1
|
Chew LA, Grigsby D, Hester CG, Amason J, McPherson WK, Flynn EJ, Visel M, Starr CR, Flannery JG, Lewis TR, Bowes Rickman C. Truncated complement factor H Y402 gene therapy rescues C3 glomerulonephritis. Mol Ther 2025:S1525-0016(25)00314-4. [PMID: 40285355 DOI: 10.1016/j.ymthe.2025.04.035] [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: 12/16/2024] [Revised: 03/06/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025] Open
Abstract
There are no effective therapies for patients with dry age-related macular degeneration (AMD) or C3 glomerulonephritis (C3G). Unfortunately, past efforts to treat C3G using exogenous human complement factor H (CFH) found limited success due to immune rejection of a foreign protein response. AMD research has also faced myriad challenges, including the absence of an ideal therapeutic target and difficulties with treatment delivery in certain preclinical models. In pursuit of an AMD therapy to overcome these obstacles, we ultimately capitalized on parallels in complement dysregulation between AMD and C3G. Here, we investigate the potential for CFH supplementation as a strategy to rescue C3G. Our findings demonstrate restored inhibition of complement's alternative pathway and long-term reversal of disease without immune rejection using adeno-associated virus (AAV)-mediated delivery of truncated CFH (tCFH) in a Cfh-/- mouse model of C3G. We tested three different tCFH vectors and found significant differences in their relative transduction efficiency and therapeutic efficacy. These discoveries motivate the development of AAV-mediated tCFH replacement therapy for patients with C3G while simultaneously demonstrating proof of concept for AAV-mediated tCFH gene augmentation therapy for patients with AMD.
Collapse
Affiliation(s)
- Lindsey A Chew
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Daniel Grigsby
- Genetically Engineered Murine Model (GEMM) Core, University of Virginia, Charlottesville, VA 22903, USA
| | - C Garren Hester
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Joshua Amason
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710, USA
| | - W Kyle McPherson
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Edward J Flynn
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Meike Visel
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Christopher R Starr
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - John G Flannery
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Tylor R Lewis
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
2
|
Zhang Q, He J, Zhu D, Chen Y, Fu M, Lu S, Qiu Y, Zhou G, Yang G, Jiang Z. Genetically modified organoids for tissue engineering and regenerative medicine. Adv Colloid Interface Sci 2025; 335:103337. [PMID: 39547125 DOI: 10.1016/j.cis.2024.103337] [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: 01/18/2024] [Revised: 07/23/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
To date, genetically modified organoids are emerging as a promising 3D modeling tool aimed at solving genetically relevant clinical and biomedical problems for regenerative medicine and tissue engineering. As an optimal vehicle for gene delivery, genetically modified organoids can enhance or reduce the expression of target genes through virus and non-virus-based gene transfection methods to achieve tissue regeneration. Animal experiments and preclinical studies have demonstrated the beneficial role of genetically modified organoids in various aspects of organ regeneration, including thymus, lacrimal glands, brain, lung, kidney, photoreceptors, etc. Furthermore, the technology offers a potential treatment option for various diseases, such as Fabry disease, non-alcoholic steatohepatitis, and Lynch syndrome. Nevertheless, the uncertain safety of genetic modification, the risk of organoid application, and bionics of current genetically modified organoids are still challenging. This review summarizes the researches on genetically modified organoids in recent years, and describes the transfection methods and functions of genetically modified organoids, then introduced their applications at length. Also, the limitations and future development directions of genetically modified organoids are included.
Collapse
Affiliation(s)
- Qinmeng Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Jin He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Danji Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Yunxuan Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Mengdie Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Shifan Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Yuesheng Qiu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guodong Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
3
|
Vats A, Xi Z, Byrne LC, Chen Y. Retinal Explant Culture from Mouse, Human, and Nonhuman Primates and Its Applications in Vision Research. Methods Mol Biol 2025; 2848:169-186. [PMID: 39240523 DOI: 10.1007/978-1-0716-4087-6_11] [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
The retinal explant culture system is a valuable tool for studying the pharmacological, toxicological, and developmental aspects of the retina. It is also used for translational studies such as gene therapy. While no photoreceptor-like cell lines are available for in vitro studies of photoreceptor cell biology, the retinal explant culture maintains the laminated retinal structure ex vivo for as long as a month. Human and nonhuman primate (NHP) postmortem retinal explants cut into small pieces offer the possibility of testing multiple conditions for safety and adeno-associated viral (AAV) vector optimization. In addition, the cone-enriched foveal area can be studied using the retinal explants. Here, we present a detailed working protocol for retinal explant isolation and culture from mouse, human, and NHP for testing drug efficacy and AAV transduction. Future applications of this protocol include combining live imaging and multiwell retinal explant culture for high-throughput drug screening systems in rodent and human retinal explants to identify new drugs against retinal degeneration.
Collapse
Affiliation(s)
- Abhishek Vats
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhouhuan Xi
- Department of Ophthalmology, Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - 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
| | - Yuanyuan Chen
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
4
|
Chew LA, Grigsby D, Hester CG, Amason J, McPherson WK, Flynn EJ, Visel M, Flannery JG, Rickman CB. Truncated Complement Factor H Y402 Gene Therapy Cures C3 Glomerulonephritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613471. [PMID: 39345485 PMCID: PMC11429740 DOI: 10.1101/2024.09.17.613471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Patients with both age-related macular degeneration (AMD) and C3 glomerulonephritis (C3G) are challenged by the absence of effective therapies to reverse and eliminate their disease burden. Capitalizing on complement dysregulation as both a significant risk factor for AMD and the known pathophysiology of C3G, we investigated the potential for adeno-associated virus (AAV) delivery of complement factor H (CFH) to rescue C3G in a Cfh-/- mouse model of C3G. While past efforts to treat C3G using exogenous human CFH resulted in limited success before immune rejection led to a foreign protein response, our findings demonstrate the capacity for long-term AAV-mediated delivery of truncated CFH (tCFH) to restore inhibition of the alternative pathway of complement and ultimately reverse C3G without immune rejection. Comparing results from the administration of several tCFH vectors also revealed significant differences in their relative efficiency and efficacy. These discoveries pave the way for subsequent development of AAV-mediated tCFH replacement therapy for patients with C3G, while simultaneously demonstrating proof of concept for a parallel AAV-mediated tCFH gene augmentation therapy for patients with AMD.
Collapse
Affiliation(s)
- Lindsey A. Chew
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710
| | - Daniel Grigsby
- Genetically Engineered Murine Model (GEMM) Core, University of Virginia, Charlottesville, VA, 22903
| | - C. Garren Hester
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710
| | - Joshua Amason
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710
| | - W. Kyle McPherson
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710
| | - Edward J. Flynn
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710
| | - Meike Visel
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720
| | - John G. Flannery
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, 94720
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke Eye Center, Duke University Medical Center, Durham, NC 27710
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
5
|
Xia CH, Liu H, Li M, Zhang H, Xing X, Gong X. Identification and Characterization of Retinitis Pigmentosa in a Novel Mouse Model Caused by PDE6B-T592I. Biomedicines 2023; 11:3173. [PMID: 38137394 PMCID: PMC10740990 DOI: 10.3390/biomedicines11123173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
The cGMP-phosphodiesterase 6 beta subunit (PDE6B) is an essential component in the phototransduction pathway for light responses in photoreceptor cells. PDE6B gene mutations cause the death of rod photoreceptors, named as hereditary retinitis pigmentosa (RP) in humans and retinal degeneration (RD) in rodents. Here, we report a new RD model, identified from a phenotypic screen of N-ethyl-N-nitrosourea (ENU)-induced mutant mice, which displays retinal degeneration caused by a point mutation in the Pde6b gene that results in PDE6B-T592I mutant protein. The homozygous mutant mice show an extensive loss of rod photoreceptors at the age of 3 weeks; unexpectedly, the loss of rod photoreceptors can be partly rescued by dark rearing. Thus, this RD mutant model displays a light-dependent loss of rod photoreceptors. Both western blot and immunostaining results show very low level of mutant PDE6B-T592I protein in the retina. Structure modeling suggests that the T592I mutation probably affects the function and stability of PDE6B protein by changing intramolecular interactions. We further demonstrate that the expression of wild-type PDE6B delivered by subretinally injected adeno-associated virus (rAAV) prevents photoreceptor cell death in this RD model in vivo. The PDE6B-T592I mutant is, therefore, a valuable RD model for evaluating rAAV-mediated treatment and for investigating the molecular mechanism of light-dependent rod photoreceptor cell death that is related to impaired PDE6B function.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaohua Gong
- Herbert Wertheim School of Optometry and Vision Science Program, University of California, Berkeley, CA 94720, USA; (C.-H.X.); (H.L.); (M.L.); (H.Z.); (X.X.)
| |
Collapse
|
6
|
Panikker P, Roy S, Ghosh A, Poornachandra B, Ghosh A. Advancing precision medicines for ocular disorders: Diagnostic genomics to tailored therapies. Front Med (Lausanne) 2022; 9:906482. [PMID: 35911417 PMCID: PMC9334564 DOI: 10.3389/fmed.2022.906482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/29/2022] [Indexed: 11/20/2022] Open
Abstract
Successful sequencing of the human genome and evolving functional knowledge of gene products has taken genomic medicine to the forefront, soon combining broadly with traditional diagnostics, therapeutics, and prognostics in patients. Recent years have witnessed an extraordinary leap in our understanding of ocular diseases and their respective genetic underpinnings. As we are entering the age of genomic medicine, rapid advances in genome sequencing, gene delivery, genome surgery, and computational genomics enable an ever-increasing capacity to provide a precise and robust diagnosis of diseases and the development of targeted treatment strategies. Inherited retinal diseases are a major source of blindness around the world where a large number of causative genes have been identified, paving the way for personalized diagnostics in the clinic. Developments in functional genetics and gene transfer techniques has also led to the first FDA approval of gene therapy for LCA, a childhood blindness. Many such retinal diseases are the focus of various clinical trials, making clinical diagnoses of retinal diseases, their underlying genetics and the studies of natural history important. Here, we review methodologies for identifying new genes and variants associated with various ocular disorders and the complexities associated with them. Thereafter we discuss briefly, various retinal diseases and the application of genomic technologies in their diagnosis. We also discuss the strategies, challenges, and potential of gene therapy for the treatment of inherited and acquired retinal diseases. Additionally, we discuss the translational aspects of gene therapy, the important vector types and considerations for human trials that may help advance personalized therapeutics in ophthalmology. Retinal disease research has led the application of precision diagnostics and precision therapies; therefore, this review provides a general understanding of the current status of precision medicine in ophthalmology.
Collapse
Affiliation(s)
| | - Shomereeta Roy
- Grow Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| | - Anuprita Ghosh
- Grow Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| | | | - Arkasubhra Ghosh
- Grow Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| |
Collapse
|
7
|
Miyadera K, Santana E, Roszak K, Iffrig S, Visel M, Iwabe S, Boyd RF, Bartoe JT, Sato Y, Gray A, Ripolles-Garcia A, Dufour VL, Byrne LC, Flannery JG, Beltran WA, Aguirre GD. Targeting ON-bipolar cells by AAV gene therapy stably reverses LRIT3-congenital stationary night blindness. Proc Natl Acad Sci U S A 2022; 119:e2117038119. [PMID: 35316139 PMCID: PMC9060458 DOI: 10.1073/pnas.2117038119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/10/2022] [Indexed: 01/08/2023] Open
Abstract
SignificanceCanine models of inherited retinal diseases have helped advance adeno-associated virus (AAV)-based gene therapies targeting specific cells in the outer retina for treating blinding diseases in patients. However, therapeutic targeting of diseases such as congenital stationary night blindness (CSNB) that exhibit defects in ON-bipolar cells (ON-BCs) of the midretina remains underdeveloped. Using a leucine-rich repeat, immunoglobulin-like and transmembrane domain 3 (LRIT3) mutant canine model of CSNB exhibiting ON-BC dysfunction, we tested the ability of cell-specific AAV capsids and promotors to specifically target ON-BCs for gene delivery. Subretinal injection of one vector demonstrated safety and efficacy with robust and stable rescue of electroretinography signals and night vision up to 1 y, paving the way for clinical trials in patients.
Collapse
Affiliation(s)
- Keiko Miyadera
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Evelyn Santana
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Karolina Roszak
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Sommer Iffrig
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Meike Visel
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Simone Iwabe
- Ophthalmology Services, Charles River Laboratories, Mattawan, MI 49071
| | - Ryan F. Boyd
- Ophthalmology Services, Charles River Laboratories, Mattawan, MI 49071
| | - Joshua T. Bartoe
- Ophthalmology Services, Charles River Laboratories, Mattawan, MI 49071
| | - Yu Sato
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Alexa Gray
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Ana Ripolles-Garcia
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Valérie L. Dufour
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Leah C. Byrne
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - John G. Flannery
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - William A. Beltran
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Gustavo D. Aguirre
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
8
|
Zin EA, Han D, Tran J, Morisson-Welch N, Visel M, Kuronen M, Flannery JG. Outcomes of progranulin gene therapy in the retina are dependent on time and route of delivery. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 22:40-51. [PMID: 34485593 PMCID: PMC8390452 DOI: 10.1016/j.omtm.2021.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/19/2021] [Indexed: 11/25/2022]
Abstract
Neuronal ceroid lipofuscinosis (NCL) is a family of neurodegenerative diseases caused by mutations to genes related to lysosomal function. One variant, CNL11, is caused by mutations to the gene encoding the protein progranulin, which regulates neuronal lysosomal function. Absence of progranulin causes cerebellar atrophy, seizures, dementia, and vision loss. As progranulin gene therapies targeting the brain are developed, it is advantageous to focus on the retina, as its characteristics are beneficial for gene therapy development: the retina is easily visible through direct imaging, can be assessed through quantitative methods in vivo, and requires smaller amounts of adeno-associated virus (AAV). In this study we characterize the retinal degeneration in a progranulin knockout mouse model of CLN11 and study the effects of gene replacement at different time points. Mice heterologously expressing progranulin showed a reduction in lipofuscin deposits and microglia infiltration. While mice that receive systemic AAV92YF-scCAG-PGRN at post-natal day 3 or 4 show a reduction in retina thinning, mice injected intravitreally at months 1 and 6 with AAV2.7m8-scCAG-PGRN exhibit no improvement, and mice injected at 12 months of age have thinner retinas than do their controls. Thus, delivery of progranulin proves to be time sensitive and dependent on route of administration, requiring early delivery for optimal therapeutic benefit.
Collapse
Affiliation(s)
- Emilia A Zin
- Vision Science Group, School of Optometry, UC Berkeley, Berkeley, CA 94720, USA
| | - Daisy Han
- Department of Integrative Biology, UC Berkeley, Berkeley, CA 94720, USA
| | - Jennifer Tran
- School of Optometry, UC Berkeley, Berkeley, CA 94720, USA
| | | | - Meike Visel
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, CA 94720, USA
| | - Mervi Kuronen
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, CA 94720, USA
| | - John G Flannery
- Vision Science Group, School of Optometry, UC Berkeley, Berkeley, CA 94720, USA.,School of Optometry, UC Berkeley, Berkeley, CA 94720, USA.,Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
9
|
Hu ML, Edwards TL, O'Hare F, Hickey DG, Wang JH, Liu Z, Ayton LN. Gene therapy for inherited retinal diseases: progress and possibilities. Clin Exp Optom 2021; 104:444-454. [PMID: 33689657 DOI: 10.1080/08164622.2021.1880863] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Inherited retinal diseases (IRDs) comprise a heterogeneous group of genetic disorders affecting the retina. Caused by mutations in over 300 genes, IRDs result in visual impairment due to dysfunction and degeneration of photoreceptors, retinal pigment epithelium, or the choroid. Important photoreceptor IRDs include retinitis pigmentosa and Leber congenital amaurosis. Macular dystrophies include Stargardt and Best disease. Currently, IRDs are largely incurable but the landscape of treatment options is rapidly changing for these diseases which, untreated, result in severe visual impairment and blindness.Advances in DNA delivery to the retina and improved genetic diagnosis of IRDs have led to a new era of research into gene therapy for these vision-threatening disorders. Gene therapy is a compelling approach due to the monogenic nature of most IRDs, with the retina being a favourable target for administering genetic vectors due to its immunoprivileged environment, direct visibility, and multiple methods to assess sensitivity and function. Generally, retinal gene therapy involves a subretinal or intravitreal injection of a viral vector, which infects target cells to deliver a therapeutic gene, or transgene. A gene augmentation strategy introduces a functioning copy of a gene to restore expression of a mutated gene, whereas a gene-editing strategy aims to directly edit and correct the mutation. Common delivery vectors include adeno-associated virus (AAV) and lentivirus.Voretigene neparvovec-rzyl (Luxturna) became the first FDA-approved direct gene therapy in December 2017, and the Australian TGA followed suit in August 2020. More are projected to follow, with clinical trials underway for many other IRDs.This review provides an overview of gene therapy for IRDs, including current progress and challenges. A companion article in this issue details target patient populations for IRD gene therapy, and how optometrists can assist in assessing individuals who may be eligible for current and future therapies.
Collapse
Affiliation(s)
- Monica L Hu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Thomas L Edwards
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Department of Surgery (Ophthalmology), Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Australia
| | - Fleur O'Hare
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Department of Surgery (Ophthalmology), Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Australia.,Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Australia
| | - Doron G Hickey
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Jiang-Hui Wang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Zhengyang Liu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Lauren N Ayton
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia.,Department of Surgery (Ophthalmology), Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Australia.,Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Australia
| |
Collapse
|
10
|
Tummala G, Crain A, Rowlan J, Pepple KL. Characterization of Gene Therapy Associated Uveitis Following Intravitreal Adeno-Associated Virus Injection in Mice. Invest Ophthalmol Vis Sci 2021; 62:41. [PMID: 33630023 PMCID: PMC7910624 DOI: 10.1167/iovs.62.2.41] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/30/2021] [Indexed: 12/26/2022] Open
Abstract
Purpose To characterize the intraocular immune cell infiltrate induced by intravitreal adeno-associated virus (AAV) gene therapy. Methods AAV vectors carrying plasmids expressing green fluorescent protein under the control of PR2.1 were injected intravitreally into AAV naive and AAV primed C57Bl/6 mice. Clinical inflammation was assessed using optical coherence tomography. Intraocular immune cell populations were identified and quantified by flow cytometry on days 1, 7, and 29 after intravitreal injection and compared with sham and fellow eye controls. Results Optical coherence tomography inflammation score and total CD45+ cell number were significantly higher in AAV injected eyes compared to uninjected fellow eye and sham injected controls. Clinically apparent inflammation (vitritis on optical coherence tomography) and cellular inflammation (CD45+ cell number) was significantly increased in AAV injected eyes and peaked around day 7. Vitritis resolved by day 29, but cellular inflammation persisted through day 29. On day 1, neutrophils and activated monocytes were the dominant cell populations in all AAV injected eyes. On day 7, eyes of AAV exposed animals had significantly more dendritic cells and T cells than eyes of AAV naive animals. By day 29, CD8- T cells were the dominant CD45+ cell population in AAV injected eyes. Conclusions Intravitreal AAV injection in mice generates clinically evident inflammation that is mild and seems to resolve spontaneously. However, the total number of intraocular CD45+ cells, particularly T cells, remain elevated. Both innate and adaptive immune cells respond to intravitreal AAV regardless of prior immune status, but the adaptive response is delayed in AAV naive eyes.
Collapse
Affiliation(s)
- Gayathri Tummala
- University of Washington, Department of Ophthalmology, Seattle, Washington, United States
| | - Adam Crain
- University of Washington, Department of Ophthalmology, Seattle, Washington, United States
| | - Jessica Rowlan
- University of Washington, Department of Ophthalmology, Seattle, Washington, United States
| | - Kathryn L. Pepple
- University of Washington, Department of Ophthalmology, Seattle, Washington, United States
| |
Collapse
|
11
|
Krueger JN, Wilmot JH, Teratani-Ota Y, Puhger KR, Nemes SE, Crestani AP, Lafreniere MM, Wiltgen BJ. Amnesia for context fear is caused by widespread disruption of hippocampal activity. Neurobiol Learn Mem 2020; 175:107295. [PMID: 32822864 PMCID: PMC8562570 DOI: 10.1016/j.nlm.2020.107295] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/26/2022]
Abstract
The hippocampus plays an essential role in the formation and retrieval of episodic memories in humans and contextual memories in animals. However, amnesia is not always observed when this structure is compromised. To determine why this is the case, we compared the effects of several different circuit manipulations on memory retrieval and hippocampal activity. Mice were first trained on context fear conditioning and then optogenetic and chemogenetic tools were used to alter activity during memory retrieval. We found that retrieval was only impaired when manipulations caused widespread changes (increases or decreases) in hippocampal activity. Widespread increases occurred when pyramidal cells were excited and widespread decreases were found when GABAergic neurons were stimulated. Direct hyperpolarization of excitatory neurons only moderately reduced activity and did not produce amnesia. Surprisingly, widespread decreases in hippocampal activity did not prevent retrieval if they occurred gradually prior to testing. This suggests that intact brain regions can express contextual memories if they are given adequate time to compensate for the loss of the hippocampus.
Collapse
Affiliation(s)
- Jamie N Krueger
- Center for Neuroscience, University of California Davis, 1544 Newton Ct., Davis, CA 95618, United States.
| | - Jacob H Wilmot
- Department of Psychology, University of California Davis, 135 Young Hall, Davis, CA 95616, United States.
| | - Yusuke Teratani-Ota
- Department of Psychology, University of California Davis, 135 Young Hall, Davis, CA 95616, United States.
| | - Kyle R Puhger
- Department of Psychology, University of California Davis, 135 Young Hall, Davis, CA 95616, United States.
| | - Sonya E Nemes
- Center for Neuroscience, University of California Davis, 1544 Newton Ct., Davis, CA 95618, United States.
| | - Ana P Crestani
- Department of Psychology, University of California Davis, 135 Young Hall, Davis, CA 95616, United States.
| | - Marrisa M Lafreniere
- Center for Neuroscience, University of California Davis, 1544 Newton Ct., Davis, CA 95618, United States.
| | - Brian J Wiltgen
- Center for Neuroscience, University of California Davis, 1544 Newton Ct., Davis, CA 95618, United States; Department of Psychology, University of California Davis, 135 Young Hall, Davis, CA 95616, United States.
| |
Collapse
|
12
|
Chung SH, Mollhoff IN, Nguyen U, Nguyen A, Stucka N, Tieu E, Manna S, Meleppat RK, Zhang P, Nguyen EL, Fong J, Zawadzki R, Yiu G. Factors Impacting Efficacy of AAV-Mediated CRISPR-Based Genome Editing for Treatment of Choroidal Neovascularization. Mol Ther Methods Clin Dev 2020; 17:409-417. [PMID: 32128346 PMCID: PMC7044682 DOI: 10.1016/j.omtm.2020.01.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022]
Abstract
Frequent injections of anti-vascular endothelial growth factor (anti-VEGF) agents are a clinical burden for patients with neovascular age-related macular degeneration (AMD). Genomic disruption of VEGF-A using adeno-associated viral (AAV) delivery of clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 has the potential to permanently suppress aberrant angiogenesis, but the factors that determine the optimal efficacy are unknown. Here, we investigate two widely used Cas9 endonucleases, SpCas9 and SaCas9, and evaluate the relative contribution of AAV-delivery efficiency and genome-editing rates in vivo to determine the mechanisms that drive successful CRISPR-based suppression of VEGF-A, using a mouse model of laser-induced choroidal neovascularization (CNV). We found that SpCas9 demonstrated higher genome-editing rates, greater VEGF reduction, and more effective CNV suppression than SaCas9, despite similar AAV transduction efficiency between a dual-vector approach for SpCas9 and single-vector system for SaCas9 to deliver the Cas9 orthologs and single guide RNAs (gRNAs). Our results suggest that successful VEGF knockdown using AAV-mediated CRISPR systems may be determined more by the efficiency of genome editing rather than viral transduction and that SpCas9 may be more effective than SaCas9 as a potential therapeutic strategy for CRISPR-based treatment of CNV in neovascular AMD.
Collapse
Affiliation(s)
- Sook Hyun Chung
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Iris Natalie Mollhoff
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Uyen Nguyen
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Amy Nguyen
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Natalie Stucka
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Eric Tieu
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Suman Manna
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Ratheesh Kumar Meleppat
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Pengfei Zhang
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Emerald Lovece Nguyen
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Jared Fong
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Robert Zawadzki
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| | - Glenn Yiu
- Department of Ophthalmology & Vision Science, University of California, Davis, Davis, CA, USA
| |
Collapse
|
13
|
Yiu G, Chung SH, Mollhoff IN, Nguyen UT, Thomasy SM, Yoo J, Taraborelli D, Noronha G. Suprachoroidal and Subretinal Injections of AAV Using Transscleral Microneedles for Retinal Gene Delivery in Nonhuman Primates. Mol Ther Methods Clin Dev 2020; 16:179-191. [PMID: 32055646 PMCID: PMC7005511 DOI: 10.1016/j.omtm.2020.01.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 01/06/2020] [Indexed: 02/01/2023]
Abstract
Retinal gene therapy using adeno-associated viruses (AAVs) is constrained by the mode of viral vector delivery. Intravitreal AAV injections are impeded by the internal limiting membrane barrier, while subretinal injections require invasive surgery and produce a limited region of therapeutic effect. In this study, we introduce a novel mode of ocular gene delivery in rhesus macaques using transscleral microneedles to inject AAV8 into the subretinal or suprachoroidal space, a potential space between the choroid and scleral wall of the eye. Using in vivo imaging, we found that suprachoroidal AAV8 produces diffuse, peripheral expression in retinal pigment epithelial (RPE) cells, but it elicited local infiltration of inflammatory cells. Transscleral subretinal injection of AAV8 using microneedles leads to focal gene expression with transduction of RPE and photoreceptors, and minimal intraocular inflammation. In comparison, intravitreal AAV8 shows minimal transduction of retinal cells, but elicits greater systemic humoral immune responses. Our study introduces a novel mode of transscleral viral delivery that can be performed without vitreoretinal surgery, with focal or diffuse transgene expression patterns suitable for different applications. The decoupling of local and systemic immune responses reveals important insights into the immunological consequences of AAV delivery to different ocular compartments surrounding the blood-retinal barrier.
Collapse
Affiliation(s)
- Glenn Yiu
- Department of Ophthalmology & Vision Science, University of California, Davis, 4860 Y Street, Suite 2400, Sacramento, CA 95817, USA
| | - Sook Hyun Chung
- Department of Ophthalmology & Vision Science, University of California, Davis, 4860 Y Street, Suite 2400, Sacramento, CA 95817, USA
| | - Iris N. Mollhoff
- Department of Ophthalmology & Vision Science, University of California, Davis, 4860 Y Street, Suite 2400, Sacramento, CA 95817, USA
| | - Uyen Tu Nguyen
- Department of Ophthalmology & Vision Science, University of California, Davis, 4860 Y Street, Suite 2400, Sacramento, CA 95817, USA
| | - Sara M. Thomasy
- Department of Ophthalmology & Vision Science, University of California, Davis, 4860 Y Street, Suite 2400, Sacramento, CA 95817, USA
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, 1 Garrod Drive, Davis, CA 95616, USA
| | - Jesse Yoo
- Clearside Biomedical, 900 North Point Parkway, Suite 200, Alpharetta, GA 30005, USA
| | - Donna Taraborelli
- Clearside Biomedical, 900 North Point Parkway, Suite 200, Alpharetta, GA 30005, USA
| | - Glenn Noronha
- Clearside Biomedical, 900 North Point Parkway, Suite 200, Alpharetta, GA 30005, USA
| |
Collapse
|
14
|
Ghosh S, Brown AM, Jenkins C, Campbell K. Viral Vector Systems for Gene Therapy: A Comprehensive Literature Review of Progress and Biosafety Challenges. APPLIED BIOSAFETY 2020; 25:7-18. [PMID: 36033383 PMCID: PMC9134621 DOI: 10.1177/1535676019899502] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Introduction National Institutes of Health (NIH) defines gene therapy as an experimental technique that uses genes to treat or prevent disease. Although gene therapy is a promising treatment option for a number of diseases (including inherited disorders, some types of cancer, and certain viral infections), the technique remains risky and is still under study to make sure that it will be effective and safe. Methods Applications of viral vectors and nonviral gene delivery systems have found an encouraging new beginning in gene therapy in recent years. Although several viral vectors and nonviral gene delivery systems have been developed in the past 3 decades, no one delivery system can be applied in gene therapy to all cell types in vitro and in vivo. Furthermore, the use of viral vector systems (both in vitro and in vivo) present unique occupational health and safety challenges. In this review article, we discuss the biosafety challenges and the current framework of risk assessment for working with the viral vector systems. Discussion The recent advances in the field of gene therapy is exciting, but it is important for scientists, institutional biosafety committees, and biosafety officers to safeguard public trust in the use of this technology in clinical trials and make conscious efforts to engage the public through ongoing forums and discussions.
Collapse
Affiliation(s)
- Sumit Ghosh
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Alex M. Brown
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Chris Jenkins
- Clinical Biosafety Services, A Division of Sabai Global, Wildwood, MO, USA
| | - Katie Campbell
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
15
|
Day TP, Byrne LC, Flannery JG, Schaffer DV. Screening for Neutralizing Antibodies Against Natural and Engineered AAV Capsids in Nonhuman Primate Retinas. Methods Mol Biol 2018; 1715:239-249. [PMID: 29188518 DOI: 10.1007/978-1-4939-7522-8_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Adeno-associated virus (AAV) has shown promise as a therapeutic gene delivery vector for inherited retinal degenerations in both preclinical disease models and human clinical trials. The retinas of nonhuman primates (NHPs) share many anatomical similarities to humans and are an important model for evaluating AAV gene delivery. Recent evidence has shown that preexisting immunity in the form of neutralizing antibodies (NABs) in NHPs strongly correlates with weak or lack of AAV transduction in the retina when administered intravitreally, work with translational implications. This necessitates prescreening of NHPs before intravitreal delivery of AAV. In this chapter, we describe a method for screening NHP serum for preexisting NABs.
Collapse
Affiliation(s)
- Timothy P Day
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Leah C Byrne
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John G Flannery
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Vision Science Graduate Group, School of Optometry, University of California, Berkeley, CA, USA
| | - David V Schaffer
- The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA.
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
- Department of Bioengineering, University of California, Berkeley, CA, USA.
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA.
| |
Collapse
|
16
|
Notch Suppression Collaborates with Ascl1 and Lin28 to Unleash a Regenerative Response in Fish Retina, But Not in Mice. J Neurosci 2018; 38:2246-2261. [PMID: 29378863 DOI: 10.1523/jneurosci.2126-17.2018] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 01/15/2018] [Accepted: 01/19/2018] [Indexed: 11/21/2022] Open
Abstract
Müller glial (MG) cells in the zebrafish retina respond to injury by acquiring retinal stem-cell characteristics. Thousands of gene expression changes are associated with this event. Key among these changes is the induction of Ascl1a and Lin28a, two reprogramming factors whose expression is necessary for retina regeneration. Whether these factors are sufficient to drive MG proliferation and subsequent neuronal-fate specification remains unknown. To test this, we conditionally expressed Ascl1a and Lin28a in the uninjured retina of male and female fish. We found that together, their forced expression only stimulates sparse MG proliferation. However, in combination with Notch signaling inhibition, widespread MG proliferation and neuron regeneration ensued. Remarkably, Ascl1 and Lin28a expression in the retina of male and female mice also stimulated sparse MG proliferation, although this was not enhanced when combined with inhibitors of Notch signaling. Lineage tracing in both fish and mice suggested that the proliferating MG generated multipotent progenitors; however, this process was much more efficient in fish than mice. Overall, our studies suggest that the overexpression of Ascl1a and Lin28a in zebrafish, in combination with inhibition of Notch signaling, can phenocopy the effects of retinal injury in Müller glia. Interestingly, Ascl1 and Lin28a seem to have similar effects in fish and mice, whereas Notch signaling may differ. Understanding the different consequences of Notch signaling inhibition in fish and mice, may suggest additional strategies for enhancing retina regeneration in mammals.SIGNIFICANCE STATEMENT Mechanisms underlying retina regeneration in fish may suggest strategies for stimulating this process in mammals. Here we report that forced expression of Ascl1 and Lin28a can stimulate sparse MG proliferation in fish and mice; however, only in fish does Notch signaling inhibition collaborate with Ascl1a and Lin28a to stimulate widespread MG proliferation in the uninjured retina. Discerning differences in Notch signaling between fish and mice MG may reveal strategies for stimulating retina regeneration in mammals.
Collapse
|
17
|
Gaub BM, Berry MH, Visel M, Holt A, Isacoff EY, Flannery JG. Optogenetic Retinal Gene Therapy with the Light Gated GPCR Vertebrate Rhodopsin. Methods Mol Biol 2018; 1715:177-189. [PMID: 29188513 DOI: 10.1007/978-1-4939-7522-8_12] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In retinal disease, despite the loss of light sensitivity as photoreceptors die, many retinal interneurons survive in a physiologically and metabolically functional state for long periods. This provides an opportunity for treatment by genetically adding a light sensitive function to these cells. Optogenetic therapies are in development, but, to date, they have suffered from low light sensitivity and narrow dynamic response range of microbial opsins. Expression of light-sensitive G protein coupled receptors (GPCRs), such as vertebrate rhodopsin , can increase sensitivity by signal amplification , as shown by several groups. Here, we describe the methods to (1) express light gated GPCRs in retinal neurons, (2) record light responses in retinal explants in vitro, (3) record cortical light responses in vivo, and (4) test visually guided behavior in treated mice.
Collapse
Affiliation(s)
- Benjamin M Gaub
- Department of Biosystems Science and Engineering (D-BSSE), Eidgenössische Technische Hochschule (ETH) Zürich, Basel, Switzerland
| | - Michael H Berry
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
| | - Meike Visel
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
| | - Amy Holt
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
| | - Ehud Y Isacoff
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA.,Physical Bioscience Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.,Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - John G Flannery
- Vision Science Graduate Group, School of Optometry, University of California, Berkeley, CA, USA. .,The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA. .,Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| |
Collapse
|
18
|
Kuo CH, Chang BI, Lee FT, Chen PK, Lee JS, Shi GY, Wu HL. Development of Recombinant Adeno-Associated Virus Serotype 2/8 Carrying Kringle Domains of Human Plasminogen for Sustained Expression and Cancer Therapy. Hum Gene Ther 2016; 26:603-13. [PMID: 25950911 DOI: 10.1089/hum.2013.220] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Angiostatin and other plasminogen derivatives exhibit antitumor activities directly or indirectly, have demonstrated promising anticancer effects in preclinical studies, but have mostly failed in clinical trials partly due to their short serum half-lives. Our previous studies demonstrated that recombinant human plasminogen kringle 1-5 (K1-5) has superior antitumor activity compared with angiostatin. In addition, optimization of recombinant K1-5 with three amino acid substitutions enhances its antitumor effect. The current study was thus undertaken to evaluate prolonged expression of optimized K1-5 as cancer gene therapy. The recombinant adeno-associated virus (AAV) vector was used to express a secreted form of the optimized K1-5 (AAV-sK15tm) to improve its pharmacokinetic profile, which was considered to be the hurdle in angiostatin treatment of cancer. We successfully generated high-titer recombinant AAV vectors and observed sustained transgene expression for 567 days after a single injection of virus. The treated animals did not display any visible signs of abnormalities and showed normal serum biochemistry. The therapeutic potential of this treatment modality was demonstrated by both a strong inhibition of lung metastasis in the mouse B16F10 melanoma model and significant growth retardation of Lewis lung carcinoma xenografts in C57BL/6N mice as well as human A2058 melanoma xenografts in NOD/SCID (nonobese diabetic/severe combined immunodeficient) mice. Taken together, our results suggested that AAV-sK15tm produced long-term suppressive effects on cancer growth in vivo and should warrant serious consideration for clinical development.
Collapse
Affiliation(s)
- Cheng-Hsiang Kuo
- 1 Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University , Tainan, Taiwan
| | - Bi-Ing Chang
- 1 Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University , Tainan, Taiwan .,3 Cardiovascular Research Center, National Cheng Kung University , Tainan, Taiwan
| | - Fang-Tzu Lee
- 1 Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University , Tainan, Taiwan .,3 Cardiovascular Research Center, National Cheng Kung University , Tainan, Taiwan
| | - Po-Ku Chen
- 1 Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University , Tainan, Taiwan
| | - Jeng-Shin Lee
- 4 Harvard Gene Therapy Initiative, Harvard Medical School , Boston, Massachusetts
| | - Guey-Yueh Shi
- 1 Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University , Tainan, Taiwan .,2 Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University , Tainan, Taiwan .,3 Cardiovascular Research Center, National Cheng Kung University , Tainan, Taiwan
| | - Hua-Lin Wu
- 1 Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University , Tainan, Taiwan .,2 Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University , Tainan, Taiwan .,3 Cardiovascular Research Center, National Cheng Kung University , Tainan, Taiwan
| |
Collapse
|
19
|
Rosa JM, Bos R, Sack GS, Fortuny C, Agarwal A, Bergles DE, Flannery JG, Feller MB. Neuron-glia signaling in developing retina mediated by neurotransmitter spillover. eLife 2015; 4. [PMID: 26274565 PMCID: PMC4566075 DOI: 10.7554/elife.09590] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 08/13/2015] [Indexed: 12/21/2022] Open
Abstract
Neuron-glia interactions play a critical role in the maturation of neural circuits; however, little is known about the pathways that mediate their communication in the developing CNS. We investigated neuron-glia signaling in the developing retina, where we demonstrate that retinal waves reliably induce calcium transients in Müller glial cells (MCs). During cholinergic waves, MC calcium transients were blocked by muscarinic acetylcholine receptor antagonists, whereas during glutamatergic waves, MC calcium transients were inhibited by ionotropic glutamate receptor antagonists, indicating that the responsiveness of MCs changes to match the neurotransmitter used to support retinal waves. Using an optical glutamate sensor we show that the decline in MC calcium transients is caused by a reduction in the amount of glutamate reaching MCs. Together, these studies indicate that neurons and MCs exhibit correlated activity during a critical period of retinal maturation that is enabled by neurotransmitter spillover from retinal synapses.
Collapse
Affiliation(s)
- Juliana M Rosa
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Rémi Bos
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Georgeann S Sack
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Cécile Fortuny
- Vision Science Graduate Program, University of California, Berkeley, Berkeley, United States
| | - Amit Agarwal
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, United States
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, United States
| | - John G Flannery
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Marla B Feller
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
20
|
Xia CH, Liu H, Cheung D, Tang F, Chang B, Li M, Gong X. NHE8 is essential for RPE cell polarity and photoreceptor survival. Sci Rep 2015; 5:9358. [PMID: 25791178 PMCID: PMC4366848 DOI: 10.1038/srep09358] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/02/2015] [Indexed: 11/09/2022] Open
Abstract
A new N-ethyl-N-nitrosourea (ENU)-induced mouse recessive mutation, identified by fundus examination of the eye, develops depigmented patches, indicating retinal disorder. Histology data show aberrant retinal pigment epithelium (RPE) and late-onset photoreceptor cell loss in the mutant retina. Chromosomal mapping and DNA sequencing reveal a point mutation (T to A) of the Slc9a8 gene, resulting in mutant sodium/proton exchanger 8 (NHE8)-M120K protein. The lysine substitution decreases the probability of forming the 3(rd) transmembrane helix, which impairs the pore structure of the Na(+)/H(+) exchanger. Various RPE defects, including mislocalization of the apical marker ezrin, and disrupted apical microvilli and basal infoldings are observed in mutant mice. We have further generated NHE8 knockout mice and confirmed similar phenotypes, including abnormal RPE cells and late-onset photoreceptor cell loss. Both in vivo and in vitro data indicate that NHE8 co-localizes with ER, Golgi and intracellular vesicles in RPE cells. Thus, NHE8 function is necessary for the survival of photoreceptor cells and NHE8 is important for RPE cell polarity and function. Dysfunctional RPE may ultimately lead to photoreceptor cell death in the NHE8 mutants. Further studies will be needed to elucidate whether or not NHE8 regulates pH homeostasis in the protein secretory pathways of RPE.
Collapse
Affiliation(s)
- Chun-hong Xia
- School of Optometry and Vision Science Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Haiquan Liu
- School of Optometry and Vision Science Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Debra Cheung
- School of Optometry and Vision Science Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Felicia Tang
- School of Optometry and Vision Science Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Bo Chang
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Mei Li
- School of Optometry and Vision Science Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Xiaohua Gong
- School of Optometry and Vision Science Program, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
21
|
Abstract
Significant advances have been made over the last decade or two in the elucidation of the molecular pathogenesis of inherited ocular disorders. In particular, remarkable successes have been achieved in exploration of gene-based medicines for these conditions, both in preclinical and in clinical studies. Progress in the development of gene therapies targeted toward correcting the primary genetic defect or focused on modulating secondary effects associated with retinal pathologies are discussed in the review. Likewise, the recent utilization of genes encoding light-sensing molecules to provide new functions to residual retinal cells in the degenerating retina is discussed. While a great deal has been learned over the last two decades, the next decade should result in an increasing number of preclinical studies progressing to human clinical trial, an exciting prospect for patients, those active in research and development and bystanders alike.
Collapse
|
22
|
Pellissier LP, Hoek RM, Vos RM, Aartsen WM, Klimczak RR, Hoyng SA, Flannery JG, Wijnholds J. Specific tools for targeting and expression in Müller glial cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14009. [PMID: 26015954 PMCID: PMC4362388 DOI: 10.1038/mtm.2014.9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/12/2014] [Indexed: 12/17/2022]
Abstract
Despite their physiological roles, Müller glial cells are involved directly or indirectly in retinal disease pathogenesis and are an interesting target for therapeutic approaches for retinal diseases and regeneration such as CRB1 inherited retinal dystrophies. In this study, we characterized the efficiency of adeno-associated virus (AAV) capsid variants and different promoters to drive protein expression in Müller glial cells. ShH10Y and AAV9 were the most powerful capsids to infect mouse Müller glial cells. Retinaldehyde-binding protein 1 (RLBP1) promoter was the most powerful promoter to transduce Müller glial cells. ShH10Y capsids and RLBP1 promoter targeted human Müller glial cells in vitro. We also developed and tested smaller promoters to express the large CRB1 gene via AAV vectors. Minimal cytomegalovirus (CMV) promoter allowed expression of full-length CRB1 protein in Müller glial cells. In summary, ShH10Y and AAV9 capsids, and RLBP1 or minimal CMV promoters are of interest as specific tools to target and express in mouse or human Müller glial cells.
Collapse
Affiliation(s)
- Lucie P Pellissier
- Department of Neuromedical Genetics, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| | - Robert M Hoek
- Department of Neuromedical Genetics, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| | - Rogier M Vos
- Department of Neuromedical Genetics, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| | - Wendy M Aartsen
- Department of Neuromedical Genetics, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| | - Ryan R Klimczak
- Department of Molecular and Cellular Biology and The Helen Wills Neuroscience Institute, University of California , Berkeley, California, USA
| | - Stefan A Hoyng
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| | - John G Flannery
- Department of Molecular and Cellular Biology and The Helen Wills Neuroscience Institute, University of California , Berkeley, California, USA
| | - Jan Wijnholds
- Department of Neuromedical Genetics, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| |
Collapse
|
23
|
Ochoa GP, Sesma JZ, Díez MA, Díaz-Tahoces A, Avilés-Trigeros M, Grijalvo S, Eritja R, Fernández E, Pedraz JL. A novel formulation based on 2,3-di(tetradecyloxy)propan-1-amine cationic lipid combined with polysorbate 80 for efficient gene delivery to the retina. Pharm Res 2014; 31:1665-75. [PMID: 24449439 DOI: 10.1007/s11095-013-1271-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/19/2013] [Indexed: 11/28/2022]
Abstract
PURPOSE The aim of the present study was to evaluate the potential application of a novel formulation based on a synthesized cationic lipid 2,3-di(tetradecyloxy)propan-1-amine, combined with polysorbate 80 to deliver the pCMS-EGFP plasmid into the rat retina. METHODS We elaborated lipoplexes by mixing the formulation containing the cationic lipid and the polysorbate 80 with the plasmid at different cationic lipid/DNA ratios (w/w). Resulted lipoplexes were characterized in terms of size, charge, and capacity to condense, protect and release the DNA. In vitro transfection studies were performed in HEK-293 and ARPE-19 cells. Formulations were also tested in vivo by monitoring the expression of the EGFP after intravitreal and subretinal injections in rat eyes. RESULTS At 2/1 cationic lipid/DNA mass ratio, the resulted lipoplexes had 200 nm of hydrodynamic diameter; were positive charged, spherical, protected DNA against enzymatic digestion and transfected efficiently HEK-293 and ARPE-19 cultured cells exhibiting lower cytotoxicity than LipofectamineTM 2000. Subretinal administrations transfected mainly photoreceptors and retinal pigment epithelial cells; whereas intravitreal injections produced a more uniform distribution of transfection through the inner part of the retina. CONCLUSIONS These results hold great expectations for other gene delivery formulations based on this cationic lipid for retinal gene therapy purposes.
Collapse
|