1
|
Sarkis S, Chamard C, Johansen B, Daien V, Michon F. Challenging glaucoma with emerging therapies: an overview of advancements against the silent thief of sight. Front Med (Lausanne) 2025; 12:1527319. [PMID: 40206485 PMCID: PMC11979169 DOI: 10.3389/fmed.2025.1527319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Glaucoma, a leading cause of irreversible blindness, represents a significant challenge in ophthalmology. This review examines recent advancements in glaucoma treatment, focusing on innovative medications and creative strategies. While new agents offer promising methods for lowering intraocular pressure (IOP), they also pose challenges related to efficacy and side effects. Alongside IOP reduction, emerging neuroprotective approaches are being explored to safeguard retinal ganglion cells (RGCs) from glaucoma-induced damage. The review also evaluates the potential of novel drug delivery systems, such as biodegradable implants and nanoparticles, to enhance treatment effectiveness and patient adherence. Additionally, it highlights the role of personalized medicine in identifying new biomarkers and customizing therapies based on individual genetic and environmental factors.
Collapse
Affiliation(s)
- Solange Sarkis
- Institute for Neurosciences of Montpellier, Univ Montpellier, Institut national de la santé et de la recherche médicale (INSERM), Montpellier, France
- Laboratoires Théa, Clermont-Ferrand, France
| | - Chloé Chamard
- Institute for Neurosciences of Montpellier, Univ Montpellier, Institut national de la santé et de la recherche médicale (INSERM), Montpellier, France
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
| | | | - Vincent Daien
- Institute for Neurosciences of Montpellier, Univ Montpellier, Institut national de la santé et de la recherche médicale (INSERM), Montpellier, France
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
- Sydney Medical School, The Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Frederic Michon
- Institute for Neurosciences of Montpellier, Univ Montpellier, Institut national de la santé et de la recherche médicale (INSERM), Montpellier, France
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
| |
Collapse
|
2
|
Suzuki R, Katada Y, Fujii M, Serizawa N, Negishi K, Kurihara T. Tropism of the AAV6.2 Vector in the Murine Retina. Int J Mol Sci 2025; 26:1580. [PMID: 40004046 PMCID: PMC11855373 DOI: 10.3390/ijms26041580] [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: 12/10/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Retinitis pigmentosa (RP) is a progressive inherited retinal dystrophy (IRD) that primarily affects rod photoreceptor cells, leading to the degeneration of photoreceptors and the gradual loss of vision. While RP is one of the most studied IRDs, other neurodegenerative diseases affecting the retina and optic nerve, such as glaucoma, also involve common mechanisms of cellular stress and degeneration. Current therapeutic approaches under investigation include gene therapy, retina prosthesis, and neuroprotection. Among these approaches, gene therapy has shown promise, though challenges related to viral vector tropism and transduction efficiency persist. The adeno-associated virus (AAV) vector is commonly employed for gene delivery, but novel serotypes and engineered variants are being explored to improve specificity and efficacy. This study evaluates the gene transfer efficiency of the AAV6.2 vector following intravitreal injection into the murine retina. Male C57BL/6 mice (9 weeks old) were intravitreally injected with 1 µL of AAV2-CMV-EGFP, AAV6-CMV-EGFP, or AAV6.2-CMV-EGFP at a titer of 3.2 × 1012 vg/mL per eye. Retinal transduction was assessed using in vivo fluorescence imaging, flat-mount imaging, and immunohistochemistry. EGFP expression in retinal ganglion cells, Müller cells, amacrine cells, and bipolar cells was quantitatively analyzed. All three AAV serotypes effectively transduced retinal ganglion cells, but AAV6.2 exhibited enhanced transduction in Müller cells and other neuronal retinal cells, including bipolar and amacrine cells. AAV6.2 demonstrated more localized expression around retinal blood vessels compared to the diffuse expression observed with AAV2. Immunohistochemical analysis revealed that AAV6.2 had significantly higher transduction efficiency in Müller cells (p < 0.001) compared to AAV2 and AAV6. AAV6.2 shows superior transduction efficiency in Müller cells, positioning it as a promising vector for gene therapies targeting retinal degenerative diseases such as RP. Its ability to effectively transduce Müller cells suggests potential applications in neuroprotection and gene replacement therapies.
Collapse
Affiliation(s)
- Ryo Suzuki
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-0016, Japan; (R.S.); (Y.K.); (M.F.); (N.S.)
| | - Yusaku Katada
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-0016, Japan; (R.S.); (Y.K.); (M.F.); (N.S.)
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan;
| | - Momo Fujii
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-0016, Japan; (R.S.); (Y.K.); (M.F.); (N.S.)
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan;
| | - Naho Serizawa
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-0016, Japan; (R.S.); (Y.K.); (M.F.); (N.S.)
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan;
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan;
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-0016, Japan; (R.S.); (Y.K.); (M.F.); (N.S.)
| |
Collapse
|
3
|
Oikawa K, Eaton JS, Kiland JA, Torné O, Mathu V, Nickells RW, McLellan GJ. Intravitreal AAV2 gene delivery to feline retinal ganglion cells. Vision Res 2025; 226:108519. [PMID: 39549467 PMCID: PMC11658774 DOI: 10.1016/j.visres.2024.108519] [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: 07/18/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024]
Abstract
Effective strategies for the neuroprotection and preservation of retinal ganglion cells (RGCs) remain elusive in the management of glaucoma. A spontaneous genetic model of glaucoma has been identified in cats and extensively characterized as a viable translational model, with eye size and anatomy similar to humans. In this study we sought to establish initial proof of concept for gene delivery to feline RGCs via intravitreal injection of AAV2 in normal cats. Pre-retinal, posterior vitreal injection of AAV2/2-CMV-GFP, was performed overlying the area centralis in 5 adult cats. Immunosuppressive oral prednisolone was administered perioperatively and gradually tapered over 6-10wks post-injection. Ophthalmic examination was performed pre- and post-injection. The GFP reporter expression and morphological effects of viral transduction on the retina were monitored in vivo using confocal scanning laser ophthalmoscopy (cSLO) and optical coherence tomography (OCT), respectively (Spectralis OCT-HRA, Heidelberg), at 1-2wk intervals over 6-10wks. Full-field electroretinograms (ERG) and visual evoked potentials (VEP) were recorded at baseline and post-injection. Retinas were examined by histology and immunolabeling for the RGC marker RBPMS and Müller cell and astrocyte marker SOX9, and GFP expression was examined in the retina, optic nerve (ON), optic tract and lateral geniculate nucleus (LGN). GFP+ retinal cells and RGC axons were visualized by cSLO at 1-2 weeks post-injection. No retinal morphological changes were observed by OCT in vivo but 3/5 eyes exhibited mild retinal inflammation on histology. Retinal and ON function were preserved in injected eyes compared to baseline and untreated eyes. GFP expression was predominantly identified in RBPMS+ RGC cells as well as SOX9+ Müller cells. GFP fluorescence was observed throughout RGC nerve fiber tract in the central visual pathway. Peak transduction in RGCs (up to ∼ 20 %) was observed in the regions with high GFP expression, but < 1 % of RGCs expressed GFP across the whole retina. Our data provide proof of concept that pre-retinal injection of AAV2/2 may represent a feasible platform for gene delivery to feline RGCs in vivo but highlight a need for further refinement to improve RGC transduction efficiency and control low-grade retinal inflammation.
Collapse
Affiliation(s)
- Kazuya Oikawa
- Department of Ophthalmology & Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States; Department of Surgical Sciences, University of Wisconsin-Madison, Madison, WI, United States; McPherson Eye Research Institute, Madison, WI, United States
| | - J Seth Eaton
- Department of Surgical Sciences, University of Wisconsin-Madison, Madison, WI, United States; McPherson Eye Research Institute, Madison, WI, United States
| | - Julie A Kiland
- Department of Ophthalmology & Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Odalys Torné
- Department of Ophthalmology & Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States; Department of Surgical Sciences, University of Wisconsin-Madison, Madison, WI, United States; McPherson Eye Research Institute, Madison, WI, United States
| | - Virginia Mathu
- Department of Ophthalmology & Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States; Department of Surgical Sciences, University of Wisconsin-Madison, Madison, WI, United States; McPherson Eye Research Institute, Madison, WI, United States
| | - Robert W Nickells
- Department of Ophthalmology & Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States; McPherson Eye Research Institute, Madison, WI, United States
| | - Gillian J McLellan
- Department of Ophthalmology & Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States; Department of Surgical Sciences, University of Wisconsin-Madison, Madison, WI, United States; McPherson Eye Research Institute, Madison, WI, United States.
| |
Collapse
|
4
|
Siontas O, Ahn S. Challenges in AAV-Based Retinal Gene Therapies and the Role of Magnetic Nanoparticle Platforms. J Clin Med 2024; 13:7385. [PMID: 39685843 DOI: 10.3390/jcm13237385] [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: 11/06/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Retinal diseases, leading to various visual impairments and blindness, are on the rise. However, the advancement of retinal gene therapies offers new hope for treatment of such diseases. Among different vector systems for conferring therapeutic genetic load to retinal cells, adeno-associated viruses (AAVs) have been most intensively explored and have already successfully gained multiple clinical approvals. AAV-based retinal gene therapies have shown great promise in treating retinal disorders, but usually rely on the heavily disruptive administration methods such as subretinal injection. This is because the clinically well-established, minimally invasive alternative of intravitreal injection (IVI) necessitates AAVs to traverse the retinal inner limiting membrane (ILM), which is hard to penetrate in higher eye models, like human or porcine eyes. Additionally, AAVs' natural transduction preference, known as tropism, is commonly not specific to cells of only one target retinal layer, which is another ongoing challenge in retinal gene therapy. This review examines strategies to overcome these obstacles with a focus on the potential of magnetic nanoparticles (MNPs) for improved retinal AAV delivery.
Collapse
Affiliation(s)
- Oliver Siontas
- Eidgenössische Technische Hochschule (ETH) Zürich, Department of Biosystems Science and Engineering, 4056 Basel, Switzerland
| | - Seungkuk Ahn
- Eidgenössische Technische Hochschule (ETH) Zürich, Department of Biosystems Science and Engineering, 4056 Basel, Switzerland
- UCD Charles Institute of Dermatology, School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| |
Collapse
|
5
|
Ezan P, Hardy E, Bemelmans A, Taiel M, Dossi E, Rouach N. Retinal damage promotes mitochondrial transfer in the visual system of a mouse model of Leber hereditary optic neuropathy. Neurobiol Dis 2024; 201:106681. [PMID: 39332508 DOI: 10.1016/j.nbd.2024.106681] [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: 07/16/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024] Open
Abstract
Lenadogene nolparvovec is a gene therapy which has been developed to treat Leber hereditary optic neuropathy (LHON) caused by a point mutation in the mitochondrial NADH dehydrogenase 4 (ND4) gene. Clinical trials have demonstrated a significant improvement of visual acuity up to 5 years after treatment by lenadogene nolparvovec but, surprisingly, unilateral treatment resulted in bilateral improvement of vision. This contralateral effect - similarly observed with other gene therapy products in development for MT-ND4-LHON - is supported by the migration of viral vector genomes and their transcripts to the contralateral eye, as reported in animals, and post-mortem samples from two patients. In this study, we used an AAV2 encoding fluorescent proteins targeting mitochondria to investigate whether these organelles themselves could transfer from the treated eye to the fellow one. We found that mitochondria travel along the visual system (optic chiasm and primary visual cortex) and reach the contralateral eye (optic nerve and retina) in physiological conditions. We also observed that, in a rotenone-induced model of retinal damage mimicking LHON, mitochondrial transfer from the healthy to the damaged eye was accelerated and enhanced. Our results thus provide a further explanation for the contralateral beneficial effect observed during clinical studies with lenadogene nolparvovec.
Collapse
Affiliation(s)
- Pascal Ezan
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Eléonore Hardy
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Alexis Bemelmans
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | | | - Elena Dossi
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France.
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| |
Collapse
|
6
|
Gurtsieva D, Minskaia E, Zhuravleva S, Subcheva E, Sakhibgaraeva E, Brovin A, Tumaev A, Karabelsky A. Engineered AAV2.7m8 Serotype Shows Significantly Higher Transduction Efficiency of ARPE-19 and HEK293 Cell Lines Compared to AAV5, AAV8 and AAV9 Serotypes. Pharmaceutics 2024; 16:138. [PMID: 38276507 PMCID: PMC10818700 DOI: 10.3390/pharmaceutics16010138] [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: 12/13/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
The level of transduction efficiency of the target retinal cells affects the choice of AAV serotype and the outcome of gene replacement therapy for inherited retinal diseases. This study focused on the tropism and transduction efficiency of AAV2.7m8-, AAV5-, AAV8-, and AAV9-GFP in ARPE-19 and HEK293 cells. Fluorescence intensity was assessed bi-hourly by means of IncuCyte S3 live imaging microscopy. Within 12 h, AAV2.7m8 demonstrated the highest transduction efficiency at four viral concentrations of 1-, 3-, 6-, and 8 × 104 VG/cell in a dose-dependent manner, followed by AAV5 in ARPE-19 and AAV9 in HEK293 cells. The transduction efficiency of AAV2.7m8 at a dose of 6 × 104 VG/cell was 21, 202, and 323 times higher in ARPE-19 cells and 324, 100, and 52 times higher in HEK293 cells compared to AAV5, AAV8, and AAV9, respectively. This trend remained for 4 days at all viral concentrations, as additionally shown by flow cytometry. At a dose of 6 × 104 VG/cell, AAV2.7m8 (97% GFP-positive cells, GFP +) was nearly two and 10 times as efficient as AAV5 (52% GFP+) and AAV9 or AAV8 (both 9%), respectively, in ARPE-19 cells. In HEK293 cells, 95% of AAV2.7m8-, 26% of AAV9-, 17% of AAV8-, and 12% of AAV5-transduced cells were GFP-positive.
Collapse
|
7
|
Sulak R, Liu X, Smedowski A. The concept of gene therapy for glaucoma: the dream that has not come true yet. Neural Regen Res 2024; 19:92-99. [PMID: 37488850 PMCID: PMC10479832 DOI: 10.4103/1673-5374.375319] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/29/2023] [Accepted: 04/06/2023] [Indexed: 07/26/2023] Open
Abstract
Gene therapies, despite of being a relatively new therapeutic approach, have a potential to become an important alternative to current treatment strategies in glaucoma. Since glaucoma is not considered a single gene disease, the identified goals of gene therapy would be rather to provide neuroprotection of retinal ganglion cells, especially, in intraocular-pressure-independent manner. The most commonly reported type of vector for gene delivery in glaucoma studies is adeno-associated virus serotype 2 that has a high tropism to retinal ganglion cells, resulting in long-term expression and low immunogenic profile. The gene therapy studies recruit inducible and genetic animal models of optic neuropathy, like DBA/2J mice model of high-tension glaucoma and the optic nerve crush-model. Reported gene therapy-based neuroprotection of retinal ganglion cells is targeting specific genes translating to growth factors (i.e., brain derived neurotrophic factor, and its receptor TrkB), regulation of apoptosis and neurodegeneration (i.e., Bcl-xl, Xiap, FAS system, nicotinamide mononucleotide adenylyl transferase 2, Digit3 and Sarm1), immunomodulation (i.e., Crry, C3 complement), modulation of neuroinflammation (i.e., erythropoietin), reduction of excitotoxicity (i.e., CamKIIα) and transcription regulation (i.e., Max, Nrf2). On the other hand, some of gene therapy studies focus on lowering intraocular pressure, by impacting genes involved in both, decreasing aqueous humor production (i.e., aquaporin 1), and increasing outflow facility (i.e., COX2, prostaglandin F2α receptor, RhoA/RhoA kinase signaling pathway, MMP1, Myocilin). The goal of this review is to summarize the current state-of-art and the direction of development of gene therapy strategies for glaucomatous neuropathy.
Collapse
Affiliation(s)
- Robert Sulak
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Xiaonan Liu
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Adrian Smedowski
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
- GlaucoTech Co., Katowice, Poland
| |
Collapse
|
8
|
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
|
9
|
Shamshad A, Kang C, Jenny LA, Persad-Paisley EM, Tsang SH. Translatability barriers between preclinical and clinical trials of AAV gene therapy in inherited retinal diseases. Vision Res 2023; 210:108258. [PMID: 37244011 PMCID: PMC10526971 DOI: 10.1016/j.visres.2023.108258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/02/2023] [Accepted: 05/09/2023] [Indexed: 05/29/2023]
Abstract
Inherited retinal diseases (IRDs) are progressive degenerative diseases which cause gradual vision loss or complete blindness. As over 270 gene mutations have been identified in the underlying pathology of IRDs, gene therapy as a treatment modality has been an increasingly active realm of investigation. Currently, the most common vehicle of ocular gene delivery is the adeno-associated virus (AAV) vector. This is injected into the immune-privileged subretinal space to mediate transgene expression in retinal cells. Although numerous animal models of IRDs have demonstrated successful outcomes following AAV-mediated gene delivery, many of these studies fail to translate into successful outcomes in clinical trials. The purpose of this review is to A) comparatively assess preclinical and clinical IRD trials in which the success of AAV-mediated therapy failed to translate between animal and human participants B) discuss factors which may complicate the translatability of gene therapy in animals to results in humans.
Collapse
Affiliation(s)
| | - Chaerim Kang
- Warren Alpert Medical School of Brown University, USA
| | - Laura A Jenny
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA; Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia University, New York, NY, USA
| | | | - Stephen H Tsang
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA; Jonas Children's Vision Care, and Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY, USA; Department of Biomedical Engineering, Columbia University, New York, NY, USA; Columbia Stem Cell Initiative, Columbia University, New York, NY, USA; Insitute of Human Nutrition, Columbia University, New York, NY, USA
| |
Collapse
|
10
|
Hammadi S, Tzoumas N, Ferrara M, Meschede IP, Lo K, Harris C, Lako M, Steel DH. Bruch's Membrane: A Key Consideration with Complement-Based Therapies for Age-Related Macular Degeneration. J Clin Med 2023; 12:2870. [PMID: 37109207 PMCID: PMC10145879 DOI: 10.3390/jcm12082870] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
The complement system is crucial for immune surveillance, providing the body's first line of defence against pathogens. However, an imbalance in its regulators can lead to inappropriate overactivation, resulting in diseases such as age-related macular degeneration (AMD), a leading cause of irreversible blindness globally affecting around 200 million people. Complement activation in AMD is believed to begin in the choriocapillaris, but it also plays a critical role in the subretinal and retinal pigment epithelium (RPE) spaces. Bruch's membrane (BrM) acts as a barrier between the retina/RPE and choroid, hindering complement protein diffusion. This impediment increases with age and AMD, leading to compartmentalisation of complement activation. In this review, we comprehensively examine the structure and function of BrM, including its age-related changes visible through in vivo imaging, and the consequences of complement dysfunction on AMD pathogenesis. We also explore the potential and limitations of various delivery routes (systemic, intravitreal, subretinal, and suprachoroidal) for safe and effective delivery of conventional and gene therapy-based complement inhibitors to treat AMD. Further research is needed to understand the diffusion of complement proteins across BrM and optimise therapeutic delivery to the retina.
Collapse
Affiliation(s)
- Sarah Hammadi
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Nikolaos Tzoumas
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Sunderland Eye Infirmary, Queen Alexandra Rd., Sunderland SR2 9H, UK
| | | | - Ingrid Porpino Meschede
- Gyroscope Therapeutics Limited, a Novartis Company, Rolling Stock Yard, 6th Floor, 188 York Way, London N7 9AS, UK
| | - Katharina Lo
- Gyroscope Therapeutics Limited, a Novartis Company, Rolling Stock Yard, 6th Floor, 188 York Way, London N7 9AS, UK
| | - Claire Harris
- Gyroscope Therapeutics Limited, a Novartis Company, Rolling Stock Yard, 6th Floor, 188 York Way, London N7 9AS, UK
- Clinical and Translational Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Majlinda Lako
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - David H. Steel
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Sunderland Eye Infirmary, Queen Alexandra Rd., Sunderland SR2 9H, UK
| |
Collapse
|
11
|
Swirski S, May O, Ahlers M, Wissinger B, Greschner M, Jüschke C, Neidhardt J. In Vivo Efficacy and Safety Evaluations of Therapeutic Splicing Correction Using U1 snRNA in the Mouse Retina. Cells 2023; 12:cells12060955. [PMID: 36980294 PMCID: PMC10047704 DOI: 10.3390/cells12060955] [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: 01/20/2023] [Revised: 03/14/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Efficacy and safety considerations constitute essential steps during development of in vivo gene therapies. Herein, we evaluated efficacy and safety of splice factor-based treatments to correct mutation-induced splice defects in an Opa1 mutant mouse line. We applied adeno-associated viruses to the retina. The viruses transduced retinal cells with an engineered U1 snRNA splice factor designed to correct the Opa1 splice defect. We found the treatment to be efficient in increasing wild-type Opa1 transcripts. Correspondingly, Opa1 protein levels increased significantly in treated eyes. Measurements of retinal morphology and function did not reveal therapy-related side-effects supporting the short-term safety of the treatment. Alterations of potential off-target genes were not detected. Our data suggest that treatments of splice defects applying engineered U1 snRNAs represent a promising in vivo therapeutic approach. The therapy increased wild-type Opa1 transcripts and protein levels without detectable morphological, functional or genetic side-effects in the mouse eye. The U1 snRNA-based therapy can be tailored to specific disease gene mutations, hence, raising the possibility of a wider applicability of this promising technology towards treatment of different inherited retinal diseases.
Collapse
Affiliation(s)
- Sebastian Swirski
- Human Genetics, Department of Human Medicine, Faculty of Medicine and Health Sciences, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| | - Oliver May
- Human Genetics, Department of Human Medicine, Faculty of Medicine and Health Sciences, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| | - Malte Ahlers
- Visual Neuroscience, Department of Neuroscience, Faculty of Medicine and Health Sciences, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| | - Bernd Wissinger
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Elfriede-Aulhorn-Straße 7, 72076 Tübingen, Germany
| | - Martin Greschner
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Elfriede-Aulhorn-Straße 7, 72076 Tübingen, Germany
- Research Center Neurosensory Science, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| | - Christoph Jüschke
- Human Genetics, Department of Human Medicine, Faculty of Medicine and Health Sciences, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| | - John Neidhardt
- Human Genetics, Department of Human Medicine, Faculty of Medicine and Health Sciences, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
- Research Center Neurosensory Science, University of Oldenburg, Carl-von-Ossietzky-Straße 9-11, 26129 Oldenburg, Germany
| |
Collapse
|
12
|
Nieuwenhuis B, Laperrousaz E, Tribble JR, Verhaagen J, Fawcett JW, Martin KR, Williams PA, Osborne A. Improving adeno-associated viral (AAV) vector-mediated transgene expression in retinal ganglion cells: comparison of five promoters. Gene Ther 2023:10.1038/s41434-022-00380-z. [PMID: 36635457 DOI: 10.1038/s41434-022-00380-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 12/02/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023]
Abstract
Recombinant adeno-associated viral vectors (AAVs) are an effective system for gene transfer. AAV serotype 2 (AAV2) is commonly used to deliver transgenes to retinal ganglion cells (RGCs) via intravitreal injection. The AAV serotype however is not the only factor contributing to the effectiveness of gene therapies. Promoters influence the strength and cell-selectivity of transgene expression. This study compares five promoters designed to maximise AAV2 cargo space for gene delivery: chicken β-actin (CBA), cytomegalovirus (CMV), short CMV early enhancer/chicken β-actin/short β-globulin intron (sCAG), mouse phosphoglycerate kinase (PGK), and human synapsin (SYN). The promoters driving enhanced green fluorescent protein (eGFP) were examined in adult C57BL/6J mice eyes and tissues of the visual system. eGFP expression was strongest in the retina, optic nerves and brain when driven by the sCAG and SYN promoters. CBA, CMV, and PGK had moderate expression by comparison. The SYN promoter had almost exclusive transgene expression in RGCs. The PGK promoter had predominant expression in both RGCs and AII amacrine cells. The ubiquitous CBA, CMV, and sCAG promoters expressed eGFP in a variety of cell types across multiple retinal layers including Müller glia and astrocytes. We also found that these promoters could transduce human retina ex vivo, although expression was predominantly in glial cells due to low RGC viability. Taken together, this promoter comparison study contributes to optimising AAV-mediated transduction in the retina, and could be valuable for research in ocular disorders, particularly those with large or complex genetic cargos.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK. .,Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK.
| | - Elise Laperrousaz
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands.,Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - James W Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Centre of Reconstructive Neuroscience, Institute of Experimental Medicine, Prague, Czech Republic
| | - Keith R Martin
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Andrew Osborne
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK. .,Ikarovec Ltd, The Norwich Research Park Innovation Centre, Norwich, UK.
| |
Collapse
|
13
|
Nieuwenhuis B, Osborne A. Intravitreal Injection of AAV for the Transduction of Mouse Retinal Ganglion Cells. Methods Mol Biol 2023; 2708:155-174. [PMID: 37558970 DOI: 10.1007/978-1-0716-3409-7_17] [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] [Indexed: 08/11/2023]
Abstract
The injection of therapies into the eye is common practice, both clinically and pre-clinically. The most straightforward delivery route is via an intravitreal injection, which introduces the treatment into the largest cavity at the posterior of the eye. This technique is frequently used to deliver gene therapies, including those containing recombinant adeno-associated viral vectors (AAVs), to the back of the eye to enable inner retinal targeting. This chapter provides detailed methodology on how to successfully perform an intravitreal injection in mice. The chapter covers vector preparation considerations, advice on how to minimize vector loss in the injection device, and ways to reduce vector reflux from the eye when administering a therapy. Finally, a protocol is provided on common retinal histology processing techniques to assess vector-mediated expression in retinal ganglion cells. It is hoped that this chapter will enable researchers to carry out effective and consistent intravitreal injections that transduce the inner retinal surface while avoiding common pitfalls.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Andrew Osborne
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
- Ikarovec Ltd, The Norwich Research Park Innovation Centre, Norwich, UK.
| |
Collapse
|
14
|
Newman NJ, Yu-Wai-Man P, Subramanian PS, Moster ML, Wang AG, Donahue SP, Leroy BP, Carelli V, Biousse V, Vignal-Clermont C, Sergott RC, Sadun AA, Fernández GR, Chwalisz BK, Banik R, Bazin F, Roux M, Cox ED, Taiel M, Sahel JA. Randomized trial of bilateral gene therapy injection for m.11778G > A MT-ND4 Leber optic neuropathy. Brain 2022; 146:1328-1341. [PMID: 36350566 PMCID: PMC10115230 DOI: 10.1093/brain/awac421] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/20/2022] [Accepted: 10/02/2022] [Indexed: 11/11/2022] Open
Abstract
Leber hereditary optic neuropathy (LHON) is an important example of mitochondrial blindness with the m.11778G > A mutation in the MT-ND4 gene being the most common disease-causing mitochondrial DNA (mtDNA) variant worldwide. The REFLECT phase 3 pivotal study is a randomized, double-masked, placebo-controlled trial investigating the efficacy and safety of bilateral intravitreal injection of lenadogene nolparvovec in patients with a confirmed m.11778G > A mutation, using a recombinant adeno-associated virus vector 2, serotype 2 (rAAV2/2-ND4). The first-affected eye received gene therapy; the fellow (affected/not-yet-affected) eye was randomly injected with gene therapy or placebo. The primary endpoint was the difference in change from baseline of best-corrected visual acuity (BCVA) in second-affected/not-yet-affected eyes treated with lenadogene nolparvovec versus placebo at 1.5 years post-treatment, expressed in logarithm of the minimal angle of resolution (LogMAR). Forty-eight patients were treated bilaterally and 50 unilaterally. At 1.5 years, the change from baseline in BCVA was not statistically different between second-affected/not-yet-affected eyes receiving lenadogene nolparvovec and placebo (primary endpoint). A statistically significant improvement in BCVA was reported from baseline to 1.5 years in lenadogene nolparvovec-treated eyes: -0.23 LogMAR for the first-affected eyes of bilaterally treated patients (p < 0.01); and -0.15 LogMAR for second-affected/not-yet-affected eyes of bilaterally treated patients and the first-affected eyes of unilaterally treated patients (p < 0.05). The mean improvement in BCVA from nadir to 1.5 years was -0.38 (0.052) LogMAR and -0.33 (0.052) LogMAR in first-affected and second-affected/not-yet-affected eyes treated with lenadogene nolparvovec, respectively (bilateral treatment group). A mean improvement of -0.33 (0.051) LogMAR and -0.26 (0.051) LogMAR was observed in first-affected lenadogene nolparvovec-treated eyes and second-affected/not-yet-affected placebo-treated eyes, respectively (unilateral treatment group). The proportion of patients with one or both eyes on-chart at 1.5 years was 85.4% and 72.0% for bilaterally and unilaterally treated patients, respectively. The gene therapy was well tolerated, with no systemic issues. Intraocular inflammation, which was mostly mild and well controlled with topical corticosteroids, occurred in 70.7% of lenadogene nolparvovec-treated eyes versus 10.2% of placebo-treated eyes. Among eyes treated with lenadogene nolparvovec, there was no difference in the incidence of intraocular inflammation between bilaterally and unilaterally treated patients. Overall, the REFLECT trial demonstrated an improvement of BCVA in LHON eyes carrying the m.11778G > A mtDNA mutation treated with lenadogene nolparvovec or placebo to a degree not reported in natural history studies and supports an improved benefit/risk profile for bilateral injections of lenadogene nolparvovec relative to unilateral injections.
Collapse
Affiliation(s)
- Nancy J Newman
- Departments of Ophthalmology, Neurology and Neurological Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Patrick Yu-Wai-Man
- Cambridge Centre for Brain Repair and MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Eye Unit, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, UK
- Moorfields Eye Hospital, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Prem S Subramanian
- Sue Anschutz-Rodgers University of Colorado Eye Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Mark L Moster
- Departments of Neurology and Ophthalmology, Wills Eye Hospital and Thomas Jefferson University, Philadelphia, PA, USA
| | - An-Guor Wang
- Department of Ophthalmology, Taipei Veterans General Hospital, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Sean P Donahue
- Department of Ophthalmology, Neurology, and Pediatrics, Vanderbilt University, and Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bart P Leroy
- Department of Ophthalmology and Center for Medical Genetics, Ghent University Hospital, and Department of Head & Skin, Ghent University, Ghent, Belgium
| | - Valerio Carelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
- Unit of Neurology, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Valerie Biousse
- Departments of Ophthalmology, Neurology and Neurological Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Catherine Vignal-Clermont
- Department of Neuro Ophthalmology and Emergencies, Rothschild Foundation Hospital, Paris, France
- Centre Hospitalier National D'Ophtalmologie des Quinze Vingts, Paris, France
| | - Robert C Sergott
- Departments of Neurology and Ophthalmology, Wills Eye Hospital and Thomas Jefferson University, Philadelphia, PA, USA
| | - Alfredo A Sadun
- Doheny Eye Institute, UCLA School of Medicine, Los Angeles, CA, USA
| | | | - Bart K Chwalisz
- Department of Ophthalmology, Massachusetts Eye & Ear, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston MA, USA
| | - Rudrani Banik
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Fondation Ophtalmologique A. de Rothschild, Paris, France
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- CHNO des Quinze-Vingts, Institut Hospitalo-Universitaire FOReSIGHT, INSERM-DGOS CIC, Paris, France
| | | |
Collapse
|
15
|
Patel DD, Marsic D, Periasamy R, Zolotukhin S, Lipinski DM. Identification of Novel Retinal Pericyte-Targeting rAAV Vectors Through Directed Evolution. Transl Vis Sci Technol 2022; 11:28. [PMID: 36018583 PMCID: PMC9428359 DOI: 10.1167/tvst.11.8.28] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Retinal pericytes play a vital role in maintaining retinal homeostasis, and their dysfunction underlies pathogenesis in such vascular eye diseases as diabetic retinopathy and wet age-related macular degeneration. Consequently, retinal pericytes are attractive therapeutic targets for gene therapy, but effectively targeting pericytes with recombinant adeno-associated virus (rAAV) vectors remains a challenge. Methods We introduced genetic modifications into the surface-exposed variable regions of the rAAV2/2 capsid to generate a complex library (>1 × 107) of capsid mutants that were then screened for preferential tropism toward retinal pericytes. Using the Tg(Cspg4-DsRed.T1)1Akik/J reporter mouse model, which has red fluorescent pericytes that can be isolated via flow cytometry in order to recover vector genomes, we performed three rounds of screening and identified seven putative mutants capable of transducing retinal pericytes. Results Following intravitreal administration of mutant vectors packaging ubiquitously expressing green fluorescent protein reporters and postmortem flow cytometry of enzymatically digested retinae, two mutants in particular, Peri-E and Peri-G, demonstrated significantly greater transduction of retinal pericytes than unmodified rAAV2/2 (1.4-fold and 2.8-fold, respectively). Conclusions Although difficult to characterize the effect of each point mutation in the context of multiple amino acid variations from the wild-type AAV2 sequence, we identified several point mutations that may play critical roles in limiting HSPG binding, evading neutralization by murine A20 monoclonal antibodies, modulating antigenicity, and evading ubiquitination to ultimately improve transduction efficiency of retinal pericytes. Translational Relevance Identification of novel retinal pericyte targeting rAAV vectors enables the development of new, long-lasting gene therapies for retinal diseases such as diabetic retinopathy and wet age-related macular degeneration.
Collapse
Affiliation(s)
- Dwani D Patel
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Ophthalmology and Visual Science, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Damien Marsic
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA.,Porton Advanced Solutions, Suzhou, Jiangsu, China
| | - Ramesh Periasamy
- Department of Ophthalmology and Visual Science, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sergei Zolotukhin
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Daniel M Lipinski
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Ophthalmology and Visual Science, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
16
|
Chen BS, Yu-Wai-Man P. From Bench to Bedside-Delivering Gene Therapy for Leber Hereditary Optic Neuropathy. Cold Spring Harb Perspect Med 2022; 12:a041282. [PMID: 35863905 PMCID: PMC9310952 DOI: 10.1101/cshperspect.a041282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Leber hereditary optic neuropathy (LHON) is a rare, maternally inherited mitochondrial disorder that presents with severe bilateral sequential vision loss, due to the selective degeneration of retinal ganglion cells (RGCs). Since the mitochondrial genetic basis for LHON was uncovered in 1988, considerable progress has been made in understanding the pathogenetic mechanisms driving RGC loss, which has enabled the development of therapeutic approaches aimed at mitigating the underlying mitochondrial dysfunction. In this review, we explore the genetics of LHON, from bench to bedside, focusing on the pathogenetic mechanisms and how these have informed the development of different gene therapy approaches, in particular the technique of allotopic expression with adeno-associated viral vectors. Finally, we provide an overview of the recent gene therapy clinical trials and consider the unanswered questions, challenges, and future prospects.
Collapse
Affiliation(s)
- Benson S Chen
- John van Geest Centre for Brain Repair and MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, United Kingdom
- Cambridge Eye Unit, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge CB2 0QQ, United Kingdom
| | - Patrick Yu-Wai-Man
- John van Geest Centre for Brain Repair and MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, United Kingdom
- Cambridge Eye Unit, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge CB2 0QQ, United Kingdom
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, United Kingdom
- Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| |
Collapse
|
17
|
Rudnick ND, Kim LA, Comander J. Adeno-associated Viral Vectors in the Retina: Delivering Gene Therapy to the Right Destination. Int Ophthalmol Clin 2022; 62:215-229. [PMID: 35325920 DOI: 10.1097/iio.0000000000000416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
|
18
|
Cwerman-Thibault H, Lechauve C, Malko-Baverel V, Augustin S, Le Guilloux G, Reboussin É, Degardin-Chicaud J, Simonutti M, Debeir T, Corral-Debrinski M. Neuroglobin effectively halts vision loss in Harlequin mice at an advanced stage of optic nerve degeneration. Neurobiol Dis 2021; 159:105483. [PMID: 34400304 DOI: 10.1016/j.nbd.2021.105483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/06/2021] [Accepted: 08/11/2021] [Indexed: 10/20/2022] Open
Abstract
Mitochondrial diseases are among the most prevalent groups of inherited neurological disorders, affecting up to 1 in 5000 adults. Despite the progress achieved on the identification of gene mutations causing mitochondrial pathologies, they cannot be cured so far. Harlequin mice, a relevant model of mitochondrial pathology due to apoptosis inducing factor depletion, suffer from progressive disappearance of retinal ganglion cells leading to optic neuropathy. In our previous work, we showed that administering adeno-associated virus encompassing the coding sequences for neuroglobin, (a neuroprotective molecule belonging to the globin family) or apoptosis-inducing factor, before neurodegeneration onset, prevented retinal ganglion cell loss and preserved visual function. One of the challenges to develop an effective treatment for optic neuropathies is to consider that by the time patients become aware of their handicap, a large amount of nerve fibers has already disappeared. Gene therapy was performed in Harlequin mice aged between 4 and 5 months with either a neuroglobin or an apoptosis-inducing factor vector to determine whether the increased abundance of either one of these proteins in retinas could preserve visual function at this advanced stage of the disease. We demonstrated that gene therapy, by preserving the connectivity of transduced retinal ganglion cells and optic nerve bioenergetics, results in the enhancement of visual cortex activity, ultimately rescuing visual impairment. This study demonstrates that: (a) An increased abundance of neuroglobin functionally overcomes apoptosis-inducing factor absence in Harlequin mouse retinas at a late stage of neuronal degeneration; (b) The beneficial effect for visual function could be mediated by neuroglobin localization to the mitochondria, thus contributing to the maintenance of the organelle homeostasis.
Collapse
Affiliation(s)
| | - Christophe Lechauve
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012 Paris, France
| | | | - Sébastien Augustin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012 Paris, France
| | | | - Élodie Reboussin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012 Paris, France
| | | | - Manuel Simonutti
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, F-75012 Paris, France
| | | | | |
Collapse
|
19
|
Fuller-Carter PI, Basiri H, Harvey AR, Carvalho LS. Focused Update on AAV-Based Gene Therapy Clinical Trials for Inherited Retinal Degeneration. BioDrugs 2021; 34:763-781. [PMID: 33136237 DOI: 10.1007/s40259-020-00453-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inherited retinal diseases (IRDs) comprise a clinically and genetically heterogeneous group of disorders that can ultimately result in photoreceptor dysfunction/death and vision loss. With over 270 genes known to be involved in IRDs, translation of treatment strategies into clinical applications has been historically difficult. However, in recent years there have been significant advances in basic research findings as well as translational studies, culminating in an increasing number of clinical trials with the ultimate goal of reducing vision loss and associated morbidities. The recent approval of Luxturna® (voretigene neparvovec-rzyl) for Leber congenital amaurosis type 2 (LCA2) prompts a review of the current clinical trials for IRDs, with a particular focus on the importance of adeno-associated virus (AAV)-based gene therapies. The present article reviews the current state of AAV use in gene therapy clinical trials for IRDs, with a brief background on AAV and the reasons behind its dominance in ocular gene therapy. It will also discuss pre-clinical progress in AAV-based therapies aimed at treating other ocular conditions that can have hereditable links, and what alternative technologies are progressing in the same therapeutic space.
Collapse
Affiliation(s)
- Paula I Fuller-Carter
- Centre for Ophthalmology and Visual Sciences (Incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| | - Hamed Basiri
- Centre for Ophthalmology and Visual Sciences (Incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia
| | - Alan R Harvey
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Livia S Carvalho
- Centre for Ophthalmology and Visual Sciences (Incorporating Lions Eye Institute), The University of Western Australia, Nedlands, WA, Australia.
| |
Collapse
|
20
|
Wei Z, Liu X, Li T, Li X, Zhou Q, Wu J, Zhang C. Transduction of mouse retina by insect cell packaged recombinant adeno-associated viruses and their mutants via intravitreal injection. Future Virol 2021. [DOI: 10.2217/fvl-2020-0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: Adeno-associated virus (AAV) is the most preferred gene therapy vector. The purpose of our research is to compare the infection tropism and gene expression efficiency of vitreous injection of recombinant AAVs (rAAVs) and their capsid mutants in mouse retina. Materials & methods: We packaged wild-type rAAV2/2,6,8,9 and their capsid mutants carrying EGFP expression cassette using insect cells. The gene expression profiles of rAAVs and their mutants in mouse retina were evaluated by optical imaging of retinal tissue flat mount and cryosections. Results & conclusion: The results showed that rAAV2 and rAAV2-Y444F mainly targeted retinal ganglion cell; rAAV8, rAAV8-Y733F, rAAV9 and mutants had obvious EGFP expression in retinal pigment epithelium cells. Compared with the wild-type rAAVs, capsid mutants have an improved transduction efficiency in mouse retina cells.
Collapse
Affiliation(s)
- Zheng Wei
- Suzhou Institute of Biomedical Engineering & Technology, Chinese Academy of Sciences, Suzhou, China
- School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaomei Liu
- Suzhou Institute of Biomedical Engineering & Technology, Chinese Academy of Sciences, Suzhou, China
| | - Taiming Li
- School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaofang Li
- Suzhou Institute of Biomedical Engineering & Technology, Chinese Academy of Sciences, Suzhou, China
- School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Qungang Zhou
- Department of Blood Collection, Suzhou Red Cross Blood Center, Suzhou, China
| | - Jianxiang Wu
- Department of Blood Collection, Suzhou Red Cross Blood Center, Suzhou, China
| | - Chun Zhang
- Suzhou Institute of Biomedical Engineering & Technology, Chinese Academy of Sciences, Suzhou, China
| |
Collapse
|
21
|
Komáromy AM, Koehl KL, Park SA. Looking into the future: Gene and cell therapies for glaucoma. Vet Ophthalmol 2021; 24 Suppl 1:16-33. [PMID: 33411993 PMCID: PMC7979454 DOI: 10.1111/vop.12858] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022]
Abstract
Glaucoma is a complex group of optic neuropathies that affects both humans and animals. Intraocular pressure (IOP) elevation is a major risk factor that results in the loss of retinal ganglion cells (RGCs) and their axons. Currently, lowering IOP by medical and surgical methods is the only approved treatment for primary glaucoma, but there is no cure, and vision loss often progresses despite therapy. Recent technologic advances provide us with a better understanding of disease mechanisms and risk factors; this will permit earlier diagnosis of glaucoma and initiation of therapy sooner and more effectively. Gene and cell therapies are well suited to target these mechanisms specifically with the potential to achieve a lasting therapeutic effect. Much progress has been made in laboratory settings to develop these novel therapies for the eye. Gene and cell therapies have already been translated into clinical application for some inherited retinal dystrophies and age-related macular degeneration (AMD). Except for the intravitreal application of ciliary neurotrophic factor (CNTF) by encapsulated cell technology for RGC neuroprotection, there has been no other clinical translation of gene and cell therapies for glaucoma so far. Possible application of gene and cell therapies consists of long-term IOP control via increased aqueous humor drainage, including inhibition of fibrosis following filtration surgery, RGC neuroprotection and neuroregeneration, modification of ocular biomechanics for improved IOP tolerance, and inhibition of inflammation and neovascularization to prevent the development of some forms of secondary glaucoma.
Collapse
Affiliation(s)
- András M. Komáromy
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Kristin L. Koehl
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Shin Ae Park
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
- College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
22
|
Koponen S, Kokki E, Kinnunen K, Ylä-Herttuala S. Viral-Vector-Delivered Anti-Angiogenic Therapies to the Eye. Pharmaceutics 2021; 13:pharmaceutics13020219. [PMID: 33562561 PMCID: PMC7915489 DOI: 10.3390/pharmaceutics13020219] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/17/2022] Open
Abstract
Pathological vessel growth harms vision and may finally lead to vision loss. Anti-angiogenic gene therapy with viral vectors for ocular neovascularization has shown great promise in preclinical studies. Most of the studies have been conducted with different adeno-associated serotype vectors. In addition, adeno- and lentivirus vectors have been used. Therapy has been targeted towards blocking vascular endothelial growth factors or other pro-angiogenic factors. Clinical trials of intraocular gene therapy for neovascularization have shown the treatment to be safe without severe adverse events or systemic effects. Nevertheless, clinical studies have not proceeded further than Phase 2 trials.
Collapse
Affiliation(s)
- Sanna Koponen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland; (S.K.); (E.K.)
| | - Emmi Kokki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland; (S.K.); (E.K.)
| | - Kati Kinnunen
- Department of Ophthalmology, Kuopio University Hospital, 70211 Kuopio, Finland;
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland; (S.K.); (E.K.)
- Gene Therapy Unit, Kuopio University Hospital, 70211 Kuopio, Finland
- Correspondence: ; Tel./Fax: +358-403-552-075
| |
Collapse
|
23
|
O'Carroll SJ, Cook WH, Young D. AAV Targeting of Glial Cell Types in the Central and Peripheral Nervous System and Relevance to Human Gene Therapy. Front Mol Neurosci 2021; 13:618020. [PMID: 33505247 PMCID: PMC7829478 DOI: 10.3389/fnmol.2020.618020] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Different glial cell types are found throughout the central (CNS) and peripheral nervous system (PNS), where they have important functions. These cell types are also involved in nervous system pathology, playing roles in neurodegenerative disease and following trauma in the brain and spinal cord (astrocytes, microglia, oligodendrocytes), nerve degeneration and development of pain in peripheral nerves (Schwann cells, satellite cells), retinal diseases (Müller glia) and gut dysbiosis (enteric glia). These cell type have all been proposed as potential targets for treating these conditions. One approach to target these cell types is the use of gene therapy to modify gene expression. Adeno-associated virus (AAV) vectors have been shown to be safe and effective in targeting cells in the nervous system and have been used in a number of clinical trials. To date, a number of studies have tested the use of different AAV serotypes and cell-specific promoters to increase glial cell tropism and expression. However, true glial-cell specific targeting for a particular glial cell type remains elusive. This review provides an overview of research into developing glial specific gene therapy and discusses some of the issues that still need to be addressed to make glial cell gene therapy a clinical reality.
Collapse
Affiliation(s)
- Simon J O'Carroll
- Spinal Cord Injury Research Group, Department of Anatomy and Medical Imaging, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - William H Cook
- Molecular Neurotherapeutics Group, Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Deborah Young
- Molecular Neurotherapeutics Group, Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
24
|
Wang Y, Bruggeman KF, Franks S, Gautam V, Hodgetts SI, Harvey AR, Williams RJ, Nisbet DR. Is Viral Vector Gene Delivery More Effective Using Biomaterials? Adv Healthc Mater 2021; 10:e2001238. [PMID: 33191667 DOI: 10.1002/adhm.202001238] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/03/2020] [Indexed: 12/16/2022]
Abstract
Gene delivery has been extensively investigated for introducing foreign genetic material into cells to promote expression of therapeutic proteins or to silence relevant genes. This approach can regulate genetic or epigenetic disorders, offering an attractive alternative to pharmacological therapy or invasive protein delivery options. However, the exciting potential of viral gene therapy has yet to be fully realized, with a number of clinical trials failing to deliver optimal therapeutic outcomes. Reasons for this include difficulty in achieving localized delivery, and subsequently lower efficacy at the target site, as well as poor or inconsistent transduction efficiency. Thus, ongoing efforts are focused on improving local viral delivery and enhancing its efficiency. Recently, biomaterials have been exploited as an option for more controlled, targeted and programmable gene delivery. There is a growing body of literature demonstrating the efficacy of biomaterials and their potential advantages over other delivery strategies. This review explores current limitations of gene delivery and the progress of biomaterial-mediated gene delivery. The combination of biomaterials and gene vectors holds the potential to surmount major challenges, including the uncontrolled release of viral vectors with random delivery duration, poorly localized viral delivery with associated off-target effects, limited viral tropism, and immune safety concerns.
Collapse
Affiliation(s)
- Yi Wang
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Kiara F. Bruggeman
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Stephanie Franks
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Vini Gautam
- Department of Biomedical Engineering The University of Melbourne Melbourne Victoria 3010 Australia
| | - Stuart I. Hodgetts
- School of Human Sciences The University of Western Australia Perth WA 6009 Australia
- Perron Institute for Neurological and Translational Science Perth WA 6009 Australia
| | - Alan R. Harvey
- School of Human Sciences The University of Western Australia Perth WA 6009 Australia
- Perron Institute for Neurological and Translational Science Perth WA 6009 Australia
| | - Richard J. Williams
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT) School of Medicine Deakin University Waurn Ponds VIC 3216 Australia
- Biofab3D St. Vincent's Hospital Fitzroy 3065 Australia
| | - David R. Nisbet
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
- Biofab3D St. Vincent's Hospital Fitzroy 3065 Australia
| |
Collapse
|
25
|
Ocular delivery of CRISPR/Cas genome editing components for treatment of eye diseases. Adv Drug Deliv Rev 2021; 168:181-195. [PMID: 32603815 DOI: 10.1016/j.addr.2020.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 06/02/2020] [Accepted: 06/12/2020] [Indexed: 12/26/2022]
Abstract
A variety of inherited or multifactorial ocular diseases call for novel treatment paradigms. The newly developed genome editing technology, CRISPR, has shown great promise in treating these diseases, but delivery of the CRISPR/Cas components to target ocular tissues and cells requires appropriate use of vectors and routes of administration to ensure safety, efficacy and specificity. Although adeno-associated viral (AAV) vectors are thus far the most commonly used tool for ocular gene delivery, sustained expression of CRISPR/Cas components may cause immune reactions and an increased risk of off-target editing. In this review, we summarize the ocular administration routes and discuss the advantages and disadvantages of viral and non-viral vectors for delivery of CRISPR/Cas components to the eye. We review the existing studies of CRISPR/Cas genome editing for ocular diseases and discuss the major challenges of the technology in ocular applications. We also discuss the most recently developed CRISPR tools such as base editing and prime editing which may be used for future ocular applications.
Collapse
|
26
|
Millington-Ward S, Chadderton N, Berkeley M, Finnegan LK, Hanlon KS, Carrigan M, Humphries P, Kenna PF, Palfi A, Farrar GJ. Novel 199 base pair NEFH promoter drives expression in retinal ganglion cells. Sci Rep 2020; 10:16515. [PMID: 33020509 PMCID: PMC7536420 DOI: 10.1038/s41598-020-73257-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 09/08/2020] [Indexed: 01/13/2023] Open
Abstract
Retinal ganglion cells (RGCs) are known to be involved in several ocular disorders, including glaucoma and Leber hereditary optic neuropathy (LHON), and hence represent target cells for gene therapies directed towards these diseases. Restricting gene therapeutics to the target cell type in many situations may be preferable compared to ubiquitous transgene expression, stimulating researchers to identify RGC-specific promoters, particularly promoter sequences that may also be appropriate in size to fit readily into recombinant adeno associated viral (AAV) vectors, the vector of choice for many ocular gene therapies. In the current study we analysed EGFP expression driven by various sequences of the putative human NEFH promoter in order to define sequences required for preferential expression in RGCs. EGFP expression profiles from four different potential NEFH promoter constructs were compared in vivo in mice using retinal histology and mRNA expression analysis. Notably, two efficient promoter sequences, one comprising just 199 bp, are presented in the study.
Collapse
Affiliation(s)
| | - Naomi Chadderton
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
| | - Megan Berkeley
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
| | - Laura K Finnegan
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
| | - Killian S Hanlon
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
| | - Matthew Carrigan
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
| | - Peter Humphries
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
| | - Paul F Kenna
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland.,The Research Foundation, Royal Victoria Eye and Ear Hospital, Dublin 2, Ireland
| | - Arpad Palfi
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
| | - G Jane Farrar
- The School of Genetics and Microbiology, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
27
|
Andrews MR. Gene therapy in the CNS-one size does not fit all. Gene Ther 2020; 28:393-395. [PMID: 32978509 DOI: 10.1038/s41434-020-00196-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/25/2020] [Accepted: 09/18/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Melissa R Andrews
- School of Biological Sciences, University of Southampton, Southampton, SO171BJ, UK.
| |
Collapse
|
28
|
Song H, Zeng Y, Sardar Pasha SPB, Bush RA, Vijayasarathy C, Qian H, Wei L, Wiley HE, Wu Z, Sieving PA. Trans-Ocular Electric Current In Vivo Enhances AAV-Mediated Retinal Transduction in Large Animal Eye After Intravitreal Vector Administration. Transl Vis Sci Technol 2020; 9:28. [PMID: 32844051 PMCID: PMC7416894 DOI: 10.1167/tvst.9.7.28] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/06/2020] [Indexed: 01/25/2023] Open
Abstract
Purpose Electric micro-current has been shown to enhance penetration and transduction of adeno-associated viral (AAV) vectors in mouse retina after intravitreal administration. We termed this: “electric-current vector mobility (ECVM).” The present study considered whether ECVM could augment retinal transduction efficiency of intravitreal AAV8-CMV-EGFP in normal rabbit and nonhuman primate (NHP) macaque. Potential mechanisms underlying enhanced retinal transduction by ECVM were also studied. Methods We applied an electric micro-current across the intact eye of normal rabbit and monkey in vivo for a brief period immediately after intravitreal injection of AAV8-CMV-EGFP. Retinal GFP expression was evaluated by fundus imaging in vivo. Retinal immunohistochemistry was performed to assess the distribution of retinal cells transduced by the AAV8-EGFP. Basic fibroblast growth factor (bFGF) was analyzed by quantitative RT-polymerase chain reaction (PCR). Müller glial reactivity and inner limiting membrane (ILM) were examined by the glial fibrillary acidic protein (GFAP) and vimentin staining in mouse retina, respectively. Results ECVM significantly increased the efficiency of AAV reaching and transducing the rabbit retina following intravitreal injection, with gene expression in inner nuclear layer, ganglion cells, and Müller cells. Similar trend of improvement was observed in the ECVM-treated monkey eye. The electric micro-current upregulated bFGF expression in Müller cells and vimentin showed ILM structural changes in mouse retina. Conclusions ECVM promotes the transduction efficiency of AAV8-CMV-GFP in normal rabbit and monkey retinas following intravitreal injection. Translational Relevance This work has potential translational relevance to human ocular gene therapy by increasing retinal expression of therapeutic vectors given by intravitreal administration.
Collapse
Affiliation(s)
- Hongman Song
- Section for Translational Research on Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, USA.,National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yong Zeng
- Section for Translational Research on Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, USA.,National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Ronald A Bush
- Section for Translational Research on Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, USA.,National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Camasamudram Vijayasarathy
- Section for Translational Research on Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, USA.,National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Haohua Qian
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa Wei
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Henry E Wiley
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhijian Wu
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul A Sieving
- Section for Translational Research on Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, Bethesda, MD, USA.,National Eye Institute, National Institutes of Health, Bethesda, MD, USA.,Department of Ophthalmology, School of Medicine, University of California at Davis, Sacramento, CA, USA
| |
Collapse
|
29
|
α-synuclein overexpression in the retina leads to vision impairment and degeneration of dopaminergic amacrine cells. Sci Rep 2020; 10:9619. [PMID: 32541823 PMCID: PMC7295803 DOI: 10.1038/s41598-020-66497-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 04/29/2020] [Indexed: 11/08/2022] Open
Abstract
The presence of α-synuclein aggregates in the retina of Parkinson's disease patients has been associated with vision impairment. In this study we sought to determine the effects of α-synuclein overexpression on the survival and function of dopaminergic amacrine cells (DACs) in the retina. Adult mice were intravitreally injected with an adeno-associated viral (AAV) vector to overexpress human wild-type α-synuclein in the inner retina. Before and after systemic injections of levodopa (L-DOPA), retinal responses and visual acuity-driven behavior were measured by electroretinography (ERG) and a water maze task, respectively. Amacrine cells and ganglion cells were counted at different time points after the injection. α-synuclein overexpression led to an early loss of DACs associated with a decrease of light-adapted ERG responses and visual acuity that could be rescued by systemic injections of L-DOPA. The data show that α-synuclein overexpression affects dopamine neurons in the retina. The approach provides a novel accessible method to model the underlying mechanisms implicated in the pathogenesis of synucleinopathies and for testing novel treatments.
Collapse
|
30
|
Madrakhimov SB, Yang JY, Ahn DH, Han JW, Ha TH, Park TK. Peripapillary Intravitreal Injection Improves AAV-Mediated Retinal Transduction. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:647-656. [PMID: 32300611 PMCID: PMC7152690 DOI: 10.1016/j.omtm.2020.03.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/23/2020] [Indexed: 12/31/2022]
Abstract
The intravitreal (IVT) injection method is a choice when targeting the inner retina for gene therapy. However, the transduction efficiency of adeno-associated virus (AAV) vectors administered by the IVT route is usually low and may be affected by several factors. To improve the transduction efficiency, we developed a novel illuminated long-needle attached injection system and injected AAV2-CMV (cytomegalovirus)-EGFP in front of the retina in rabbit eyes. Ophthalmological examinations were performed and the levels of pro-inflammatory cytokines in the aqueous humor were assessed at the baseline and 1 month, and the results were compared with those of the conventional injection method. Retinal tissues were used for immunohistochemistry. In the ophthalmological examinations, no significant inflammatory signs were detected in both groups, except for transient, mild hyperemia. In the tissues of the rabbits in the peripapillary injection group, significantly increased GFP expression was detected at the ganglion cell and the inner nuclear layers (p < 0.01). There were no differences between groups in glial activation and expressions of interleukin (IL)-6 and IL-8. These results suggest that peripapillary IVT injection in front of the retina would be safe and efficiently transduce viral vectors into the retina of large animals and is considered as a potential method for use in clinical trials.
Collapse
Affiliation(s)
- Sanjar Batirovich Madrakhimov
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research on Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Jin Young Yang
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research on Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Dong Hyuck Ahn
- Laboratory for Translational Research on Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Jung Woo Han
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Tae Ho Ha
- CMLAB, Convergence Technologies for Bio-Medical Science, Seoul, South Korea
| | - Tae Kwann Park
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research on Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Cheonan, Choongchungnam-do, South Korea
- Ex Lumina Therapeutics and Technologies, Bucheon, South Korea
- Corresponding author: Tae Kwann Park, MD, PhD, Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, #170, Jomaru-ro, Wonmi-gu, Bucheon 14584, South Korea.
| |
Collapse
|
31
|
Guimaraes TACD, Georgiou M, Robson AG, Michaelides M. KCNV2 retinopathy: clinical features, molecular genetics and directions for future therapy. Ophthalmic Genet 2020; 41:208-215. [PMID: 32441199 PMCID: PMC7446039 DOI: 10.1080/13816810.2020.1766087] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
-associated retinopathy or “cone dystrophy with supernormal rod responses” is an
autosomal recessive cone-rod dystrophy with pathognomonic ERG findings. This gene
encodes Kv8.2, a voltage-gated potassium channel subunit that acts as a modulator by
shifting the activation range of the K+ channels in photoreceptor inner
segments. Currently, no treatment is available for the condition. However, there is a
lack of prospective long-term data in large molecularly confirmed cohorts, which is a
prerequisite for accurate patient counselling/prognostication, to identify an optimal
window for intervention and outcome measures, and ultimately to design future therapy
trials. Herein we provide a detailed review of the clinical features, retinal imaging,
electrophysiology and psychophysical studies, molecular genetics, and briefly discuss
future prospects for therapy trials.
Collapse
Affiliation(s)
- Thales A C De Guimaraes
- UCL Institute of Ophthalmology, University College London , London, UK.,Moorfields Eye Hospital , London, UK
| | - Michalis Georgiou
- UCL Institute of Ophthalmology, University College London , London, UK.,Moorfields Eye Hospital , London, UK
| | - Anthony G Robson
- UCL Institute of Ophthalmology, University College London , London, UK.,Moorfields Eye Hospital , London, UK
| | - Michel Michaelides
- UCL Institute of Ophthalmology, University College London , London, UK.,Moorfields Eye Hospital , London, UK
| |
Collapse
|
32
|
Guimaraes TACD, Georgiou M, Bainbridge JWB, Michaelides M. Gene therapy for neovascular age-related macular degeneration: rationale, clinical trials and future directions. Br J Ophthalmol 2020; 105:151-157. [PMID: 32269060 PMCID: PMC7848059 DOI: 10.1136/bjophthalmol-2020-316195] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 01/19/2023]
Abstract
Age-related macular degeneration (AMD) is one of the leading causes of irreversible blindness in the developed world. Antivascular endothelial growth factor therapy has transformed the management and outcome of neovascular AMD (nAMD), although the need for repeated intravitreal injections—even lifelong—and the related complications, high drug costs, frequent clinic visits and repeated imaging have resulted in an enormous burden both to healthcare systems and patients. The application of gene therapy approaches for sustained delivery of a range of antiangiogenic proteins has the promise of helping to address these aforementioned challenges. A number of early phase clinical trials of gene therapy in nAMD have provided encouraging results, with many more ongoing or anticipated. There remain significant areas of controversy, including regarding the optimal treatment targets, routes of administration and potential safety concerns. In this review we aim to provide an update of the current status of gene therapy for nAMD and briefly discuss future prospects.
Collapse
Affiliation(s)
| | - Michalis Georgiou
- UCL Institute of Ophthalmology, University College London, London, UK.,Moorfields Eye Hospital, London, UK
| | - James W B Bainbridge
- UCL Institute of Ophthalmology, University College London, London, UK.,Moorfields Eye Hospital, London, UK
| | - Michel Michaelides
- UCL Institute of Ophthalmology, University College London, London, UK .,Moorfields Eye Hospital, London, UK
| |
Collapse
|
33
|
Reese H, Bordelon T, Odeh F, Broussard A, Kormos C, Murphy A, Shanahan C, Menegatti S. Purification of animal immunoglobulin G (IgG) using peptoid affinity ligands. Biotechnol Prog 2020; 36:e2994. [DOI: 10.1002/btpr.2994] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/07/2020] [Accepted: 03/02/2020] [Indexed: 01/19/2023]
Affiliation(s)
- Hannah Reese
- Department of Chemical and Biomolecular EngineeringNorth Carolina State University Raleigh North Carolina USA
| | | | - Fuad Odeh
- LigaTrap LLC Raleigh North Carolina USA
| | | | | | | | - Calvin Shanahan
- Department of Chemical and Biomolecular EngineeringNorth Carolina State University Raleigh North Carolina USA
| | - Stefano Menegatti
- Department of Chemical and Biomolecular EngineeringNorth Carolina State University Raleigh North Carolina USA
- Biomanufacturing Training and Education Center (BTEC)North Carolina State University Raleigh North Carolina USA
| |
Collapse
|
34
|
Touahri Y, Dixit R, Kofoed RH, Mikloska K, Park E, Raeisossadati R, Markham-Coultes K, David LA, Rijal H, Zhao J, Lynch M, Hynynen K, Aubert I, Schuurmans C. Focused ultrasound as a novel strategy for noninvasive gene delivery to retinal Müller glia. Theranostics 2020; 10:2982-2999. [PMID: 32194850 PMCID: PMC7053200 DOI: 10.7150/thno.42611] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
Müller glia are specialized retinal cells with stem cell properties in fish and frogs but not in mammals. Current efforts to develop gene therapies to activate mammalian Müller glia for retinal repair will require safe and effective delivery strategies for recombinant adeno-associated viruses (AAVs), vectors of choice for clinical translation. Intravitreal and subretinal injections are currently used for AAV gene delivery in the eye, but less invasive methods efficiently targeting Müller glia have yet to be developed. Methods: As gene delivery strategies have been more extensively studied in the brain, to validate our vectors, we initially compared the glial tropism of AAV-PHP.eB, an AAV9 that crosses the blood-brain and blood-retinal barriers, for its ability to drive fluorescent protein expression in glial cells in both the brain and retina. We then tested the glial transduction of AAV2/8-GFAP-mCherry, a virus that does not cross blood-brain and blood-retinal barriers, for its effectiveness in transducing Müller glia in murine retinal explants ex vivo. For in vivo assays we used larger rat eyes, performing invasive intravitreal injections, and non-invasive intravenous delivery using focused ultrasound (FUS) (pressure amplitude: 0.360 - 0.84 MPa) and microbubbles (Definity, 0.2 ml/kg). Results: We showed that AAV-PHP.eB carrying a ubiquitous promoter (CAG) and green fluorescent protein (GFP) reporter, readily crossed the blood-brain and blood-retinal barriers after intravenous delivery in mice. However, murine Müller glia did not express GFP, suggesting that they were not transduced by AAV-PHP.eB. We thus tested an AAV2/8 variant, which was selected based on its safety record in multiple clinical trials, adding a glial fibrillary acidic protein (GFAP) promoter and mCherry (red fluorescent protein) reporter. We confirmed the glial specificity of AAV2/8-GFAP-mCherry, showing effective expression of mCherry in astrocytes after intracranial injection in the mouse brain, and of Müller glia in murine retinal explants. For in vivo experiments we switched to rats because of their larger size, injecting AAV2/8-GFAP-mCherry intravitreally, an invasive procedure, demonstrating passage across the inner limiting membrane, leading to Müller glia transduction. We then tested an alternative non-invasive delivery approach targeting a different barrier - the inner blood-retinal-barrier, applying focused ultrasound (FUS) to the retina after intravenous injection of AAV2/8 and microbubbles in rats, using magnetic resonance imaging (MRI) for FUS targeting. FUS permeabilized the rat blood-retinal-barrier and allowed the passage of macromolecules to the retina (Evans blue, IgG, IgM), with minimal extravasation of platelets and red blood cells. Intravenous injection of microbubbles and AAV2/8-GFAP-mCherry followed by FUS resulted in mCherry expression in rat Müller glia. However, systemic delivery of AAV2/8 also had off-target effects, transducing several murine peripheral organs, particularly the liver. Conclusions: Retinal permeabilisation via FUS in the presence of microbubbles is effective for delivering AAV2/8 across the inner blood-retinal-barrier, targeting Müller glia, which is less invasive than intravitreal injections that bypass the inner limiting membrane. However, implementing FUS in the clinic will require a comprehensive consideration of any off-target tropism of the AAV in peripheral organs, combined ideally, with the development of Müller glia-specific promoters.
Collapse
Affiliation(s)
- Yacine Touahri
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Rajiv Dixit
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Rikke Hahn Kofoed
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Kristina Mikloska
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - EunJee Park
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Reza Raeisossadati
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Kelly Markham-Coultes
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Luke Ajay David
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Hibo Rijal
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Jiayi Zhao
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Madelaine Lynch
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Isabelle Aubert
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Carol Schuurmans
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
35
|
Structure comparison of the chimeric AAV2.7m8 vector with parental AAV2. J Struct Biol 2019; 209:107433. [PMID: 31859208 DOI: 10.1016/j.jsb.2019.107433] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/28/2019] [Accepted: 12/03/2019] [Indexed: 12/26/2022]
Abstract
The AAV2.7m8 vector is an engineered capsid with a 10-amino acid insertion in adeno-associated virus (AAV) surface variable region VIII (VR-VIII) resulting in the alteration of an antigenic region of AAV2 and the ability to efficiently transduce retina cells following intravitreal administration. Directed evolution and in vivo screening in the mouse retina isolated this vector. In the present study, we sought to identify the structural differences between a recombinant AAV2.7m8 (rAAV2.7m8) vector packaging a GFP genome and its parental serotype, AAV2, by cryo-electron microscopy (cryo-EM) and image reconstruction. The structures of rAAV2.7m8 and AAV2 were determined to 2.91 and 3.02 Å resolution, respectively. The rAAV2.7m8 amino acid side-chains for residues 219-745 (the last C-terminal residue) were interpretable in the density map with the exception of the 10 inserted amino acids. While observable in a low sigma threshold density, side-chains were only resolved at the base of the insertion, likely due to flexibility at the top of the loop. A comparison to parental AAV2 (ordered from residues 217-735) showed the structures to be similar, except at some side-chains that had different orientations and, in VR-VIII containing the 10 amino acid insertion. VR-VIII is part of an AAV2 antigenic epitope, and the difference is consistent with rAAV2.7m8's escape from a known AAV2 monoclonal antibody, C37-B. The observations provide valuable insight into the configuration of inserted surface peptides on the AAV capsid and structural differences to be leveraged for future AAV vector rational design, especially for retargeted tropism and antibody escape.
Collapse
|
36
|
Alrashdi B, Dawod B, Schampel A, Tacke S, Kuerten S, Marshall JS, Côté PD. Nav1.6 promotes inflammation and neuronal degeneration in a mouse model of multiple sclerosis. J Neuroinflammation 2019; 16:215. [PMID: 31722722 PMCID: PMC6852902 DOI: 10.1186/s12974-019-1622-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In multiple sclerosis (MS) and in the experimental autoimmune encephalomyelitis (EAE) model of MS, the Nav1.6 voltage-gated sodium (Nav) channel isoform has been implicated as a primary contributor to axonal degeneration. Following demyelination Nav1.6, which is normally co-localized with the Na+/Ca2+ exchanger (NCX) at the nodes of Ranvier, associates with β-APP, a marker of neural injury. The persistent influx of sodium through Nav1.6 is believed to reverse the function of NCX, resulting in an increased influx of damaging Ca2+ ions. However, direct evidence for the role of Nav1.6 in axonal degeneration is lacking. METHODS In mice floxed for Scn8a, the gene that encodes the α subunit of Nav1.6, subjected to EAE we examined the effect of eliminating Nav1.6 from retinal ganglion cells (RGC) in one eye using an AAV vector harboring Cre and GFP, while using the contralateral either injected with AAV vector harboring GFP alone or non-targeted eye as control. RESULTS In retinas, the expression of Rbpms, a marker for retinal ganglion cells, was found to be inversely correlated to the expression of Scn8a. Furthermore, the gene expression of the pro-inflammatory cytokines Il6 (IL-6) and Ifng (IFN-γ), and of the reactive gliosis marker Gfap (GFAP) were found to be reduced in targeted retinas. Optic nerves from targeted eyes were shown to have reduced macrophage infiltration and improved axonal health. CONCLUSION Taken together, our results are consistent with Nav1.6 promoting inflammation and contributing to axonal degeneration following demyelination.
Collapse
Affiliation(s)
- Barakat Alrashdi
- Department of Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Department of Biology, Al-Jouf University, Sakaka, Saudi Arabia
| | - Bassel Dawod
- Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Andrea Schampel
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sabine Tacke
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jean S Marshall
- Department of Pathology, Dalhousie University, Halifax, NS, B3H 4R2, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Patrice D Côté
- Department of Biology, Dalhousie University, Halifax, NS, B3H 4R2, Canada. .,Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
| |
Collapse
|
37
|
Tawarayama H, Feng Q, Murayama N, Suzuki N, Nakazawa T. Cyclin-Dependent Kinase Inhibitor 2b Mediates Excitotoxicity-Induced Death of Retinal Ganglion Cells. ACTA ACUST UNITED AC 2019; 60:4479-4488. [DOI: 10.1167/iovs.19-27396] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Hiroshi Tawarayama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Qiwei Feng
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Namie Murayama
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Noriyuki Suzuki
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
38
|
Waldner DM, Visser F, Fischer AJ, Bech-Hansen NT, Stell WK. Avian Adeno-Associated Viral Transduction of the Postembryonic Chicken Retina. Transl Vis Sci Technol 2019; 8:1. [PMID: 31293820 PMCID: PMC6608088 DOI: 10.1167/tvst.8.4.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023] Open
Abstract
Purpose The posthatching chicken is a valuable animal model for research, but molecular tools needed for altering its gene expression are not yet available. Our purpose here was to adapt the adeno-associated viral (AAV) vector method, used widely in mammalian studies, for use in investigations of the chicken retina. We hypothesized that the recently characterized avian AAV (A3V) vector could effectively transduce chick retinal cells for manipulation of gene expression, after intravitreal or subretinal injection. Methods A3V encoding enhanced green fluorescent protein (EGFP) was injected intravitreally or subretinally into P1-3 chick eye and left for 7 to 10 days. Retinas were then sectioned or flat-mounted and visualized via laser-scanning confocal microscopy for analysis of expression and imaging of retinal cells. Results Intravitreal A3V-EGFP injection resulted in EGFP expression in a small percent of retinal cells, primarily those with processes and/or cell bodies near the vitreal surface. In contrast, subretinal injection of A3V-EGFP within confined retinal “blebs” produced high rates of transduction of rods and all types of cones. Some examples of all other major retinal cell types, including horizontal, amacrine, bipolar, ganglion, and Müller cells, were also transduced, although with much lower frequency than photoreceptors. Conclusions A3V is a promising tool for investigating chick retinal cells and circuitry in situ. This novel vector can be used for studies in which local photoreceptor transduction is sufficient for meaningful observations. Translational Relevance With this vector, the postembryonic chick retina can now be used for preclinical trials of gene therapy for prevention and treatment of human retinal disease.
Collapse
Affiliation(s)
- Derek M Waldner
- Graduate Department of Neuroscience, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Frank Visser
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Andy J Fischer
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - N Torben Bech-Hansen
- Department of Medical Genetics, and Department of Surgery, Alberta Children's Hospital Research Institute, and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - William K Stell
- Department of Cell Biology and Anatomy and Department of Surgery, Hotchkiss Brain Institute, and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
39
|
Devoldere J, Peynshaert K, Dewitte H, Vanhove C, De Groef L, Moons L, Özcan SY, Dalkara D, De Smedt SC, Remaut K. Non-viral delivery of chemically modified mRNA to the retina: Subretinal versus intravitreal administration. J Control Release 2019; 307:315-330. [PMID: 31265881 DOI: 10.1016/j.jconrel.2019.06.042] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/20/2019] [Accepted: 06/29/2019] [Indexed: 12/20/2022]
Abstract
mRNA therapeutics have recently experienced a new wave of interest, mainly due to the discovery that chemical modifications to mRNA's molecular structure could drastically reduce its inherent immunogenicity and perceived instability. On this basis, we aimed to explore the potential of chemically stabilized mRNA for ocular applications. More specifically, we investigated the behavior of mRNA-loaded lipid-based carriers in human retinal cells (in vitro), in bovine retinal explants (ex vivo) and in mouse retinas (in vivo). We demonstrate a clear superiority of mRNA over pDNA to induce protein expression in different retinal cell types, which was further enhanced by chemical modification of the mRNA, providing up to ~1800-fold higher reporter gene expression compared to pDNA. Moreover, transgene expression could be detected for at least 20 days after a single administration of chemically modified mRNA in vitro. We furthermore determined the localization and extent of mRNA expression depending on the administration route. After subretinal (SR) administration, mRNA expression was observed in vivo and ex vivo. By contrast, intravitreal (IVT) administration resulted in limited expression in vivo. Using ex vivo bovine explants with an intact vitreoretinal (VR) interface we could attribute this to the inner limiting membrane (ILM), which presents a large barrier for non-viral delivery of mRNA, trapping mRNA complexes at the vitreal side. When the vitreous was removed, which compromises the ILM, mRNA expression was apparent and seemed to colocalize with Müller cells or photoreceptors after respectively IVT or SR administration. Taken together, this study represents a first step towards mRNA-mediated therapy for retinal diseases.
Collapse
Affiliation(s)
- Joke Devoldere
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Karen Peynshaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Medical School of the Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1050 Jette, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Christian Vanhove
- Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | - Sinem Yilmaz Özcan
- Neurological Sciences and Psychiatry Institute; Hacettepe University, Ankara, Turkey
| | - Deniz Dalkara
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
40
|
Devoldere J, Wels M, Peynshaert K, Dewitte H, De Smedt SC, Remaut K. The obstacle course to the inner retina: Hyaluronic acid-coated lipoplexes cross the vitreous but fail to overcome the inner limiting membrane. Eur J Pharm Biopharm 2019; 141:161-171. [PMID: 31150809 DOI: 10.1016/j.ejpb.2019.05.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/27/2019] [Accepted: 05/27/2019] [Indexed: 10/26/2022]
Abstract
Considerable research over the last few years has revealed dysregulation of growth factors in various retinal diseases, such as glaucoma, diabetic retinopathy and photoreceptor degenerations. The use of messengerRNA (mRNA) to transiently overexpress a specific factor could compensate for this imbalance. However, a critical challenge of this approach lies in the ability to efficiently deliver mRNA molecules to the retinal target cells. In this study we found that intravitreal (IVT) injection is an attractive approach to deliver mRNA to the retina, providing two critical barriers can be overcome: the vitreous and the inner limiting membrane (ILM). We demonstrated that the vitreous is indeed a major hurdle in the delivery of the cationic mRNA-complexes to retinal cells, both in terms of vitreal mobility and cellular uptake. To improve their intravitreal mobility and avoid unwanted extracellular interactions, we evaluated the use of hyaluronic acid (HA) as an electrostatic coating strategy. This HA-coating provided the complexes with a negative surface charge, markedly enhancing their mobility in the vitreous humor, without reducing their cellular internalization and transfection efficiency. However, although this coating strategy allows the mRNA-complexes to successfully overcome the vitreal barrier, the majority of the particles accumulated at the ILM. This study therefore underscores the crucial barrier function of the ILM toward non-viral retinal gene delivery and the need to smartly design mRNA-carriers able to surmount the vitreous as well as the ILM.
Collapse
Affiliation(s)
- Joke Devoldere
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Mike Wels
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Karen Peynshaert
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Medical School of the Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1050 Jette, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Katrien Remaut
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
41
|
Hori T, Fukutome M, Maejima C, Matsushima H, Kobayashi K, Kitazawa S, Kitahara R, Kitano K, Kobayashi K, Moritoh S, Koike C. Gene delivery to cone photoreceptors by subretinal injection of rAAV2/6 in the mouse retina. Biochem Biophys Res Commun 2019; 515:222-227. [PMID: 31146917 DOI: 10.1016/j.bbrc.2019.05.117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 05/17/2019] [Indexed: 11/29/2022]
Abstract
Adeno-associated virus (AAV) has been studied as a safe delivery tool for gene therapy of retinal blinding diseases such as Leber's congenital amaurosis (LCA). The tropism of recombinant AAV (rAAV) including its specificity and efficiency in targeting retinal cell types has been studied with native or engineered capsids, along with specific promoters. However, one of the rAAV serotypes, rAAV2/6, has not been well-studied based on a report of low infection efficiency in the retina. We investigated the tropism of several rAAVs by subretinal injection in the adult mouse and found that rAAV2/6 predominantly infected cone photoreceptors including the main spectral type. Our data suggest that subretinal injection with rAAV2/6 may provide both an efficacious and specific means of gene delivery to cone photoreceptors in murine retinas.
Collapse
Affiliation(s)
- Tesshu Hori
- Graduate School of Pharmacy, Ritsumeikan University, Kusatsu, Shiga, Japan; College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Masashi Fukutome
- College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan; Graduate School of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Chiseto Maejima
- College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Hiroki Matsushima
- Graduate School of Pharmacy, Ritsumeikan University, Kusatsu, Shiga, Japan; College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Kensuke Kobayashi
- College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan; Graduate School of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Soichiro Kitazawa
- College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Ryo Kitahara
- College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan; Graduate School of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Katsunori Kitano
- College of Information Science and Engineering, Ritsumeikan University, Kusatsu, Shiga, Japan; Center for Systems Vision Science, Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Satoru Moritoh
- College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan; Center for Systems Vision Science, Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Chieko Koike
- College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan; Graduate School of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan; Center for Systems Vision Science, Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, Shiga, Japan.
| |
Collapse
|
42
|
Adeno-associated virus neutralising antibodies in type 1 diabetes mellitus. Gene Ther 2019; 26:250-263. [PMID: 30962537 DOI: 10.1038/s41434-019-0076-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 12/26/2022]
Abstract
Recombinant Adeno-associated viruses (AAVs) are an attractive vector for gene therapy delivery which may be blocked by AAV neutralising antibodies (NAbs). As Type 1 Diabetes (T1DM) is an endocrine disease of immunological origin, it is likely that NAb profiles are altered in the disease. In this study NAb to AAV2, AAV5, AAV6, and AAV8 in 72 subjects with T1DM and 45 non-diabetic patients were measured over a 4-year follow-up period. AAV2 NAb titres were significantly lower in non-diabetic subjects (P = 0.036). The T1DM group had more AAV8 NAb activity at baseline (P = 0.019), whilst after 4 years follow-up the T1DM group displayed developed increased AAV 5 (P = 0.03), 6 (P = 0.03) and 8 (P = 0.002) activity relative to the control group, however, overall AAV5 and 8 NAb levels were very low in patients <40. AAV NAb titre activity and prevalence generally appears higher in T1DM, however, low levels of AAV 5 and 8, particular in younger adult age groups at which T1DM can be targeted, could make these attractive vectors to target the disease.
Collapse
|
43
|
Hori T, Fukutome M, Koike C. Adeno Associated Virus (AAV) as a Tool for Clinical and Experimental Delivery of Target Genes into the Mammalian Retina. Biol Pharm Bull 2019; 42:343-347. [DOI: 10.1248/bpb.b18-00913] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Tesshu Hori
- Laboratory for Systems Neuroscience and Developmental Biology, College of Pharmaceutical Sciences, Ritsumeikan University
| | | | - Chieko Koike
- Laboratory for Systems Neuroscience and Developmental Biology, College of Pharmaceutical Sciences, Ritsumeikan University
- Graduate School of Life Sciences, Ritsumeikan University
- Center for Systems Vision Science, Research Organization of Science and Technology, Ritsumeikan University
| |
Collapse
|
44
|
Peynshaert K, Devoldere J, Minnaert AK, De Smedt SC, Remaut K. Morphology and Composition of the Inner Limiting Membrane: Species-Specific Variations and Relevance toward Drug Delivery Research. Curr Eye Res 2019; 44:465-475. [PMID: 30638413 DOI: 10.1080/02713683.2019.1565890] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The inner limiting membrane (ILM) represents the structural boundary between the vitreous and the retina, and is suggested to act as a barrier for a wide range of retinal therapies. While it is widely acknowledged that the morphology of the human ILM exhibits regional variations and undergoes age-related changes, insight into its structure in laboratory animals is very limited. Besides presenting a detailed overview of the morphology and composition of the human ILM, this review specifically reflects on the species-specific differences in ILM structure. With these differences in mind, we furthermore summarize the most relevant reports on the barrier role of the ILM with regard to viral vectors, nanoparticles, anti-VEGF medication and stem cells. Overall, this review aims to deliberate on the impact of species-specific ILM variations on drug delivery research as well as to pinpoint knowledge gaps which future basic research should resolve.
Collapse
Affiliation(s)
- Karen Peynshaert
- a Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium.,b Ghent Research Group on Nanomedicines , Ghent University , Ghent , Belgium
| | - Joke Devoldere
- a Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium.,b Ghent Research Group on Nanomedicines , Ghent University , Ghent , Belgium
| | - An-Katrien Minnaert
- a Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium.,b Ghent Research Group on Nanomedicines , Ghent University , Ghent , Belgium
| | - Stefaan C De Smedt
- a Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium.,b Ghent Research Group on Nanomedicines , Ghent University , Ghent , Belgium
| | - Katrien Remaut
- a Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium.,b Ghent Research Group on Nanomedicines , Ghent University , Ghent , Belgium
| |
Collapse
|
45
|
Katada Y, Kobayashi K, Tsubota K, Kurihara T. Evaluation of AAV-DJ vector for retinal gene therapy. PeerJ 2019; 7:e6317. [PMID: 30671314 PMCID: PMC6339780 DOI: 10.7717/peerj.6317] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/19/2018] [Indexed: 01/02/2023] Open
Abstract
Purpose The most common virus vector used in gene therapy research for ophthalmologic diseases is the adeno-associated virus (AAV) vector, which has been used successfully in a number of preclinical and clinical studies. It is important to evaluate novel AAV vectors in animal models for application of clinical gene therapy. The AAV-DJ (type 2/type 8/type 9 chimera) was engineered from shuffling eight different wild-type native viruses. In this study, we investigated the efficiency of gene transfer by AAV-DJ injections into the retina. Methods One microliter of AAV-2-CAGGS-EGFP or AAV-DJ-CAGGS-EGFP vector at a titer of 1.4 × 10e12 vg/ml was injected intravitreally or subretinally in each eye of C57BL/6 mice. We evaluated the transduction characteristics of AAV-2 and -DJ vectors using fluorescence microscopy and electroretinography. Results The results confirmed that AAV-DJ could deeply transfer gene to photoreceptor layer with intravitreal injection and has an efficient gene transfer to various cell types especially the Mueller cells in the retina. Retinal function was not affected by AAV-DJ infection or ectopic EGFP expression. Conclusions The AAV-DJ vector efficiently induces the reporter gene in both the inner and outer murine retina without functional toxicity. These data indicated that the AAV-DJ vector is a useful tool for the gene therapy research targeting retinal disorders.
Collapse
Affiliation(s)
- Yusaku Katada
- Department of Ophthalmology, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan.,Laboratory of Photobiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Toshihide Kurihara
- Department of Ophthalmology, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan.,Laboratory of Photobiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
46
|
Song H, Bush RA, Zeng Y, Qian H, Wu Z, Sieving PA. Trans-ocular Electric Current In Vivo Enhances AAV-Mediated Retinal Gene Transduction after Intravitreal Vector Administration. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 13:77-85. [PMID: 30719486 PMCID: PMC6350231 DOI: 10.1016/j.omtm.2018.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/14/2018] [Indexed: 01/31/2023]
Abstract
Adeno-associated virus (AAV) vector-mediated gene delivery is a promising approach for therapy, but implementation in the eye currently is hampered by the need for delivering the vector underneath the retina, using surgical application into the subretinal space. This limits the extent of the retina that is treated and may cause surgical injury. Vector delivery into the vitreous cavity would be preferable because it is surgically less invasive and would reach more of the retina. Unfortunately, most conventional, non-modified AAV vector serotypes penetrate the retina poorly from the vitreous; this limits efficient transduction and expression by target cells (retinal pigment epithelium and photoreceptors). We developed a method of applying a small and safe electric current across the intact eye in vivo for a brief period following intravitreal vector administration. This significantly improved AAV-mediated transduction of retinal cells in wild-type mice following intravitreal delivery, with gene expression in retinal pigment epithelium and photoreceptor cells. The low-level current had no adverse effects on retinal structure and function. This method should be generally applicable for other AAV serotypes and may have broad application in both basic research and clinical studies.
Collapse
Affiliation(s)
- Hongman Song
- Section for Translational Research on Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Ronald A Bush
- Section for Translational Research on Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Yong Zeng
- Section for Translational Research on Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Haohua Qian
- National Eye Institute, NIH, Bethesda, MD 20892, USA
| | - Zhijian Wu
- National Eye Institute, NIH, Bethesda, MD 20892, USA
| | - Paul A Sieving
- Section for Translational Research on Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA.,National Eye Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
47
|
Looser ZJ, Barrett MJP, Hirrlinger J, Weber B, Saab AS. Intravitreal AAV-Delivery of Genetically Encoded Sensors Enabling Simultaneous Two-Photon Imaging and Electrophysiology of Optic Nerve Axons. Front Cell Neurosci 2018; 12:377. [PMID: 30405358 PMCID: PMC6205974 DOI: 10.3389/fncel.2018.00377] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/03/2018] [Indexed: 11/21/2022] Open
Abstract
Myelination of axons by oligodendrocytes is a key feature of the remarkably fast operating CNS. Oligodendrocytes not only tune axonal conduction speed but are also suggested to maintain long-term axonal integrity by providing metabolic support to the axons they ensheath. However, how myelinating oligodendrocytes impact axonal energy homeostasis remains poorly understood and difficult to investigate. Here, we provide a method of how to study electrically active myelinated axons expressing genetically encoded sensors by combining electrophysiology and two-photon imaging of acutely isolated optic nerves. We show that intravitreal adeno-associated viral (AAV) vector delivery is an efficient tool to achieve functional sensor expression in optic nerve axons, which is demonstrated by measuring axonal ATP dynamics following AAV-mediated sensor expression. This novel approach allows for fast expression of any optical sensor of interest to be studied in optic nerve axons without the need to go through the laborious process of producing new transgenic mouse lines. Viral-mediated biosensor expression in myelinated axons and the subsequent combination of nerve recordings and sensor imaging outlines a powerful method to investigate oligodendroglial support functions and to further interrogate cellular mechanisms governing axonal energy homeostasis under physiological and pathological conditions.
Collapse
Affiliation(s)
- Zoe J. Looser
- Institute of Pharmacology & Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Matthew J. P. Barrett
- Institute of Pharmacology & Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Johannes Hirrlinger
- Carl-Ludwig-Institute for Physiology, University of Leipzig, Leipzig, Germany
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany
| | - Bruno Weber
- Institute of Pharmacology & Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Aiman S. Saab
- Institute of Pharmacology & Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
48
|
Human Vascular Endothelial Growth Factor A 165 Expression Induces the Mouse Model of Neovascular Age-Related Macular Degeneration. Genes (Basel) 2018; 9:genes9090438. [PMID: 30200369 PMCID: PMC6162490 DOI: 10.3390/genes9090438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/24/2018] [Accepted: 08/28/2018] [Indexed: 12/27/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) expression induces age-related macular degeneration (AMD), which is a common vision-threatening disease due to choroidal neovascularization and a fibrovascular membrane. We describe a mouse model of neovascular AMD with the local expression of human VEGF-A165 in the eye. We use a transgenic mouse in which human VEGF-A165 has been silenced with the loxP-STOP fragment. The choroidal neovascularization and human VEGF-A165 expression in the mouse are induced by subretinal adenoviral Cre gene delivery. Cre gene transfer is compared with adenoviral LacZ gene transfer control. We characterize the AMD phenotype and changes in the vasculature by using fluorescein angiography, optical coherence tomography, and immunohistochemistry. At early time points, mice exhibit increases in retinal thickness (348 ± 114 µm vs. 231 ± 32 µm) and choroidal neovascularization area (12000 ± 15174 µm² vs. 2169 ± 3495 µm²) compared with the control. At later time points, choroidal neovascularization develops into subretinal fibrovascular membrane. Human VEGF-A165 expression lasts several weeks. In conclusion, the retinas display vascular abnormalities consistent with choroidal neovascularization. Together with immunohistochemical findings, these changes resemble clinical AMD-like ocular pathologies. We conclude that this mouse model of Cre-induced choroidal neovascularization is useful for mimicking the pathogenesis of AMD, studying the effects of human VEGF-A165 in the retina, and evaluating anti-VEGF treatments for choroidal neovascularization.
Collapse
|
49
|
Sullivan JA, Stanek LM, Lukason MJ, Bu J, Osmond SR, Barry EA, O'Riordan CR, Shihabuddin LS, Cheng SH, Scaria A. Rationally designed AAV2 and AAVrh8R capsids provide improved transduction in the retina and brain. Gene Ther 2018; 25:205-219. [PMID: 29785047 DOI: 10.1038/s41434-018-0017-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/25/2018] [Accepted: 03/27/2018] [Indexed: 01/18/2023]
Abstract
The successful application of adeno-associated virus (AAV) gene delivery vectors as a therapeutic paradigm will require efficient gene delivery to the appropriate cells in affected organs. In this study, we utilized a rational design approach to introduce modifications to the AAV2 and AAVrh8R capsids and the resulting variants were evaluated for transduction activity in the retina and brain. The modifications disrupted either capsid/receptor binding or altered capsid surface charge. Specifically, we mutated AAV2 amino acids R585A and R588A, which are required for binding to its receptor, heparan sulfate proteoglycans, to generate a variant referred to as AAV2-HBKO. In contrast to parental AAV2, the AAV2-HBKO vector displayed low-transduction activity following intravitreal delivery to the mouse eye; however, following its subretinal delivery, AAV2-HBKO resulted in significantly greater photoreceptor transduction. Intrastriatal delivery of AAV2-HBKO to mice facilitated widespread striatal and cortical expression, in contrast to the restricted transduction pattern of the parental AAV2 vector. Furthermore, we found that altering the surface charge on the AAVrh8R capsid by modifying the number of arginine residues on the capsid surface had a profound impact on subretinal transduction. The data further validate the potential of capsid engineering to improve AAV gene therapy vectors for clinical applications.
Collapse
Affiliation(s)
| | - Lisa M Stanek
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| | | | - Jie Bu
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| | | | | | | | | | - Seng H Cheng
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| | - Abraham Scaria
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| |
Collapse
|
50
|
Progress in Gene Therapy to Prevent Retinal Ganglion Cell Loss in Glaucoma and Leber's Hereditary Optic Neuropathy. Neural Plast 2018; 2018:7108948. [PMID: 29853847 PMCID: PMC5954906 DOI: 10.1155/2018/7108948] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/04/2018] [Indexed: 12/24/2022] Open
Abstract
The eye is at the forefront of the application of gene therapy techniques to medicine. In the United States, a gene therapy treatment for Leber's congenital amaurosis, a rare inherited retinal disease, recently became the first gene therapy to be approved by the FDA for the treatment of disease caused by mutations in a specific gene. Phase III clinical trials of gene therapy for other single-gene defect diseases of the retina and optic nerve are also currently underway. However, for optic nerve diseases not caused by single-gene defects, gene therapy strategies are likely to focus on slowing or preventing neuronal death through the expression of neuroprotective agents. In addition to these strategies, there has also been recent interest in the potential use of precise genome editing techniques to treat ocular disease. This review focuses on recent developments in gene therapy techniques for the treatment of glaucoma and Leber's hereditary optic neuropathy (LHON). We discuss recent successes in clinical trials for the treatment of LHON using gene supplementation therapy, promising neuroprotective strategies that have been employed in animal models of glaucoma and the potential use of genome editing techniques in treating optic nerve disease.
Collapse
|