outer segment collapses and vision impairment. Within the human retina, FAM161A has two isoforms: the long one with exon 4, and the short one without it. To restore CC in Fam161a-deficient mice shortly after the onset of cilium disorganization, we compared AAV vectors with varying promoter activities, doses, and human isoforms. While all vectors improved cell survival, only the combination of both isoforms using the weak FCBR1-F0.4 promoter enabled precise FAM161A expression in the CC and enhanced retinal function. Our investigation into FAM161A gene replacement for RP28 emphasizes the importance of precise therapeutic gene regulation, appropriate vector dosing, and delivery of both isoforms. This precision is pivotal for secure gene therapy involving structural proteins like FAM161A.
Collapse
Affiliation(s)
- Yvan Arsenijevic
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland.
| | - Ning Chang
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
- Group for Retinal Disorder Research, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Olivier Mercey
- University of Geneva, Department of Molecular and Cellular Biology, Sciences III, Geneva, Switzerland
| | - Younes El Fersioui
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
- Group for Retinal Disorder Research, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Hanna Koskiniemi-Kuendig
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Caroline Joubert
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland
| | - Alexis-Pierre Bemelmans
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives: mécanismes, thérapies, imagerie, Fontenay-aux-Roses, France
| | - Carlo Rivolta
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Eyal Banin
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Faculty of Medicine, The Hebrew of Jerusalem, Jerusalem, Israel
| | - Dror Sharon
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Faculty of Medicine, The Hebrew of Jerusalem, Jerusalem, Israel
| | - Paul Guichard
- University of Geneva, Department of Molecular and Cellular Biology, Sciences III, Geneva, Switzerland
| | - Virginie Hamel
- University of Geneva, Department of Molecular and Cellular Biology, Sciences III, Geneva, Switzerland
| | - Corinne Kostic
- Group for Retinal Disorder Research, Department of Ophthalmology, University of Lausanne, Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Lausanne, Switzerland.
| |
Collapse
|
4
|
Shoemaker A. Bardet-Biedl syndrome: A clinical overview focusing on diagnosis, outcomes and best-practice management. Diabetes Obes Metab 2024; 26 Suppl 2:25-33. [PMID: 38383825 DOI: 10.1111/dom.15494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/16/2024] [Accepted: 01/27/2024] [Indexed: 02/23/2024]
Abstract
Bardet-Biedl syndrome (BBS) is a genetic disorder characterized by early-onset obesity, polydactyly, genital and kidney anomalies, developmental delay and vision loss due to rod-cone dystrophy. BBS is an autosomal recessive disorder with >20 implicated genes. The genotype-phenotype relationship in BBS is not clear, and there may be additional modifying factors. The underlying mechanism is dysfunction of primary cilia. In BBS, receptor trafficking in and out of the cilia is compromised, affecting multiple organ systems. Along with early-onset obesity, hyperphagia is a prominent symptom and contributes significantly to clinical morbidity and caregiver burden. While there is no cure for BBS, setmelanotide is a new pharmacotherapy approved for treatment of obesity in BBS. The differential diagnosis for BBS includes other ciliopathies, such as Alstrom syndrome, and other genetic obesity syndromes, such as Prader-Willi syndrome. Careful clinical history and genetic testing can help determine the diagnosis and a multidisciplinary team is necessary to guide clinical management.
Collapse
Affiliation(s)
- Ashley Shoemaker
- Division of Pediatric Endocrinology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
5
|
Hassan S, Hsu Y, Thompson JM, Kalmanek E, VandeLune JA, Stanley S, Drack AV. The dose-response relationship of subretinal gene therapy with rAAV2tYF-CB-h RS1 in a mouse model of X-linked retinoschisis. Front Med (Lausanne) 2024; 11:1304819. [PMID: 38414621 PMCID: PMC10898246 DOI: 10.3389/fmed.2024.1304819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/16/2024] [Indexed: 02/29/2024] Open
Abstract
Purpose X-linked retinoschisis (XLRS), due to loss-of-function mutations in the retinoschisin (RS1) gene, is characterized by a modest to severe decrease in visual acuity. Clinical trials for XLRS utilizing intravitreal (IVT) gene therapy showed ocular inflammation. We conducted a subretinal dose-response preclinical study using rAAV2tYF-CB-hRS1 utilizing the Rs1 knockout (Rs1-KO) mouse to investigate short- and long-term retinal rescue after subretinal gene delivery. Methods Rs1-KO mice were subretinally injected with 2 μL of rAAV2tYF-CB-hRS1 vector with 8E9 viral genomes (vg)/eye, 8E8 vg/eye, 8E7 vg/eye, or sham injection, and compared to untreated eyes. Reconstitution of human RS1 protein was detected using western blotting. Analysis of retinal function by electroretinography (ERG) and structural analysis by optical coherence tomography (OCT) were performed at 1, 2, 3, 5, 7, and 12 months post injection (MPI). Immunohistochemistry (IHC) was performed to evaluate cone rescue on the cellular level. Functional vision was evaluated using a visually guided swim assay (VGSA). Results Western blotting analysis showed human RS1 protein expression in a dose-dependent manner. Quantification of western blotting showed that the RS1 protein expression in mice treated with the 8E8 vg dose was near the wild-type (WT) expression levels. ERG demonstrated dose-dependent effects: At 1 MPI the 8E8 vg dose treated eyes had higher light-adapted (LA) ERG amplitudes in 3.0 flash and 5 Hz flicker compared to untreated (p < 0.0001) and sham-treated eyes (p < 0.0001) which persisted until the 12 MPI endpoint, consistent with improved cone function. ERG b-wave amplitudes were higher in response to dark-adapted (DA) 0.01 dim flash and 3.0 standard combined response (SCR) compared to sham-treated (p < 0.01) and untreated eyes (p < 0.001) which persisted until 3 MPI, suggesting short-term improvement of the rod photoreceptors. All injections, including sham-treated, resulted in a cyst severity score of 1 (no cavities), with significant reductions compared to untreated eyes up to 3 MPI (p < 0.05). The high and low dose groups showed inconsistent ERG improvements, despite reduced cyst severity, emphasizing the dose-dependent nature of gene augmentation's efficacy and the tenuous connection between cyst reduction and ERG improvement. IHC data showed a significant cone rescue in eyes treated with the 8E8 vg dose compared to sham-treated and untreated eyes. VGSA showed better functional vision in 8E8 vg dose treated mice. Eyes treated with the highest dose showed occasional localized degeneration in the outer nuclear layer. Conclusion Our data suggest that a dose of 8E8 vg/eye subretinally improves retinal function and structure in the Rs1-KO mouse. It improves cone function, rod function, and reduces cyst severity. Sham treatment resolves schisis cysts, but 8E8 vg/eye is needed for optimal retinal electrical function rescue. These findings offer a promising path for clinical translation to human trials.
Collapse
Affiliation(s)
- Salma Hassan
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Biomedical Science-Cell and Developmental Biology Graduate Program, Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, United States
| | - Ying Hsu
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Jacob M Thompson
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, United States
| | - Emily Kalmanek
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Joel A VandeLune
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Sarah Stanley
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Arlene V Drack
- Department of Ophthalmology and Visual Sciences, Institute for Vision Research, and Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Biomedical Science-Cell and Developmental Biology Graduate Program, Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, United States
- Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
6
|
He X, Fu Y, Ma L, Yao Y, Ge S, Yang Z, Fan X. AAV for Gene Therapy in Ocular Diseases: Progress and Prospects. RESEARCH (WASHINGTON, D.C.) 2023; 6:0291. [PMID: 38188726 PMCID: PMC10768554 DOI: 10.34133/research.0291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 11/27/2023] [Indexed: 01/09/2024]
Abstract
Owing to the promising therapeutic effect and one-time treatment advantage, gene therapy may completely change the management of eye diseases, especially retinal diseases. Adeno-associated virus (AAV) is considered one of the most promising viral gene delivery tools because it can infect various types of tissues and is considered as a relatively safe gene delivery vector. The eye is one of the most popular organs for gene therapy, since its limited volume is suitable for small doses of AAV stably transduction. Recently, an increasing number of clinical trials of AAV-mediated gene therapy are underway. This review summarizes the biological functions of AAV and its application in the treatment of various ocular diseases, as well as the characteristics of different AAV delivery routes in clinical applications. Here, the latest research progresses in AAV-mediated gene editing and silencing strategies to modify that the genetic ocular diseases are systematically outlined, especially by base editing and prime editing. We discuss the progress of AAV in ocular optogenetic therapy. We also summarize the application of AAV-mediated gene therapy in animal models and the difficulties in its clinical transformation.
Collapse
Affiliation(s)
- Xiaoyu He
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yidian Fu
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Liang Ma
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yizheng Yao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University; Clinical Research Center of Neurological Disease,
The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Zhi Yang
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People’s Hospital,
Shanghai JiaoTong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
7
|
Tian X, Zhao H, Zhou J. Organization, functions, and mechanisms of the BBSome in development, ciliopathies, and beyond. eLife 2023; 12:e87623. [PMID: 37466224 DOI: 10.7554/elife.87623] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
The BBSome is an octameric protein complex that regulates ciliary transport and signaling. Mutations in BBSome subunits are closely associated with ciliary defects and lead to ciliopathies, notably Bardet-Biedl syndrome. Over the past few years, there has been significant progress in elucidating the molecular organization and functions of the BBSome complex. An improved understanding of BBSome-mediated biological events and molecular mechanisms is expected to help advance the development of diagnostic and therapeutic approaches for BBSome-related diseases. Here, we review the current literature on the structural assembly, transport regulation, and molecular functions of the BBSome, emphasizing its roles in cilium-related processes. We also provide perspectives on the pathological role of the BBSome in ciliopathies as well as how these can be exploited for therapeutic benefit.
Collapse
Affiliation(s)
- Xiaoyu Tian
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Huijie Zhao
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
8
|
Hsu Y, Bhattarai S, Thompson JM, Mahoney A, Thomas J, Mayer SK, Datta P, Garrison J, Searby CC, Vandenberghe LH, Seo S, Sheffield VC, Drack AV. Subretinal gene therapy delays vision loss in a Bardet-Biedl Syndrome type 10 mouse model. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:164-181. [PMID: 36700052 PMCID: PMC9841241 DOI: 10.1016/j.omtn.2022.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Blindness in Bardet-Biedl syndrome (BBS) is caused by dysfunction and loss of photoreceptor cells in the retina. BBS10, mutations of which account for approximately 21% of all BBS cases, encodes a chaperonin protein indispensable for the assembly of the BBSome, a cargo adaptor important for ciliary trafficking. The loss of BBSome function in the eye causes a reduced light sensitivity of photoreceptor cells, photoreceptor ciliary malformation, dysfunctional ciliary trafficking, and photoreceptor cell death. Cone photoreceptors lacking BBS10 have congenitally low electrical function in electroretinography. In this study, we performed gene augmentation therapy by injecting a viral construct subretinally to deliver the coding sequence of the mouse Bbs10 gene to treat retinal degeneration in a BBS10 mouse model. Long-term efficacy was assessed by measuring the electrical functions of the retina over time, imaging of the treated regions to visualize cell survival, conducting visually guided swim assays to measure functional vision, and performing retinal histology. We show that subretinal gene therapy slowed photoreceptor cell death and preserved retinal function in treated eyes. Notably, cone photoreceptors regained their electrical function after gene augmentation. Measurement of functional vision showed that subretinal gene therapy provided a significant benefit in delaying vision loss.
Collapse
Affiliation(s)
- Ying Hsu
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Sajag Bhattarai
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Jacob M. Thompson
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Angela Mahoney
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Jacintha Thomas
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Sara K. Mayer
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, USA
| | - Poppy Datta
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Janelle Garrison
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | | | - Luk H. Vandenberghe
- Massachusetts Eye and Ear, Grousbeck Gene Therapy Center, Harvard Medical School, Boston, MA, USA
| | - Seongjin Seo
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Val C. Sheffield
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Arlene V. Drack
- Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
9
|
Mohan K, Dubey SK, Jung K, Dubey R, Wang QJ, Prajapati S, Roney J, Abney J, Kleinman ME. Long-Term Evaluation of Retinal Morphology and Function in Rosa26-Cas9 Knock-In Mice. Int J Mol Sci 2023; 24:5186. [PMID: 36982266 PMCID: PMC10049241 DOI: 10.3390/ijms24065186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/25/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
The CRISPR/Cas9 system is a robust, efficient, and cost-effective gene editing tool widely adopted in translational studies of ocular diseases. However, in vivo CRISPR-based editing in animal models poses challenges such as the efficient delivery of the CRISPR components in viral vectors with limited packaging capacity and a Cas9-associated immune response. Using a germline Cas9-expressing mouse model would help to overcome these limitations. Here, we evaluated the long-term effects of SpCas9 expression on retinal morphology and function using Rosa26-Cas9 knock-in mice. We observed abundant SpCas9 expression in the RPE and retina of Rosa26-Cas9 mice using the real-time polymerase chain reaction (RT-PCR), Western blotting, and immunostaining. SD-OCT imaging and histological analysis of the RPE, retinal layers, and vasculature showed no apparent structural abnormalities in adult and aged Cas9 mice. Full-field electroretinogram of adult and aged Cas9 mice showed no long-term functional changes in the retinal tissues because of constitutive Cas9 expression. The current study showed that both the retina and RPE maintain their phenotypic and functional features in Cas9 knock-in mice, establishing this as an ideal animal model for developing therapeutics for retinal diseases.
Collapse
Affiliation(s)
- Kabhilan Mohan
- Department of Surgery, East Tennessee State University, Johnson City, TN 37614, USA (S.K.D.)
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Sushil Kumar Dubey
- Department of Surgery, East Tennessee State University, Johnson City, TN 37614, USA (S.K.D.)
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Kyungsik Jung
- Department of Surgery, East Tennessee State University, Johnson City, TN 37614, USA (S.K.D.)
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Rashmi Dubey
- Department of Surgery, East Tennessee State University, Johnson City, TN 37614, USA (S.K.D.)
| | - Qing Jun Wang
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Subhash Prajapati
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Jacob Roney
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Jennifer Abney
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Mark Ellsworth Kleinman
- Department of Surgery, East Tennessee State University, Johnson City, TN 37614, USA (S.K.D.)
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
10
|
Melluso A, Secondulfo F, Capolongo G, Capasso G, Zacchia M. Bardet-Biedl Syndrome: Current Perspectives and Clinical Outlook. Ther Clin Risk Manag 2023; 19:115-132. [PMID: 36741589 PMCID: PMC9896974 DOI: 10.2147/tcrm.s338653] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
The Bardet Biedl syndrome (BBS) is a rare inherited disorder considered a model of non-motile ciliopathy. It is in fact caused by mutations of genes encoding for proteins mainly localized to the base of the cilium. Clinical features of BBS patients are widely shared with patients suffering from other ciliopathies, especially autosomal recessive syndromic disorders; moreover, mutations in cilia-related genes can cause different clinical ciliopathy entities. Besides the best-known clinical features, as retinal degeneration, learning disabilities, polydactyly, obesity and renal defects, several additional clinical signs have been reported in BBS, expanding our understanding of the complexity of its clinical spectrum. The present review aims to describe the current knowledge of BBS i) pathophysiology, ii) clinical manifestations, highlighting both the most common and the less described features, iii) current and future perspective for treatment.
Collapse
Affiliation(s)
- Andrea Melluso
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Floriana Secondulfo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giovanna Capolongo
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giovambattista Capasso
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy,Biogem Scarl, Ariano Irpino, AV, 83031, Italy
| | - Miriam Zacchia
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy,Correspondence: Miriam Zacchia, Via Pansini 5, Naples, 80131, Italy, Tel +39 081 566 6650, Fax +39 081 566 6671, Email
| |
Collapse
|
11
|
Delvallée C, Dollfus H. Retinal Degeneration Animal Models in Bardet-Biedl Syndrome and Related Ciliopathies. Cold Spring Harb Perspect Med 2023; 13:a041303. [PMID: 36596648 PMCID: PMC9808547 DOI: 10.1101/cshperspect.a041303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Retinal degeneration due to photoreceptor ciliary-related proteins dysfunction accounts for more than 25% of all inherited retinal dystrophies. The cilium, being an evolutionarily conserved and ubiquitous organelle implied in many cellular functions, can be investigated by way of many models from invertebrate models to nonhuman primates, all these models have massively contributed to the pathogenesis understanding of human ciliopathies. Taking the Bardet-Biedl syndrome (BBS) as an emblematic example as well as other related syndromic ciliopathies, the contribution of a wide range of models has enabled to characterize the role of the BBS proteins in the archetypical cilium but also at the level of the connecting cilium of the photoreceptors. There are more than 24 BBS genes encoding for proteins that form different complexes such as the BBSome and the chaperone proteins complex. But how they lead to retinal degeneration remains a matter of debate with the possible accumulation of proteins in the inner segment and/or accumulation of unwanted proteins in the outer segment that cannot return in the inner segment machinery. Many BBS proteins (but not the chaperonins for instance) can be modeled in primitive organisms such as Paramecium, Chlamydomonas reinardtii, Trypanosoma brucei, and Caenorhabditis elegans These models have enabled clarifying the role of a subset of BBS proteins in the primary cilium as well as their relations with other modules such as the intraflagellar transport (IFT) module, the nephronophthisis (NPHP) module, or the Meckel-Gruber syndrome (MKS)/Joubert syndrome (JBTS) module mostly involved with the transition zone of the primary cilia. Assessing the role of the primary cilia structure of the connecting cilium of the photoreceptor cells has been very much studied by way of zebrafish modeling (Danio rerio) as well as by a plethora of mouse models. More recently, large animal models have been described for three BBS genes and one nonhuman primate model in rhesus macaque for BBS7 In completion to animal models, human cell models can now be used notably thanks to gene editing and the use of induced pluripotent stem cells (iPSCs). All these models are not only important for pathogenesis understanding but also very useful for studying therapeutic avenues, their pros and cons, especially for gene replacement therapy as well as pharmacological triggers.
Collapse
Affiliation(s)
- Clarisse Delvallée
- Laboratoire de Génétique Médicale UMRS1112, Centre de Recherche Biomédicale de Strasbourg, CRBS, Institut de Génétique Médicale d'Alsace, IGMA, Strasbourg 67000, France
| | - Hélène Dollfus
- Laboratoire de Génétique Médicale UMRS1112, Centre de Recherche Biomédicale de Strasbourg, CRBS, Institut de Génétique Médicale d'Alsace, IGMA, Strasbourg 67000, France
| |
Collapse
|
12
|
Guo DF, Merrill RA, Qian L, Hsu Y, Zhang Q, Lin Z, Thedens DR, Usachev YM, Grumbach I, Sheffield VC, Strack S, Rahmouni K. The BBSome regulates mitochondria dynamics and function. Mol Metab 2023; 67:101654. [PMID: 36513220 PMCID: PMC9792363 DOI: 10.1016/j.molmet.2022.101654] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE The essential role of mitochondria in regulation of metabolic function and other physiological processes has garnered enormous interest in understanding the mechanisms controlling the function of this organelle. We assessed the role of the BBSome, a protein complex composed of eight Bardet-Biedl syndrome (BBS) proteins, in the control of mitochondria dynamic and function. METHODS We used a multidisciplinary approach that include CRISPR/Cas9 technology-mediated generation of a stable Bbs1 gene knockout hypothalamic N39 neuronal cell line. We also analyzed the phenotype of BBSome deficient mice in presence or absence of the gene encoding A-kinase anchoring protein 1 (AKAP1). RESULTS Our data show that the BBSome play an important role in the regulation of mitochondria dynamics and function. Disruption of the BBSome cause mitochondria hyperfusion in cell lines, fibroblasts derived from patients as well as in hypothalamic neurons and brown adipocytes of mice. The morphological changes in mitochondria translate into functional abnormalities as indicated by the reduced oxygen consumption rate and altered mitochondrial distribution and calcium handling. Mechanistically, we demonstrate that the BBSome modulates the activity of dynamin-like protein 1 (DRP1), a key regulator of mitochondrial fission, by regulating its phosphorylation and translocation to the mitochondria. Notably, rescuing the decrease in DRP1 activity through deletion of one copy of the gene encoding AKAP1 was effective to normalize the defects in mitochondrial morphology and activity induced by BBSome deficiency. Importantly, this was associated with improvement in several of the phenotypes caused by loss of the BBSome such as the neuroanatomical abnormalities, metabolic alterations and obesity highlighting the importance of mitochondria defects in the pathophysiology of BBS. CONCLUSIONS These findings demonstrate a critical role of the BBSome in the modulation of mitochondria function and point to mitochondrial defects as a key disease mechanism in BBS.
Collapse
Affiliation(s)
- Deng-Fu Guo
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Veterans Affairs Health Care System, Iowa City, IA, USA
| | - Ronald A Merrill
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Lan Qian
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Ying Hsu
- Veterans Affairs Health Care System, Iowa City, IA, USA
| | - Qihong Zhang
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Zhihong Lin
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Daniel R Thedens
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Yuriy M Usachev
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Isabella Grumbach
- Veterans Affairs Health Care System, Iowa City, IA, USA; Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Val C Sheffield
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Stefan Strack
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Veterans Affairs Health Care System, Iowa City, IA, USA; Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
13
|
Scruggs BA, Bhattarai S, Helms M, Cherascu I, Salesevic A, Stalter E, Laird J, Baker SA, Drack AV. AAV2/4-RS1 gene therapy in the retinoschisin knockout mouse model of X-linked retinoschisis. PLoS One 2022; 17:e0276298. [PMID: 36477475 PMCID: PMC9728878 DOI: 10.1371/journal.pone.0276298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 10/04/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To evaluate efficacy of a novel adeno-associated virus (AAV) vector, AAV2/4-RS1, for retinal rescue in the retinoschisin knockout (Rs1-KO) mouse model of X-linked retinoschisis (XLRS). Brinzolamide (Azopt®), a carbonic anhydrase inhibitor, was tested for its ability to potentiate the effects of AAV2/4-RS1. METHODS AAV2/4-RS1 with a cytomegalovirus (CMV) promoter (2x1012 viral genomes/mL) was delivered to Rs1-KO mice via intravitreal (N = 5; 1μL) or subretinal (N = 21; 2μL) injections at postnatal day 60-90. Eleven mice treated with subretinal therapy also received topical Azopt® twice a day. Serial full field electroretinography (ERG) was performed starting at day 50-60 post-injection. Mice were evaluated using a visually guided swim assay (VGSA) in light and dark conditions. The experimental groups were compared to untreated Rs1-KO (N = 11), wild-type (N = 12), and Rs1-KO mice receiving only Azopt® (N = 5). Immunofluorescence staining was performed to assess RS1 protein expression following treatment. RESULTS The ERG b/a ratio was significantly higher in the subretinal plus Azopt® (p<0.0001), subretinal without Azopt® (p = 0.0002), and intravitreal (p = 0.01) treated eyes compared to untreated eyes. There was a highly significant subretinal treatment effect on ERG amplitudes collectively at 7-9 months post-injection (p = 0.0003). Cones showed more effect than rods. The subretinal group showed improved time to platform in the dark VGSA compared to untreated mice (p<0.0001). RS1 protein expression was detected in the outer retina in subretinal treated mice and in the inner retina in intravitreal treated mice. CONCLUSIONS AAV2/4-RS1 shows promise for improving retinal phenotype in the Rs1-KO mouse model. Subretinal delivery was superior to intravitreal. Topical brinzolamide did not improve efficacy. AAV2/4-RS1 may be considered as a potential treatment for XLRS patients.
Collapse
Affiliation(s)
- Brittni A. Scruggs
- University of Iowa Institute for Vision Research and the Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States of America
| | - Sajag Bhattarai
- University of Iowa Institute for Vision Research and the Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States of America
| | - Megan Helms
- University of Iowa Institute for Vision Research and the Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States of America
| | - Ioana Cherascu
- University of Iowa Institute for Vision Research and the Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States of America
| | - Adisa Salesevic
- University of Iowa Institute for Vision Research and the Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States of America
| | - Elliot Stalter
- University of Iowa Institute for Vision Research and the Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States of America
- Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Joseph Laird
- Department of Biochemistry, University of Iowa, Iowa City, Iowa, United States of America
| | - Sheila A. Baker
- University of Iowa Institute for Vision Research and the Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States of America
- Department of Biochemistry, University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, United States of America
| | - Arlene V. Drack
- University of Iowa Institute for Vision Research and the Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa, United States of America
- Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
14
|
Karamali F, Behtaj S, Babaei-Abraki S, Hadady H, Atefi A, Savoj S, Soroushzadeh S, Najafian S, Nasr Esfahani MH, Klassen H. Potential therapeutic strategies for photoreceptor degeneration: the path to restore vision. J Transl Med 2022; 20:572. [PMID: 36476500 PMCID: PMC9727916 DOI: 10.1186/s12967-022-03738-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/29/2022] [Indexed: 12/12/2022] Open
Abstract
Photoreceptors (PRs), as the most abundant and light-sensing cells of the neuroretina, are responsible for converting light into electrical signals that can be interpreted by the brain. PR degeneration, including morphological and functional impairment of these cells, causes significant diminution of the retina's ability to detect light, with consequent loss of vision. Recent findings in ocular regenerative medicine have opened promising avenues to apply neuroprotective therapy, gene therapy, cell replacement therapy, and visual prostheses to the challenge of restoring vision. However, successful visual restoration in the clinical setting requires application of these therapeutic approaches at the appropriate stage of the retinal degeneration. In this review, firstly, we discuss the mechanisms of PR degeneration by focusing on the molecular mechanisms underlying cell death. Subsequently, innovations, recent developments, and promising treatments based on the stage of disorder progression are further explored. Then, the challenges to be addressed before implementation of these therapies in clinical practice are considered. Finally, potential solutions to overcome the current limitations of this growing research area are suggested. Overall, the majority of current treatment modalities are still at an early stage of development and require extensive additional studies, both pre-clinical and clinical, before full restoration of visual function in PR degeneration diseases can be realized.
Collapse
Affiliation(s)
- Fereshteh Karamali
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sanaz Behtaj
- grid.1022.10000 0004 0437 5432Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Queensland, Australia ,grid.1022.10000 0004 0437 5432Menzies Health Institute Queensland, Griffith University, Southport, QLD 4222 Australia
| | - Shahnaz Babaei-Abraki
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Hanieh Hadady
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Atefeh Atefi
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Soraya Savoj
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Sareh Soroushzadeh
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Samaneh Najafian
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr Esfahani
- grid.417689.5Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Henry Klassen
- grid.266093.80000 0001 0668 7243Gavin Herbert Eye Institute, Irvine, CA USA
| |
Collapse
|
15
|
Chang KJ, Wu HY, Yarmishyn AA, Li CY, Hsiao YJ, Chi YC, Lo TC, Dai HJ, Yang YC, Liu DH, Hwang DK, Chen SJ, Hsu CC, Kao CL. Genetics behind Cerebral Disease with Ocular Comorbidity: Finding Parallels between the Brain and Eye Molecular Pathology. Int J Mol Sci 2022; 23:9707. [PMID: 36077104 PMCID: PMC9456058 DOI: 10.3390/ijms23179707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022] Open
Abstract
Cerebral visual impairments (CVIs) is an umbrella term that categorizes miscellaneous visual defects with parallel genetic brain disorders. While the manifestations of CVIs are diverse and ambiguous, molecular diagnostics stand out as a powerful approach for understanding pathomechanisms in CVIs. Nevertheless, the characterization of CVI disease cohorts has been fragmented and lacks integration. By revisiting the genome-wide and phenome-wide association studies (GWAS and PheWAS), we clustered a handful of renowned CVIs into five ontology groups, namely ciliopathies (Joubert syndrome, Bardet-Biedl syndrome, Alstrom syndrome), demyelination diseases (multiple sclerosis, Alexander disease, Pelizaeus-Merzbacher disease), transcriptional deregulation diseases (Mowat-Wilson disease, Pitt-Hopkins disease, Rett syndrome, Cockayne syndrome, X-linked alpha-thalassaemia mental retardation), compromised peroxisome disorders (Zellweger spectrum disorder, Refsum disease), and channelopathies (neuromyelitis optica spectrum disorder), and reviewed several mutation hotspots currently found to be associated with the CVIs. Moreover, we discussed the common manifestations in the brain and the eye, and collated animal study findings to discuss plausible gene editing strategies for future CVI correction.
Collapse
Affiliation(s)
- Kao-Jung Chang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hsin-Yu Wu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | | | - Cheng-Yi Li
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yu-Jer Hsiao
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yi-Chun Chi
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Tzu-Chen Lo
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - He-Jhen Dai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yi-Chiang Yang
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Ding-Hao Liu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - De-Kuang Hwang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Shih-Jen Chen
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Chih-Chien Hsu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chung-Lan Kao
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| |
Collapse
|
16
|
Nasser F, Kohl S, Kurtenbach A, Kempf M, Biskup S, Zuleger T, Haack TB, Weisschuh N, Stingl K, Zrenner E. Ophthalmic and Genetic Features of Bardet Biedl Syndrome in a German Cohort. Genes (Basel) 2022; 13:genes13071218. [PMID: 35886001 PMCID: PMC9322102 DOI: 10.3390/genes13071218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/29/2022] [Accepted: 07/02/2022] [Indexed: 12/04/2022] Open
Abstract
The aim of this study was to characterize the ophthalmic and genetic features of Bardet Biedl (BBS) syndrome in a cohort of patients from a German specialized ophthalmic care center. Sixty-one patients, aged 5−56 years, underwent a detailed ophthalmic examination including visual acuity and color vision testing, electroretinography (ERG), visually evoked potential recording (VEP), fundus examination, and spectral domain optical coherence tomography (SD-OCT). Adaptive optics flood illumination ophthalmoscopy was performed in five patients. All patients had received diagnostic genetic testing and were selected upon the presence of apparent biallelic variants in known BBS-associated genes. All patients had retinal dystrophy with morphologic changes of the retina. Visual acuity decreased from ~0.2 (decimal) at age 5 to blindness 0 at 50 years. Visual field examination could be performed in only half of the patients and showed a concentric constriction with remaining islands of function in the periphery. ERG recordings were mostly extinguished whereas VEP recordings were reduced in about half of the patients. The cohort of patients showed 51 different likely biallelic mutations—of which 11 are novel—in 12 different BBS-associated genes. The most common associated genes were BBS10 (32.8%) and BBS1 (24.6%), and by far the most commonly observed variants were BBS10 c.271dup;p.C91Lfs*5 (21 alleles) and BBS1 c.1169T>G;p.M390R (18 alleles). The phenotype associated with the different BBS-associated genes and genotypes in our cohort is heterogeneous, with diverse features without genotype−phenotype correlation. The results confirm and expand our knowledge of this rare disease.
Collapse
Affiliation(s)
- Fadi Nasser
- Centre for Ophthalmology, University of Tübingen, 72076 Tuebingen, Germany; (S.K.); (A.K.); (M.K.); (N.W.); (K.S.); (E.Z.)
- Department of Ophthalmology, University of Leipzig, 04103 Leipzig, Germany
- Correspondence:
| | - Susanne Kohl
- Centre for Ophthalmology, University of Tübingen, 72076 Tuebingen, Germany; (S.K.); (A.K.); (M.K.); (N.W.); (K.S.); (E.Z.)
| | - Anne Kurtenbach
- Centre for Ophthalmology, University of Tübingen, 72076 Tuebingen, Germany; (S.K.); (A.K.); (M.K.); (N.W.); (K.S.); (E.Z.)
| | - Melanie Kempf
- Centre for Ophthalmology, University of Tübingen, 72076 Tuebingen, Germany; (S.K.); (A.K.); (M.K.); (N.W.); (K.S.); (E.Z.)
- Center for Rare Eye Diseases, University of Tübingen, 72076 Tuebingen, Germany
| | | | - Theresia Zuleger
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tuebingen, Germany; (T.Z.); (T.B.H.)
| | - Tobias B. Haack
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076 Tuebingen, Germany; (T.Z.); (T.B.H.)
| | - Nicole Weisschuh
- Centre for Ophthalmology, University of Tübingen, 72076 Tuebingen, Germany; (S.K.); (A.K.); (M.K.); (N.W.); (K.S.); (E.Z.)
| | - Katarina Stingl
- Centre for Ophthalmology, University of Tübingen, 72076 Tuebingen, Germany; (S.K.); (A.K.); (M.K.); (N.W.); (K.S.); (E.Z.)
- Center for Rare Eye Diseases, University of Tübingen, 72076 Tuebingen, Germany
| | - Eberhart Zrenner
- Centre for Ophthalmology, University of Tübingen, 72076 Tuebingen, Germany; (S.K.); (A.K.); (M.K.); (N.W.); (K.S.); (E.Z.)
- Werner Reichardt Centre for Integrative Neuroscience (CIN), University of Tübingen, 72076 Tuebingen, Germany
| |
Collapse
|
17
|
Bansal M. Commentary: Retinitis pigmentosa in Laurence-Moon-Bardet-Biedl syndrome: Genomic sequencing, gene therapy, and gene editing. Indian J Ophthalmol 2022; 70:2539. [PMID: 35791151 PMCID: PMC9426160 DOI: 10.4103/ijo.ijo_398_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Mayank Bansal
- CSIR Insitute of Genomics and Integrative Biology (IGIB), New Delhi; Academy of Scientific and Innovative Research (AcSIR); Fortis Memorial Research Insitute, Gurgaon, Haryana, India
| |
Collapse
|
18
|
Ryan MM. Gene therapy for neuromuscular disorders: prospects and ethics. Arch Dis Child 2022; 107:421-426. [PMID: 34462265 DOI: 10.1136/archdischild-2020-320908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 08/11/2021] [Indexed: 11/04/2022]
Abstract
Most childhood neuromuscular disorders are caused by mutations causing abnormal expression or regulation of single genes or genetic pathways. The potential for gene therapy, gene editing and genetic therapies to ameliorate the course of these conditions is extraordinarily exciting, but there are significant challenges associated with their use, particularly with respect to safety, efficacy, cost and equity. Engagement with these novel technologies mandates careful assessment of the benefits and burdens of treatment for the patient, their family and their society. The examples provided by spinal muscular atrophy and Duchenne muscular dystrophy illustrate the potential value and challenges of gene and genetic therapies for paediatric neurological conditions. The cost and complexity of administration of these agents is a challenge for all countries. Jurisdictional variations in availability of newborn screening, genetic diagnostics, drug approval and reimbursement pathways, treatment and rehabilitation will affect equity of access, nationally and internationally. These challenges will best be addressed by collaboration by governments, pharma, clinicians and patient groups to establish frameworks for safe and cost-effective use of these exciting new therapies.
Collapse
Affiliation(s)
- Monique M Ryan
- Children's Neurosciences Centre, The Royal Children's Hospital Melbourne, Parkville, Victoria, Australia .,Murdoch Children's Research Institute, Parkville, Victoria, Australia
| |
Collapse
|
19
|
Husson H, Bukanov NO, Moreno S, Smith MM, Richards B, Zhu C, Picariello T, Park H, Wang B, Natoli TA, Smith LA, Zanotti S, Russo RJ, Madden SL, Klinger KW, Modur V, Ibraghimov-Beskrovnaya O. Correction of cilia structure and function alleviates multi-organ pathology in Bardet-Biedl syndrome mice. Hum Mol Genet 2021; 29:2508-2522. [PMID: 32620959 PMCID: PMC7471507 DOI: 10.1093/hmg/ddaa138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/22/2020] [Accepted: 07/01/2020] [Indexed: 12/21/2022] Open
Abstract
Bardet–Biedl syndrome (BBS) is a pleiotropic autosomal recessive ciliopathy affecting multiple organs. The development of potential disease-modifying therapy for BBS will require concurrent targeting of multi-systemic manifestations. Here, we show for the first time that monosialodihexosylganglioside accumulates in Bbs2−/− cilia, indicating impairment of glycosphingolipid (GSL) metabolism in BBS. Consequently, we tested whether BBS pathology in Bbs2−/− mice can be reversed by targeting the underlying ciliary defect via reduction of GSL metabolism. Inhibition of GSL synthesis with the glucosylceramide synthase inhibitor Genz-667161 decreases the obesity, liver disease, retinal degeneration and olfaction defect in Bbs2−/− mice. These effects are secondary to preservation of ciliary structure and signaling, and stimulation of cellular differentiation. In conclusion, reduction of GSL metabolism resolves the multi-organ pathology of Bbs2−/− mice by directly preserving ciliary structure and function towards a normal phenotype. Since this approach does not rely on the correction of the underlying genetic mutation, it might translate successfully as a treatment for other ciliopathies.
Collapse
Affiliation(s)
- Hervé Husson
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Nikolay O Bukanov
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Sarah Moreno
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Mandy M Smith
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | | | - Cheng Zhu
- Translational Sciences, Sanofi, Framingham, MA 01701, USA
| | - Tyler Picariello
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Hyejung Park
- Pre-Development Sciences, Sanofi, Waltham, MA 02451, USA
| | - Bing Wang
- Pre-Development Sciences, Sanofi, Waltham, MA 02451, USA
| | - Thomas A Natoli
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Laurie A Smith
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Stefano Zanotti
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | - Ryan J Russo
- Rare and Neurologic Diseases Research, Sanofi, Framingham, MA 01701, USA
| | | | | | - Vijay Modur
- Rare Diseases Development, Sanofi, Cambridge, MA 02142, USA
| | | |
Collapse
|
20
|
Xie C, Habif JC, Uytingco CR, Ukhanov K, Zhang L, de Celis C, Sheffield VC, Martens JR. Gene therapy rescues olfactory perception in a clinically relevant ciliopathy model of Bardet-Biedl syndrome. FASEB J 2021; 35:e21766. [PMID: 34383976 DOI: 10.1096/fj.202100627r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/02/2021] [Accepted: 06/14/2021] [Indexed: 11/11/2022]
Abstract
Bardet-Biedl syndrome (BBS) is a hereditary genetic disorder that results in numerous clinical manifestations including olfactory dysfunction. Of at least 21 BBS-related genes that can carry multiple mutations, a pathogenic mutation, BBS1M390R, is the single most common mutation of clinically diagnosed BBS outcomes. While the deletion of BBS-related genes in mice can cause variable penetrance in different organ systems, the impact of the Bbs1M390R mutation in the olfactory system remains unclear. Using a clinically relevant knock-in mouse model homozygous for Bbs1M390R, we investigated the impact of the mutation on the olfactory system and tested the potential of viral-mediated, wildtype gene replacement therapy to rescue smell loss. The cilia of olfactory sensory neurons (OSNs) in Bbs1M390R/M390R mice were significantly shorter and fewer than those of wild-type mice. Also, both peripheral cellular odor detection and synaptic-dependent activity in the olfactory bulb were significantly decreased in the mutant mice. Furthermore, to gain insight into the degree to which perceptual features are impaired in the mutant mice, we used whole-body plethysmography to quantitatively measure odor-evoked sniffing. The Bbs1M390R/M390R mice showed significantly higher odor detection thresholds (reduced odor sensitivity) compared to wild-type mice; however, their odor discrimination acuity was still well maintained. Importantly, adenoviral expression of Bbs1 in OSNs restored cilia length and re-established both peripheral odorant detection and odor perception. Together, our findings further expand our understanding for the development of gene therapeutic treatment for congenital ciliopathies in the olfactory system.
Collapse
Affiliation(s)
- Chao Xie
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA.,Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL, USA
| | - Julien C Habif
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA.,Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL, USA
| | - Cedric R Uytingco
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA.,Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL, USA
| | - Kirill Ukhanov
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA.,Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL, USA
| | - Lian Zhang
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA.,Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL, USA
| | - Carlos de Celis
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA.,Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL, USA
| | - Val C Sheffield
- Division of Medical Genetics and Genomics, Department of Pediatrics, University of Iowa, Iowa City, IA, USA.,Department of Ophthalmology and Vision Research, University of Iowa, Iowa City, IA, USA
| | - Jeffrey R Martens
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA.,Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
21
|
Endothelial BBSome is essential for vascular, metabolic, and retinal functions. Mol Metab 2021; 53:101308. [PMID: 34303879 PMCID: PMC8379702 DOI: 10.1016/j.molmet.2021.101308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/29/2021] [Accepted: 07/19/2021] [Indexed: 11/21/2022] Open
Abstract
Objectives Endothelial cells that line the entire vascular system play a pivotal role in the control of various physiological processes, including metabolism. Additionally, endothelial dysfunction is associated with many pathological conditions, including obesity. Here, we assessed the role of the BBSome, a protein complex composed of eight Bardet-Biedl syndrome (BBS) proteins in endothelial cells. Methods We studied the effects of BBSome disruption in endothelial cells on vascular function, body weight, glucose homeostasis, and the liver and retina. For this, we generated mice with selective BBSome disruption in endothelial cells through Bbs1 gene deletion. Results We found that endothelial cell–specific BBSome disruption causes endothelial dysfunction, as indicated by the impaired acetylcholine-induced vasorelaxation in both the aorta and mesenteric artery. This was associated with an increase in the contractile response to thromboxane A2 receptor agonist (U46619) in the mesenteric artery. Mechanistically, we demonstrated that mice lacking the Bbs1 gene in endothelial cells show elevated vascular angiotensinogen gene expression, implicating renin-angiotensin system activation in the vascular changes evoked by endothelial BBSome deficiency. Strikingly, our data indicate that endothelial BBSome deficiency increases body weight and fat mass and causes hepatosteatosis along with alterations in hepatic expression of lipid metabolism–related genes and metabolomics profile. In addition, electroretinogram and optical coherence tomography analyses revealed functional and structural abnormalities in the retina, evoked by absence of the endothelial BBSome. Conclusions Our findings demonstrate that the BBSome in endothelial cells is required for the regulation of vascular function, adiposity, hepatic lipid metabolism, and retinal function. Disruption of the BBSome in endothelial cells alters vascular reactivity. Loss of the BBSome in endothelial cells increases vascular angiotensinogen gene expression. Endothelial BBSome deficiency increases body weight and fat mass and causes hepatosteatosis. Absence of the endothelial BBSome induces functional and structural abnormalities in the retina.
Collapse
|
22
|
Aleman TS, O'Neil EC, O'Connor K, Jiang YY, Aleman IA, Bennett J, Morgan JIW, Toussaint BW. Bardet-Biedl syndrome-7 ( BBS7) shows treatment potential and a cone-rod dystrophy phenotype that recapitulates the non-human primate model. Ophthalmic Genet 2021; 42:252-265. [PMID: 33729075 DOI: 10.1080/13816810.2021.1888132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Purpose: To provide a detailed ophthalmic phenotype of two male patients with Bardet-Biedl Syndrome (BBS) due to mutations in the BBS7 geneMethods: Two brothers ages 26 (Patient 1, P1) and 23 (P2) underwent comprehensive ophthalmic evaluations over three years. Visual function was assessed with full-field electroretinograms (ffERGs), kinetic and chromatic perimetry, multimodal imaging with spectral domain optical coherence tomography (SD-OCT), fundus autofluorescence (FAF) with short- (SW) and near-infrared (NIR) excitation lights and adaptive optics scanning light ophthalmoscopy (AOSLO).Results: Both siblings had a history of obesity and postaxial polydactyly; P2 had diagnoses of type 1 Diabetes Mellitus, Addison's disease, high-functioning autism-spectrum disorder and -12D myopia. Visual acuities were better than 20/30. Kinetic fields were moderately constricted. Cone-mediated ffERGs were undetectable, rod ERGs were ~80% of normal mean. Static perimetry showed severe central cone and rod dysfunction. Foveal to parafoveal hypoautofluorescence, most obvious on NIR-FAF, co-localized with outer segment shortening/loss and outer nuclear layer thinning by SD-OCT, and with reduced photoreceptors densities by AOSLO. A structural-functional dissociation was confirmed for cone- and rod-mediated parameters. Worsening of the above abnormalities was documented by SD-OCT and FAF in P2 at 3 years. Gene screening identified compound heterozygous mutations in BBS7 (p.Val266Glu: c.797 T > A of maternal origin; c.1781_1783delCAT, paternal) in both patients.Conclusions: BBS7-associated retinal degeneration may present as a progressive cone-rod dystrophy pattern, reminiscent of both the murine and non-human primate models of the disease. Predominantly central retinal abnormalities in both cone and rod photoreceptors showed a structural-functional dissociation, an ideal scenario for gene augmentation treatments.
Collapse
Affiliation(s)
- Tomas S Aleman
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Erin C O'Neil
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Ophthalmology of the Children's Hospital of Philadelphia, Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Keli O'Connor
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yu You Jiang
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Isabella A Aleman
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jean Bennett
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jessica I W Morgan
- Scheie Eye Institute at the Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Advanced Retinal and Ocular Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brian W Toussaint
- Christiana Care Health System, Wilmington, Delaware, USA.,Department of Ophthalmology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
23
|
Ectopic expression of BBS1 rescues male infertility, but not retinal degeneration, in a BBS1 mouse model. Gene Ther 2021; 29:227-235. [PMID: 33664503 PMCID: PMC9422088 DOI: 10.1038/s41434-021-00241-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 01/12/2021] [Accepted: 02/05/2021] [Indexed: 11/30/2022]
Abstract
Bardet-Biedl syndrome (BBS) is a rare ciliopathy for which there are no current effective treatments. BBS is a genetically heterogeneous disease, though the M390R mutation in BBS1 is involved in approximately 25% of all genetic diagnoses of BBS. The principle features of BBS include retinal degeneration, obesity, male infertility, polydactyly, intellectual disability, and renal abnormalities. Patients with mutations in BBS genes often present with night blindness within the first decade of life, which progresses to complete blindness. This is due to progressive loss of photoreceptor cells. Male infertility is caused by a lack of spermatozoa flagella, rendering them immobile. In this study, we have crossed the wild-type human BBS1 gene, driven by the CAG promoter, onto the Bbs1M390R/M390R mouse model to determine if ectopic expression of BBS1 rescues male infertility and retinal degeneration. qRT-PCR indicates that the BBS1 transgene is expressed in multiple tissues throughout the mouse, with the highest expression seen in the testes, and much lower expression in the eye and hypothalamus. Immunohistochemistry of the transgene in the eye showed little if any expression in the photoreceptor outer nuclear layer. When male Bbs1M30R/M390R;BBS1TG+ mice are housed with WT females, they are able to sire offspring, indicating that the male infertility phenotype of BBS is rescued by the transgene. Using electroretinography (ERGs) to measure retinal function and optical coherence tomography to measure retinal thickness, we show that the transgene does not confer protection against retinal degeneration in Bbs1M300R/M390R;BBS1TG+ mice. The results of this study indicate the male infertility aspect of BBS is an attractive target for gene therapy.
Collapse
|
24
|
Ravi B, Chan-Cortés MH, Sumner CJ. Gene-Targeting Therapeutics for Neurological Disease: Lessons Learned from Spinal Muscular Atrophy. Annu Rev Med 2021; 72:1-14. [PMID: 33502897 DOI: 10.1146/annurev-med-070119-115459] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The last few decades have seen an explosion in identification of genes that cause monogenetic neurological diseases, as well as advances in gene-targeting therapeutics. Neurological conditions that were once considered incurable are now increasingly tractable. At the forefront is the motor neuron disease spinal muscular atrophy (SMA), historically the leading inherited cause of infant mortality. In the last 5 years, three SMA treatments have been approved by the US Food and Drug Administration (FDA): intrathecally delivered splice-switching antisense oligonucleotide (nusinersen), systemically delivered AAV9-based gene replacement therapy (onasemnogene abeparvovec), and an orally bioavailable, small-molecule, splice-switching drug (risdiplam). Despite this remarkable progress, clinical outcomes in patients are variable. Therapeutic optimization will require improved understanding of drug pharmacokinetics and target engagement in neurons, potential toxicities, and long-term effects. We review current progress in SMA therapeutics, clinical trials, shortcomings of current treatments, and implications for the treatment of other neurogenetic diseases.
Collapse
Affiliation(s)
- Bhavya Ravi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA;
| | | | - Charlotte J Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA; .,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21202, USA
| |
Collapse
|
25
|
Kantor A, McClements ME, Peddle CF, Fry LE, Salman A, Cehajic-Kapetanovic J, Xue K, MacLaren RE. CRISPR genome engineering for retinal diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 182:29-79. [PMID: 34175046 DOI: 10.1016/bs.pmbts.2021.01.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Novel gene therapy treatments for inherited retinal diseases have been at the forefront of translational medicine over the past couple of decades. Since the discovery of CRISPR mechanisms and their potential application for the treatment of inherited human conditions, it seemed inevitable that advances would soon be made using retinal models of disease. The development of CRISPR technology for gene therapy and its increasing potential to selectively target disease-causing nucleotide changes has been rapid. In this chapter, we discuss the currently available CRISPR toolkit and how it has been and can be applied in the future for the treatment of inherited retinal diseases. These blinding conditions have until now had limited opportunity for successful therapeutic intervention, but the discovery of CRISPR has created new hope of achieving such, as we discuss within this chapter.
Collapse
Affiliation(s)
- Ariel Kantor
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom.
| | - Michelle E McClements
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Caroline F Peddle
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Lewis E Fry
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Ahmed Salman
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Jasmina Cehajic-Kapetanovic
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom; Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
26
|
Cring MR, Sheffield VC. Gene therapy and gene correction: targets, progress, and challenges for treating human diseases. Gene Ther 2020; 29:3-12. [PMID: 33037407 DOI: 10.1038/s41434-020-00197-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/24/2020] [Accepted: 09/21/2020] [Indexed: 12/28/2022]
Abstract
The field of gene therapy has made significant strides over the last several decades toward the treatment of previously untreatable genetic disease. Gene therapy techniques have been aimed at mitigating disease features of recessive and dominant disorders, as well as several cancers and other diseases. While there have been numerous disease targets of gene therapy trials, only four therapies have reached FDA and/or EMA approval for clinical use. Gene correction using CRISPR-Cas9 is an extension of gene therapy that has received considerable attention in recent years and boasts many possible uses beyond classical gene therapy approaches. While there is significant therapeutic potential using gene therapy and gene correction strategies, a number of hurdles remain to be overcome before they become more common in clinical use, particularly with regards to safety and efficacy. As research progresses in this exciting field, it is likely that these therapies will become first-line treatments and will have tremendous positive impacts on the lives of patients with genetic disorders.
Collapse
Affiliation(s)
- Matthew R Cring
- Department of Pediatrics, Division of Medical Genetics and Genomics, University of Iowa, Iowa City, IA, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA
| | - Val C Sheffield
- Department of Pediatrics, Division of Medical Genetics and Genomics, University of Iowa, Iowa City, IA, USA. .,Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
27
|
Ku CA, Pennesi ME. The new landscape of retinal gene therapy. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2020; 184:846-859. [PMID: 32888388 DOI: 10.1002/ajmg.c.31842] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 12/15/2022]
Abstract
Novel therapeutics for inherited retinal dystrophies (IRDs) have rapidly evolved since groundbreaking clinical trials for LCA due to RPE65 mutations led to the first FDA-approved in vivo gene therapy. Since then, advancements in viral vectors have led to more efficient AAV transduction and developed other viral vectors for gene augmentation therapy of large gene targets. Furthermore, significant developments in gene editing and RNA modulation technologies have introduced novel capabilities for treatment of autosomal dominant diseases, intronic mutations, and/or large genes otherwise unable to be treated with current viral vectors. We highlight strategies currently being evaluated in gene therapy clinical trials and promising preclinical developments for IRDs.
Collapse
Affiliation(s)
- Cristy A Ku
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Mark E Pennesi
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
28
|
Trapani I, Auricchio A. Has retinal gene therapy come of age? From bench to bedside and back to bench. Hum Mol Genet 2020; 28:R108-R118. [PMID: 31238338 PMCID: PMC6797000 DOI: 10.1093/hmg/ddz130] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 04/24/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023] Open
Abstract
Retinal gene therapy has advanced considerably in the past three decades. Initial efforts have been devoted to comprehensively explore and optimize the transduction abilities of gene delivery vectors, define the appropriate intraocular administration routes and obtain evidence of efficacy in animal models of inherited retinal diseases (IRDs). Successful translation in clinical trials of the initial promising proof-of-concept studies led to the important milestone of the first approved product for retinal gene therapy in both US and Europe. The unprecedented clinical development observed during the last decade in the field is however highlighting new challenges that will need to be overcome to bring gene therapy to fruition to a larger patient population within and beyond the realm of IRDs.
Collapse
Affiliation(s)
- Ivana Trapani
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.,Medical Genetics, Department of Translational Medicine, Federico II University, Naples, Italy
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.,Department of Advanced Biomedicine, Federico II University, Naples, Italy
| |
Collapse
|
29
|
Fry LE, Peddle CF, Barnard AR, McClements ME, MacLaren RE. RNA editing as a therapeutic approach for retinal gene therapy requiring long coding sequences. Int J Mol Sci 2020; 21:ijms21030777. [PMID: 31991730 PMCID: PMC7037314 DOI: 10.3390/ijms21030777] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/26/2022] Open
Abstract
RNA editing aims to treat genetic disease through altering gene expression at the transcript level. Pairing site-directed RNA-targeting mechanisms with engineered deaminase enzymes allows for the programmable correction of G>A and T>C mutations in RNA. This offers a promising therapeutic approach for a range of genetic diseases. For inherited retinal degenerations caused by point mutations in large genes not amenable to single-adeno-associated viral (AAV) gene therapy such as USH2A and ABCA4, correcting RNA offers an alternative to gene replacement. Genome editing of RNA rather than DNA may offer an improved safety profile, due to the transient and potentially reversible nature of edits made to RNA. This review considers the current site-directing RNA editing systems, and the potential to translate these to the clinic for the treatment of inherited retinal degeneration.
Collapse
Affiliation(s)
- Lewis E. Fry
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Correspondence:
| | - Caroline F. Peddle
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Alun R. Barnard
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Michelle E. McClements
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Robert E. MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| |
Collapse
|
30
|
Uytingco CR, Green WW, Martens JR. Olfactory Loss and Dysfunction in Ciliopathies: Molecular Mechanisms and Potential Therapies. Curr Med Chem 2019; 26:3103-3119. [PMID: 29303074 DOI: 10.2174/0929867325666180105102447] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/08/2017] [Accepted: 12/20/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ciliopathies are a class of inherited pleiotropic genetic disorders in which alterations in cilia assembly, maintenance, and/or function exhibit penetrance in the multiple organ systems. Olfactory dysfunction is one such clinical manifestation that has been shown in both patients and model organisms. Existing therapies for ciliopathies are limited to the treatment or management of symptoms. The last decade has seen an increase in potential curative therapeutic options including small molecules and biologics. Recent work in multiciliated olfactory sensory neurons has demonstrated the capacity of targeted gene therapy to restore ciliation in terminally differentiated cells and rescue olfactory function. This review will discuss the current understanding of the penetrance of ciliopathies in the olfactory system. Importantly, it will highlight both pharmacological and biological approaches, and their potential therapeutic value in the olfactory system and other ciliated tissues. METHODS We undertook a structured and comprehensive search of peer-reviewed research literature encompassing in vitro, in vivo, model organism, and clinical studies. From these publications, we describe the olfactory system, and discuss the penetrance of ciliopathies and impact of cilia loss on olfactory function. In addition, we outlined the developing therapies for ciliopathies across different organ and cell culture systems, and discussed their potential therapeutic application to the mammalian olfactory system. RESULTS One-hundred sixty-one manuscripts were included in the review, centering on the understanding of olfactory penetrance of ciliopathies, and discussing the potential therapeutic options for ciliopathies in the context of the mammalian olfactory system. Forty-four manuscripts were used to generate a table listing the known congenital causes of olfactory dysfunction, with the first ten listed are linked to ciliopathies. Twenty-three manuscripts were used to outline the potential of small molecules for the olfactory system. Emphasis was placed on HDAC6 inhibitors and lithium, both of which were shown to stabilize microtubule structures, contributing to ciliogenesis and cilia lengthening. Seventy-five manuscripts were used to describe gene therapy and gene therapeutic strategies. Included were the implementation of adenoviral, adeno-associated virus (AAV), and lentiviral vectors to treat ciliopathies across different organ systems and application toward the olfactory system. Thus far, adenoviral and AAVmeditated ciliary restoration demonstrated successful proof-of-principle preclinical studies. In addition, gene editing, ex vivo gene therapy, and transplantation could serve as alternative therapeutic and long-term approaches. But for all approaches, additional assessment of vector immunogenicity, specificity, and efficacy need further investigation. Currently, ciliopathy treatments are limited to symptomatic management with no curative options. However, the accessibility and amenability of the olfactory system to treatment would facilitate development and advancement of a viable therapy. CONCLUSION The findings of this review highlight the contribution of ciliopathies to a growing list of congenial olfactory dysfunctions. Promising results from other organ systems imply the feasibility of biologics, with results from gene therapies proving to be a viable therapeutic option for ciliopathies and olfactory dysfunction.
Collapse
Affiliation(s)
- Cedric R Uytingco
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, United States.,University of Florida Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Warren W Green
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, United States.,University of Florida Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Jeffrey R Martens
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, United States.,University of Florida Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, United States
| |
Collapse
|
31
|
Kretschmer V, Patnaik SR, Kretschmer F, Chawda MM, Hernandez-Hernandez V, May-Simera HL. Progressive Characterization of Visual Phenotype in Bardet-Biedl Syndrome Mutant Mice. Invest Ophthalmol Vis Sci 2019; 60:1132-1143. [PMID: 30901771 DOI: 10.1167/iovs.18-25210] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Bardet-Biedl syndrome (BBS) is an archetypical ciliopathy caused by defective ciliary trafficking and consequent function. Insights gained from BBS mouse models are applicable to other syndromic and nonsyndromic retinal diseases. This progressive characterization of the visual phenotype in three BBS mouse models sets a baseline for testing therapeutic interventions. Methods Longitudinal acquisition of electroretinograms, optical coherence tomography scans, and visual acuity using the optomotor reflex in Bbs6/Mkks, Bbs8/Ttc8, and Bbs5 knockout mice. Gene and protein expression analysis in vivo and in vitro. Results Complete loss of BBS5, BBS6, or BBS8 leads to different rates of retinal degeneration and visual function over time. BBS8-deficient mice showed the fastest rate of degeneration, and BBS8 seems to be required for cone photoreceptors to reach functional maturity. In contrast, the loss of BBS5 (a further BBSome component) showed very little degeneration. Loss of BBS8 versus BBS5 resulted in different physiologic responses both in vivo and in vitro. BBS6-deficient mice show a slower rate of degeneration with both rod and cone function reducing at a similar rate. Conclusions The mouse models analyzed show distinct and diverging courses of degeneration upon loss of BBS5, BBS6, or BBS8, which can be used as a benchmark to test therapeutic interventions. Close consideration of the different phenotypes reveal subtle but important differences relating to their function. Because we also see differences in terms of phenotype depending on the type of visual assessment used, our data highlight the importance of using a combinatorial approach for assessment of visual function.
Collapse
Affiliation(s)
- Viola Kretschmer
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes Gutenberg University, Mainz, Germany
| | - Sarita Rani Patnaik
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes Gutenberg University, Mainz, Germany
| | - Friedrich Kretschmer
- Scientific Computing Facility, Max Planck Institute for Brain Research, Frankfurt, Germany
| | - Mira Manilal Chawda
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Victor Hernandez-Hernandez
- Genetics and Genomic Medicine Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Helen Louise May-Simera
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
32
|
Barny I, Perrault I, Michel C, Goudin N, Defoort-Dhellemmes S, Ghazi I, Kaplan J, Rozet JM, Gerard X. AON-Mediated Exon Skipping to Bypass Protein Truncation in Retinal Dystrophies Due to the Recurrent CEP290 c.4723A > T Mutation. Fact or Fiction? Genes (Basel) 2019; 10:E368. [PMID: 31091803 PMCID: PMC6562928 DOI: 10.3390/genes10050368] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/09/2019] [Accepted: 05/09/2019] [Indexed: 01/09/2023] Open
Abstract
Mutations in CEP290 encoding a centrosomal protein important to cilia formation cause a spectrum of diseases, from isolated retinal dystrophies to multivisceral and sometimes embryo-lethal ciliopathies. In recent years, endogenous and/or selective non-canonical exon skipping of mutant exons have been documented in attenuated retinal disease cases. This observation led us to consider targeted exon skipping to bypass protein truncation resulting from a recurrent mutation in exon 36 (c.4723A > T, p.Lys1575*) causing isolated retinal ciliopathy. Here, we report two unrelated individuals (P1 and P2), carrying the mutation in homozygosity but affected with early-onset severe retinal dystrophy and congenital blindness, respectively. Studying skin-derived fibroblasts, we observed basal skipping and nonsense associated-altered splicing of exon 36, producing low (P1) and very low (P2) levels of CEP290 products. Consistent with a more severe disease, fibroblasts from P2 exhibited reduced ciliation compared to P1 cells displaying normally abundant cilia; both lines presented however significantly elongated cilia, suggesting altered axonemal trafficking. Antisense oligonucleotides (AONs)-mediated skipping of exon 36 increased the abundance of the premature termination codon (PTC)-free mRNA and protein, reduced axonemal length and improved cilia formation in P2 but not in P1 expressing higher levels of skipped mRNA, questioning AON-mediated exon skipping to treat patients carrying the recurrent c.4723A > T mutation.
Collapse
Affiliation(s)
- Iris Barny
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases, Imagine and Paris Descartes University, 75015 Paris, France.
| | - Isabelle Perrault
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases, Imagine and Paris Descartes University, 75015 Paris, France.
| | - Christel Michel
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases, Imagine and Paris Descartes University, 75015 Paris, France.
| | - Nicolas Goudin
- Cell Imaging Core Facility of the Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Imagine and Paris Descartes University, 75015 Paris, France.
| | - Sabine Defoort-Dhellemmes
- Service D'exploration de la Vision et Neuro-Ophtalmologie, Pôle D'imagerie et Explorations Fonctionnelles, CHRU de Lille, 59037 Lille, France.
| | - Imad Ghazi
- Department of Ophthalmology, IHU Necker-Enfants Malades, 75015 Paris, France.
| | - Josseline Kaplan
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases, Imagine and Paris Descartes University, 75015 Paris, France.
| | - Jean-Michel Rozet
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases, Imagine and Paris Descartes University, 75015 Paris, France.
| | - Xavier Gerard
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetics Diseases, Imagine and Paris Descartes University, 75015 Paris, France.
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, Hôpital Ophtalmique Jules Gonin, Fondation Asile des Aveugles, 1004 Lausanne, Switzerland.
| |
Collapse
|
33
|
Butler MC, Sullivan JM. Ultrahigh Resolution Mouse Optical Coherence Tomography to Aid Intraocular Injection in Retinal Gene Therapy Research. J Vis Exp 2018. [PMID: 30451216 DOI: 10.3791/55894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
HR-SD-OCT is utilized to monitor the progression of photoreceptor degeneration in live mouse models, assess the delivery of therapeutic agents into the subretinal space, and to evaluate toxicity and efficacy in vivo. HR-SD-OCT uses near infrared light (800-880 nm) and has optics specifically designed for the unique optics of the mouse eye with sub-2-micron axial resolution. Transgenic mouse models of outer retinal (photoreceptor) degeneration and controls were imaged to assess the disease progression. Pulled glass microneedles were used to deliver sub retinal injections of adeno-associated virus (AAV) or nanoparticles (NP) via a trans-scleral and trans-choroidal approach. Careful positioning of the needle into the subretinal space was required prior to a calibrated pressure injection, which delivers fluid into the sub retinal space. Real time subretinal surgery was conducted on our retinal imaging system (RIS). HR-SD-OCT demonstrated progressive uniform retinal degeneration due to expression of a toxic mutant human mutant rhodopsin (P347S) (RHOP347S) transgene in mice. HR-SD-OCT allows rigorous quantification of all the retinal layers. Outer nuclear layer (ONL) thickness and photoreceptor outer segment length (OSL) measurements correlate with photoreceptor vitality, degeneration, or rescue. The RIS delivery system allows real-time visualization of subretinal injections in neonatal (~P10-14) or adult mice, and HR-SD-OCT immediately determines success of delivery and maps areal extent. HR-SD-OCT is a powerful tool that can evaluate the success of subretinal surgery in mice, in addition to measuring vitality of photoreceptors in vivo. HR-SD-OCT can also be used to identify uniform animal cohorts to evaluate the extent of retinal degeneration, toxicity, and therapeutic rescue in preclinical gene therapy research studies.
Collapse
Affiliation(s)
- Mark C Butler
- Research Service, VA Western New York Healthcare System; Department of Ophthalmology, (Ross Eye Institute), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo- SUNY
| | - Jack M Sullivan
- Research Service, VA Western New York Healthcare System; Department of Ophthalmology, (Ross Eye Institute), Jacobs School of Medicine and Biomedical Sciences, University at Buffalo- SUNY; Pharmacology/Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo- SUNY; Physiology/Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo- SUNY; Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo- SUNY; The RNA Institute, University at Buffalo- SUNY; The SUNY Eye Institute;
| |
Collapse
|
34
|
Duncan JL, Pierce EA, Laster AM, Daiger SP, Birch DG, Ash JD, Iannaccone A, Flannery JG, Sahel JA, Zack DJ, Zarbin MA. Inherited Retinal Degenerations: Current Landscape and Knowledge Gaps. Transl Vis Sci Technol 2018; 7:6. [PMID: 30034950 PMCID: PMC6052953 DOI: 10.1167/tvst.7.4.6] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 05/28/2018] [Indexed: 12/11/2022] Open
Affiliation(s)
- Jacque L Duncan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Eric A Pierce
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Amy M Laster
- Foundation Fighting Blindness, Columbia, MD, USA
| | - Stephen P Daiger
- Human Genetics Center, School of Public Health, and Ruiz Department of Ophthalmology and Visual Science, The University of Texas Health Science Center, Houston, TX, USA
| | - David G Birch
- Rose-Silverthorne Retinal Degenerations Laboratory, Retina Foundation of the Southwest, Dallas, TX, USA
| | - John D Ash
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Alessandro Iannaccone
- Center for Retinal Degenerations and Ophthalmic Genetic Diseases, Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - John G Flannery
- Vision Science, the Helen Wills Neuroscience Institute, the Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - José A Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Institut de la Vision-Sorbonne Université, Inserm, CNRS-Paris, France
| | - Donald J Zack
- Departments of Ophthalmology, Neuroscience, Molecular Biology and Genetics, and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marco A Zarbin
- Institute of Ophthalmology and Visual Science, Rutgers-New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | | |
Collapse
|
35
|
Trapani I, Auricchio A. Seeing the Light after 25 Years of Retinal Gene Therapy. Trends Mol Med 2018; 24:669-681. [PMID: 29983335 DOI: 10.1016/j.molmed.2018.06.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 05/31/2018] [Accepted: 06/11/2018] [Indexed: 12/25/2022]
Abstract
The retina has been at the forefront of translational gene therapy. Proof-of-concept that gene therapy could restore vision in a large animal led to the initiation of the first successful clinical trials and, in turn, to the recent approval of the first gene therapy product for an ocular disease. As dozens of clinical trials of retinal gene therapy have begun, new challenges are identified, which include delivery of large genes, counteracting gain-of-function mutations, and safe and effective gene transfer to diseased retinas. Advancements in vector design, improvements of delivery routes, and selection of optimal timing for intervention will contribute to extend the initial success of retinal gene therapy to an increasing number of inherited blinding conditions.
Collapse
Affiliation(s)
- Ivana Trapani
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy; Medical Genetics, Department of Translational Medicine, Federico II University, Naples, Italy.
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy; Department of Advanced Biomedicine, Federico II University, Naples, Italy.
| |
Collapse
|
36
|
Burnight ER, Giacalone JC, Cooke JA, Thompson JR, Bohrer LR, Chirco KR, Drack AV, Fingert JH, Worthington KS, Wiley LA, Mullins RF, Stone EM, Tucker BA. CRISPR-Cas9 genome engineering: Treating inherited retinal degeneration. Prog Retin Eye Res 2018; 65:28-49. [PMID: 29578069 PMCID: PMC8210531 DOI: 10.1016/j.preteyeres.2018.03.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 03/15/2018] [Accepted: 03/18/2018] [Indexed: 12/18/2022]
Abstract
Gene correction is a valuable strategy for treating inherited retinal degenerative diseases, a major cause of irreversible blindness worldwide. Single gene defects cause the majority of these retinal dystrophies. Gene augmentation holds great promise if delivered early in the course of the disease, however, many patients carry mutations in genes too large to be packaged into adeno-associated viral vectors and some, when overexpressed via heterologous promoters, induce retinal toxicity. In addition to the aforementioned challenges, some patients have sustained significant photoreceptor cell loss at the time of diagnosis, rendering gene replacement therapy insufficient to treat the disease. These patients will require cell replacement to restore useful vision. Fortunately, the advent of induced pluripotent stem cell and CRISPR-Cas9 gene editing technologies affords researchers and clinicians a powerful means by which to develop strategies to treat patients with inherited retinal dystrophies. In this review we will discuss the current developments in CRISPR-Cas9 gene editing in vivo in animal models and in vitro in patient-derived cells to study and treat inherited retinal degenerative diseases.
Collapse
Affiliation(s)
- Erin R Burnight
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Joseph C Giacalone
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Jessica A Cooke
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Jessica R Thompson
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Laura R Bohrer
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Kathleen R Chirco
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Arlene V Drack
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - John H Fingert
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Kristan S Worthington
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States; Department of Biochemical Engineering, University of Iowa, Iowa City, IA, United States
| | - Luke A Wiley
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Robert F Mullins
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Edwin M Stone
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States
| | - Budd A Tucker
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, University of Iowa, Iowa City, IA, United States.
| |
Collapse
|
37
|
Clinical characteristics of a Japanese patient with Bardet-Biedl syndrome caused by BBS10 mutations. Jpn J Ophthalmol 2018; 62:458-466. [DOI: 10.1007/s10384-018-0591-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 03/02/2018] [Indexed: 11/26/2022]
|
38
|
Forsythe E, Kenny J, Bacchelli C, Beales PL. Managing Bardet-Biedl Syndrome-Now and in the Future. Front Pediatr 2018; 6:23. [PMID: 29487844 PMCID: PMC5816783 DOI: 10.3389/fped.2018.00023] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/25/2018] [Indexed: 11/13/2022] Open
Abstract
Bardet-Biedl syndrome is a rare autosomal recessive multisystem disorder caused by defects in genes encoding for proteins that localize to the primary cilium/basal body complex. Twenty-one disease-causing genes have been identified to date. It is one of the most well-studied conditions in the family of diseases caused by defective cilia collectively known as ciliopathies. In this review, we provide an update on diagnostic developments, clinical features, and progress in the management of Bardet-Biedl syndrome. Advances in diagnostic technologies including exome and whole genome sequencing are expanding the spectrum of patients who are diagnosed with Bardet-Biedl syndrome and increasing the number of cases with diagnostic uncertainty. As a result of the diagnostic developments, a small number of patients with only one or two clinical features of Bardet-Biedl syndrome are being diagnosed. Our understanding of the syndrome-associated renal disease has evolved and is reviewed here. Novel interventions are developing at a rapid pace and are explored in this review including genetic therapeutics such as gene therapy, exon skipping therapy, nonsense suppression therapy, and gene editing. Other non-genetic therapies such as gene repurposing, targeted therapies, and non-pharmacological interventions are also discussed.
Collapse
Affiliation(s)
- Elizabeth Forsythe
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Joanna Kenny
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Chiara Bacchelli
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Philip L Beales
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
39
|
CLINICAL PROGRESS IN INHERITED RETINAL DEGENERATIONS: GENE THERAPY CLINICAL TRIALS AND ADVANCES IN GENETIC SEQUENCING. Retina 2017; 37:417-423. [PMID: 27753762 DOI: 10.1097/iae.0000000000001341] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE Inherited retinal dystrophies are a significant cause of vision loss and are characterized by the loss of photoreceptors and the retinal pigment epithelium (RPE). Mutations in approximately 250 genes cause inherited retinal degenerations with a high degree of genetic heterogeneity. New techniques in next-generation sequencing are allowing the comprehensive analysis of all retinal disease genes thus changing the approach to the molecular diagnosis of inherited retinal dystrophies. This review serves to analyze clinical progress in genetic diagnostic testing and implications for retinal gene therapy. METHODS A literature search of PubMed and OMIM was conducted to relevant articles in inherited retinal dystrophies. RESULTS Next-generation genetic sequencing allows the simultaneous analysis of all the approximately 250 genes that cause inherited retinal dystrophies. Reported diagnostic rates range are high and range from 51% to 57%. These new sequencing tools are highly accurate with sensitivities of 97.9% and specificities of 100%. Retinal gene therapy clinical trials are underway for multiple genes including RPE65, ABCA4, CHM, RS1, MYO7A, CNGA3, CNGB3, ND4, and MERTK for which a molecular diagnosis may be beneficial for patients. CONCLUSION Comprehensive next-generation genetic sequencing of all retinal dystrophy genes is changing the paradigm for how retinal specialists perform genetic testing for inherited retinal degenerations. Not only are high diagnostic yields obtained, but mutations in genes with novel clinical phenotypes are also identified. In the era of retinal gene therapy clinical trials, identifying specific genetic defects will increasingly be of use to identify patients who may enroll in clinical studies and benefit from novel therapies.
Collapse
|
40
|
Weihbrecht K, Goar WA, Pak T, Garrison JE, DeLuca AP, Stone EM, Scheetz TE, Sheffield VC. Keeping an Eye on Bardet-Biedl Syndrome: A Comprehensive Review of the Role of Bardet-Biedl Syndrome Genes in the Eye. MEDICAL RESEARCH ARCHIVES 2017; 5. [PMID: 29457131 DOI: 10.18103/mra.v5i9.1526] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Upwards of 90% of individuals with Bardet-Biedl syndrome (BBS) display rod-cone dystrophy with early macular involvement. BBS is an autosomal recessive, genetically heterogeneous, pleiotropic ciliopathy for which 21 causative genes have been discovered to date. In addition to retinal degeneration, the cardinal features of BBS include obesity, cognitive impairment, renal anomalies, polydactyly, and hypogonadism. Here, we review the genes, proteins, and protein complexes involved in BBS and the BBS model organisms available for the study of retinal degeneration. We include comprehensive lists for all known BBS genes, their known phenotypes, and the model organisms available. We also review the molecular mechanisms believed to lead to retinal degeneration. We provide an overview of the mode of inheritance and describe the relationships between BBS genes and Joubert syndrome, Leber Congenital Amaurosis, Senior-Løken syndrome, and non-syndromic retinitis pigmentosa. Finally, we propose ways that new advances in technology will allow us to better understand the role of different BBS genes in retinal formation and function.
Collapse
Affiliation(s)
- Katie Weihbrecht
- Department of Pediatrics, University of Iowa; Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Wesley A Goar
- Department of Pediatrics, University of Iowa; Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Thomas Pak
- Department of Pediatrics, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Janelle E Garrison
- Department of Pediatrics, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Adam P DeLuca
- Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Edwin M Stone
- Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Todd E Scheetz
- Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| | - Val C Sheffield
- Department of Pediatrics, University of Iowa; Iowa City, IA 52242, USA.,Department of Ophthalmology and Visual Sciences, University of Iowa; Iowa City, IA 52242, USA.,Stephen A. Wynn Institute for Vision Research, University of Iowa; Iowa City, IA 52242, USA
| |
Collapse
|
41
|
Burnight ER, Gupta M, Wiley LA, Anfinson KR, Tran A, Triboulet R, Hoffmann JM, Klaahsen DL, Andorf JL, Jiao C, Sohn EH, Adur MK, Ross JW, Mullins RF, Daley GQ, Schlaeger TM, Stone EM, Tucker BA. Using CRISPR-Cas9 to Generate Gene-Corrected Autologous iPSCs for the Treatment of Inherited Retinal Degeneration. Mol Ther 2017; 25:1999-2013. [PMID: 28619647 PMCID: PMC5589061 DOI: 10.1016/j.ymthe.2017.05.015] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 05/22/2017] [Accepted: 05/22/2017] [Indexed: 01/20/2023] Open
Abstract
Patient-derived induced pluripotent stem cells (iPSCs) hold great promise for autologous cell replacement. However, for many inherited diseases, treatment will likely require genetic repair pre-transplantation. Genome editing technologies are useful for this application. The purpose of this study was to develop CRISPR-Cas9-mediated genome editing strategies to target and correct the three most common types of disease-causing variants in patient-derived iPSCs: (1) exonic, (2) deep intronic, and (3) dominant gain of function. We developed a homology-directed repair strategy targeting a homozygous Alu insertion in exon 9 of male germ cell-associated kinase (MAK) and demonstrated restoration of the retinal transcript and protein in patient cells. We generated a CRISPR-Cas9-mediated non-homologous end joining (NHEJ) approach to excise a major contributor to Leber congenital amaurosis, the IVS26 cryptic-splice mutation in CEP290, and demonstrated correction of the transcript and protein in patient iPSCs. Lastly, we designed allele-specific CRISPR guides that selectively target the mutant Pro23His rhodopsin (RHO) allele, which, following delivery to both patient iPSCs in vitro and pig retina in vivo, created a frameshift and premature stop that would prevent transcription of the disease-causing variant. The strategies developed in this study will prove useful for correcting a wide range of genetic variants in genes that cause inherited retinal degeneration.
Collapse
Affiliation(s)
- Erin R Burnight
- Stephen A. Wynn Institute for Vision Research and Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, USA
| | - Manav Gupta
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 01451, USA
| | - Luke A Wiley
- Stephen A. Wynn Institute for Vision Research and Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, USA
| | - Kristin R Anfinson
- Stephen A. Wynn Institute for Vision Research and Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, USA
| | - Audrey Tran
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 01451, USA
| | - Robinson Triboulet
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 01451, USA
| | - Jeremy M Hoffmann
- Stephen A. Wynn Institute for Vision Research and Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, USA
| | - Darcey L Klaahsen
- Stephen A. Wynn Institute for Vision Research and Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, USA
| | - Jeaneen L Andorf
- Stephen A. Wynn Institute for Vision Research and Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, USA
| | - Chunhua Jiao
- Stephen A. Wynn Institute for Vision Research and Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, USA
| | - Elliott H Sohn
- Stephen A. Wynn Institute for Vision Research and Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, USA
| | - Malavika K Adur
- Department of Animal Sciences, Iowa State University, Ames, IA 50011, USA
| | - Jason W Ross
- Department of Animal Sciences, Iowa State University, Ames, IA 50011, USA
| | - Robert F Mullins
- Stephen A. Wynn Institute for Vision Research and Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, USA
| | - George Q Daley
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 01451, USA
| | - Thorsten M Schlaeger
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 01451, USA
| | - Edwin M Stone
- Stephen A. Wynn Institute for Vision Research and Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, USA
| | - Budd A Tucker
- Stephen A. Wynn Institute for Vision Research and Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52241, USA.
| |
Collapse
|
42
|
Kenny J, Forsythe E, Beales P, Bacchelli C. Toward personalized medicine in Bardet–Biedl syndrome. Per Med 2017; 14:447-456. [DOI: 10.2217/pme-2017-0019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Personalized medicine is becoming routine in the treatment of common diseases such as cancer, but has lagged behind in the field of rare diseases. It is currently in the early stages for the treatment of Bardet–Biedl syndrome. Advances in the understanding of ciliary biology and diagnostic techniques have opened up the prospect of treating BBS in a patient-specific manner. Owing to their structure and function, cilia provide an attractive therapeutic target and genetic therapies are being explored in ciliopathy treatment. Promising avenues include gene therapy, gene editing techniques and splice-correcting and read-through therapies. Targeted drug design has been successful in the treatment of genetic disease and research is underway in the discovery of known and novel drugs to treat Bardet–Biedl syndrome.
Collapse
Affiliation(s)
- Joanna Kenny
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford St, London WC1N 1EH, UK
| | - Elizabeth Forsythe
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford St, London WC1N 1EH, UK
| | - Philip Beales
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford St, London WC1N 1EH, UK
| | - Chiara Bacchelli
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford St, London WC1N 1EH, UK
| |
Collapse
|
43
|
Zeng Y, Petralia RS, Vijayasarathy C, Wu Z, Hiriyanna S, Song H, Wang YX, Sieving PA, Bush RA. Retinal Structure and Gene Therapy Outcome in Retinoschisin-Deficient Mice Assessed by Spectral-Domain Optical Coherence Tomography. Invest Ophthalmol Vis Sci 2017; 57:OCT277-87. [PMID: 27409484 PMCID: PMC4968785 DOI: 10.1167/iovs.15-18920] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose Spectral-domain optical coherence tomography (SD-OCT) was used to characterize the retinal phenotype, natural history, and treatment responses in a mouse model of X-linked retinoschisis (Rs1-KO) and to identify new structural markers of AAV8-mediated gene therapy outcome. Methods Optical coherence tomography scans were performed on wild-type and Rs1-KO mouse retinas between 1 and 12 months of age and on Rs1-KO mice after intravitreal injection of AAV8-scRS/IRBPhRS (AAV8-RS1). Cavities and photoreceptor outer nuclear layer (ONL) thickness were measured, and outer retina reflective band (ORRB) morphology was examined with age and after AAV8-RS1 treatment. Outer retina reflective band morphology was compared to immunohistochemical staining of the outer limiting membrane (OLM) and photoreceptor inner segment (IS) mitochondria and to electron microscopy (EM) images of IS. Results Retinal cavity size in Rs1-KO mice increased between 1 and 4 months and decreased thereafter, while ONL thickness declined steadily, comparable to previous histologic studies. Wild-type retina had four ORRBs. In Rs1-KO, ORRB1was fragmented from 1 month, but was normal after 8 months; ORRB2 and ORRB3 were merged at all ages. Outer retina reflective band morphology returned to normal after AAV-RS1 therapy, paralleling the recovery of the OLM and IS mitochondria as indicated by anti–β-catenin and anti-COX4 labeling, respectively, and EM. Conclusions Spectral-domain OCT is a sensitive, noninvasive tool to monitor subtle changes in retinal morphology, disease progression, and effects of therapies in mouse models. The ORRBs may be useful to assess the outcome of gene therapy in the treatment of X-linked retinoschisis patients.
Collapse
Affiliation(s)
- Yong Zeng
- Section on Translational Research for Retinal and Macular Degeneration National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States
| | - Ronald S Petralia
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States
| | - Camasamudram Vijayasarathy
- Section on Translational Research for Retinal and Macular Degeneration National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States
| | - Zhijian Wu
- Ocular Gene Therapy Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Suja Hiriyanna
- Ocular Gene Therapy Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Hongman Song
- Section on Translational Research for Retinal and Macular Degeneration National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States
| | - Ya-Xian Wang
- Advanced Imaging Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States
| | - Paul A Sieving
- Section on Translational Research for Retinal and Macular Degeneration National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States 4National Eye Institute, National Institutes of Healt
| | - Ronald A Bush
- Section on Translational Research for Retinal and Macular Degeneration National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
44
|
Williams CL, Uytingco CR, Green WW, McIntyre JC, Ukhanov K, Zimmerman AD, Shively DT, Zhang L, Nishimura DY, Sheffield VC, Martens JR. Gene Therapeutic Reversal of Peripheral Olfactory Impairment in Bardet-Biedl Syndrome. Mol Ther 2017; 25:904-916. [PMID: 28237838 DOI: 10.1016/j.ymthe.2017.02.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/02/2017] [Accepted: 02/05/2017] [Indexed: 12/22/2022] Open
Abstract
Olfactory dysfunction is a pervasive but underappreciated health concern that affects personal safety and quality of life. Patients with olfactory dysfunctions have limited therapeutic options, particularly those involving congenital diseases. Bardet-Biedl syndrome (BBS) is one such disorder, where olfactory loss and other symptoms manifest from defective cilium morphology and/or function in various cell types/tissues. Olfactory sensory neurons (OSNs) of BBS mutant mice lack the capacity to build/maintain cilia, rendering the cells incapable of odor detection. Here we examined OSN cilium defects in Bbs1 mutant mice and assessed the utility of gene therapy to restore ciliation and function in young and adult mice. Bbs1 mutant mice possessed short residual OSN cilia in which BBSome protein trafficking and odorant detection were defective. Gene therapy with an adenovirus-delivered wild-type Bbs1 gene restored OSN ciliation, corrected BBSome cilium trafficking defects, and returned acute odor responses. Finally, using clinically approved AAV serotypes, we demonstrate, for the first time, the capacity of AAVs to restore ciliation and odor detection in OSNs of Bbs1 mutants. Together, our data demonstrate that OSN ciliogenesis can be promoted in differentiated cells of young and adult Bbs1 mutants and highlight the potential of gene therapy as a viable restorative treatment for congenital olfactory disorders.
Collapse
Affiliation(s)
- Corey L Williams
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Cedric R Uytingco
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Warren W Green
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Jeremy C McIntyre
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Kirill Ukhanov
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Arthur D Zimmerman
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Dana T Shively
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Lian Zhang
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | - Val C Sheffield
- Department of Pediatrics, University of Iowa, Iowa City, IA 52242, USA; Howard Hughes Medical Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Jeffrey R Martens
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Center for Smell and Taste, University of Florida College of Medicine, Gainesville, FL 32610, USA.
| |
Collapse
|
45
|
Ruan GX, Barry E, Yu D, Lukason M, Cheng SH, Scaria A. CRISPR/Cas9-Mediated Genome Editing as a Therapeutic Approach for Leber Congenital Amaurosis 10. Mol Ther 2017; 25:331-341. [PMID: 28109959 PMCID: PMC5368591 DOI: 10.1016/j.ymthe.2016.12.006] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 12/06/2016] [Accepted: 12/06/2016] [Indexed: 12/18/2022] Open
Abstract
As the most common subtype of Leber congenital amaurosis (LCA), LCA10 is a severe retinal dystrophy caused by mutations in the CEP290 gene. The most frequent mutation found in patients with LCA10 is a deep intronic mutation in CEP290 that generates a cryptic splice donor site. The large size of the CEP290 gene prevents its use in adeno-associated virus (AAV)-mediated gene augmentation therapy. Here, we show that targeted genomic deletion using the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system represents a promising therapeutic approach for the treatment of patients with LCA10 bearing the CEP290 splice mutation. We generated a cellular model of LCA10 by introducing the CEP290 splice mutation into 293FT cells and we showed that guide RNA pairs coupled with SpCas9 were highly efficient at removing the intronic splice mutation and restoring the expression of wild-type CEP290. In addition, we demonstrated that a dual AAV system could effectively delete an intronic fragment of the Cep290 gene in the mouse retina. To minimize the immune response to prolonged expression of SpCas9, we developed a self-limiting CRISPR/Cas9 system that minimizes the duration of SpCas9 expression. These results support further studies to determine the therapeutic potential of CRISPR/Cas9-based strategies for the treatment of patients with LCA10.
Collapse
Affiliation(s)
- Guo-Xiang Ruan
- Rare Diseases, Sanofi Genzyme, Framingham, MA 01701, USA.
| | | | - Dan Yu
- Rare Diseases, Sanofi Genzyme, Framingham, MA 01701, USA
| | | | - Seng H Cheng
- Rare Diseases, Sanofi Genzyme, Framingham, MA 01701, USA
| | - Abraham Scaria
- Rare Diseases, Sanofi Genzyme, Framingham, MA 01701, USA
| |
Collapse
|
46
|
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: 2.9] [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
|
47
|
Abstract
Primary cilia are organelles that are present on many different cell types, either transiently or permanently. They play a crucial role in receiving signals from the environment and passing these signals to other parts of the cell. In that way, they are involved in diverse processes such as adipocyte differentiation and olfactory sensation. Mutations in genes coding for ciliary proteins often have pleiotropic effects and lead to clinical conditions, ciliopathies, with multiple symptoms. In this study, we reviewed observations from ciliopathies with obesity as one of the symptoms. It shows that variation in cilia-related genes is itself not a major cause of obesity in the population but may be a part of the multifactorial aetiology of this complex condition. Both common polymorphisms and rare deleterious variants may contribute to the obesity risk. Genotype-phenotype relationships have been noticed. Among the ciliary genes, obesity differs with regard to severity and age of onset, which may relate to the influence of each gene on the balance between pro- and anti-adipogenic processes. Analysis of the function and location of the proteins encoded by these ciliary genes suggests that obesity is more linked to activities at the basal area of the cilium, including initiation of the intraflagellar transport, but less to the intraflagellar transport itself. Regarding the role of cilia, three possible mechanistic processes underlying obesity are described: adipogenesis, neuronal food intake regulation and food odour perception.
Collapse
|
48
|
Abstract
Bardet-Biedl syndrome (BBS) is a rare autosomal recessive genetic disorder. It is characterized by heterogeneous clinical manifestations including primary features of the disease (rod-cone dystrophy, polydactyly, obesity, genital abnormalities, renal defects, and learning difficulties) and secondary BBS characteristics (developmental delay, speech deficit, brachydactyly or syndactyly, dental defects, ataxia or poor coordination, olfactory deficit, diabetes mellitus, congenital heart disease, etc.); most of these symptoms may not be present at birth but appear and progressively worsen during the first and second decades of life. At least 20 BBS genes have already been identified, and all of them are involved in primary cilia functioning. Genetic diagnosis of BBS is complicated due to lack of gene-specific disease symptoms; however, it is gradually becoming more accessible with the invention of multigene sequencing technologies. Clinical management of BBS is largely limited to a symptomatic treatment. Mouse experiments demonstrate that the most debilitating complication of BBS, blindness, can be rescued by topical gene therapy. There is a published case report describing the delay of BBS symptoms by nutritional compensation of the disease-related biochemical deficiencies. Progress in DNA testing technologies is likely to rapidly resolve all limitations in BBS diagnosis; however, much slower improvement is expected with regard to BBS treatment.
Collapse
Affiliation(s)
- Evgeny N Suspitsin
- N.N. Petrov Institute of Oncology, St. Petersburg, Russia; St. Petersburg Pediatric Medical University, St. Petersburg, Russia
| | - Evgeny N Imyanitov
- N.N. Petrov Institute of Oncology, St. Petersburg, Russia; St. Petersburg Pediatric Medical University, St. Petersburg, Russia; I.I. Mechnikov North-Western Medical University, St. Petersburg, Russia; St. Petersburg State University, St. Petersburg, Russia
| |
Collapse
|
49
|
Butler MC, Sullivan JM. A Novel, Real-Time, In Vivo Mouse Retinal Imaging System. Invest Ophthalmol Vis Sci 2016; 56:7159-68. [PMID: 26551329 DOI: 10.1167/iovs.14-16370] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
PURPOSE To develop an efficient, low-cost instrument for robust real-time imaging of the mouse retina in vivo, and assess system capabilities by evaluating various animal models. METHODS Following multiple disappointing attempts to visualize the mouse retina during a subretinal injection using commercially available systems, we identified the key limitation to be inadequate illumination due to off axis illumination and poor optical train optimization. Therefore, we designed a paraxial illumination system for Greenough-type stereo dissecting microscope incorporating an optimized optical launch and an efficiently coupled fiber optic delivery system. Excitation and emission filters control spectral bandwidth. A color coupled-charged device (CCD) camera is coupled to the microscope for image capture. Although, field of view (FOV) is constrained by the small pupil aperture, the high optical power of the mouse eye, and the long working distance (needed for surgical manipulations), these limitations can be compensated by eye positioning in order to observe the entire retina. RESULTS The retinal imaging system delivers an adjustable narrow beam to the dilated pupil with minimal vignetting. The optic nerve, vasculature, and posterior pole are crisply visualized and the entire retina can be observed through eye positioning. Normal and degenerative retinal phenotypes can be followed over time. Subretinal or intraocular injection procedures are followed in real time. Real-time, intravenous fluorescein angiography for the live mouse has been achieved. CONCLUSIONS A novel device is established for real-time viewing and image capture of the small animal retina during subretinal injections for preclinical gene therapy studies.
Collapse
Affiliation(s)
- Mark C Butler
- Research Service, VA Western New York Healthcare System, Buffalo, New York, United States 2Department of Ophthalmology, Ross Eye Institute, University at Buffalo-SUNY, Buffalo, New York, United States
| | - Jack M Sullivan
- Research Service, VA Western New York Healthcare System, Buffalo, New York, United States 2Department of Ophthalmology, Ross Eye Institute, University at Buffalo-SUNY, Buffalo, New York, United States 3Department of Pharmacology/Toxicology, University at
| |
Collapse
|
50
|
Intravitreal Injection of Splice-switching Oligonucleotides to Manipulate Splicing in Retinal Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e250. [PMID: 26325627 PMCID: PMC4877449 DOI: 10.1038/mtna.2015.24] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 07/22/2015] [Indexed: 12/25/2022]
Abstract
Leber congenital amaurosis is a severe hereditary retinal dystrophy responsible for neonatal blindness. The most common disease-causing mutation (c.2991+1655A>G; 10-15%) creates a strong splice donor site that leads to insertion of a cryptic exon encoding a premature stop codon. Recently, we reported that splice-switching oligonucleotides (SSO) allow skipping of the mutant cryptic exon and the restoration of ciliation in fibroblasts of affected patients, supporting the feasibility of a SSO-mediated exon skipping strategy to correct the aberrant splicing. Here, we present data in the wild-type mouse, which demonstrate that intravitreal administration of 2'-OMePS-SSO allows selective alteration of Cep290 splicing in retinal cells, including photoreceptors as shown by successful alteration of Abca4 splicing using the same approach. We show that both SSOs and Cep290 skipped mRNA were detectable for at least 1 month and that intravitreal administration of oligonucleotides did not provoke any serious adverse event. These data suggest that intravitreal injections of SSO should be considered to bypass protein truncation resulting from the c.2991+1655A>G mutation as well as other truncating mutations in genes which like CEP290 or ABCA4 have a mRNA size that exceed cargo capacities of US Food and Drug Administration (FDA)-approved adeno-associated virus (AAV)-vectors, thus hampering gene augmentation therapy.
Collapse
|