1
|
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
|
2
|
McDonald A, Wijnholds J. Retinal Ciliopathies and Potential Gene Therapies: A Focus on Human iPSC-Derived Organoid Models. Int J Mol Sci 2024; 25:2887. [PMID: 38474133 PMCID: PMC10932180 DOI: 10.3390/ijms25052887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
The human photoreceptor function is dependent on a highly specialised cilium. Perturbation of cilial function can often lead to death of the photoreceptor and loss of vision. Retinal ciliopathies are a genetically diverse range of inherited retinal disorders affecting aspects of the photoreceptor cilium. Despite advances in the understanding of retinal ciliopathies utilising animal disease models, they can often lack the ability to accurately mimic the observed patient phenotype, possibly due to structural and functional deviations from the human retina. Human-induced pluripotent stem cells (hiPSCs) can be utilised to generate an alternative disease model, the 3D retinal organoid, which contains all major retinal cell types including photoreceptors complete with cilial structures. These retinal organoids facilitate the study of disease mechanisms and potential therapies in a human-derived system. Three-dimensional retinal organoids are still a developing technology, and despite impressive progress, several limitations remain. This review will discuss the state of hiPSC-derived retinal organoid technology for accurately modelling prominent retinal ciliopathies related to genes, including RPGR, CEP290, MYO7A, and USH2A. Additionally, we will discuss the development of novel gene therapy approaches targeting retinal ciliopathies, including the delivery of large genes and gene-editing techniques.
Collapse
Affiliation(s)
- Andrew McDonald
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
- Netherlands Institute of Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
3
|
Hu S, Chen Y, Xie D, Xu K, Fu Y, Chi W, Liu H, Huang J. Nme 2 Cas9-mediated therapeutic editing in inhibiting angiogenesis after wet age-related macular degeneration onset. Clin Transl Med 2023; 13:e1383. [PMID: 37598400 PMCID: PMC10440058 DOI: 10.1002/ctm2.1383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/17/2023] [Accepted: 08/13/2023] [Indexed: 08/22/2023] Open
Abstract
BACKGROUND Age-related macular degeneration (AMD), particularly wet AMD characterised by choroidal neovascularization (CNV), is a leading cause of vision loss in the elderly. The hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF)/VEGF receptor 2 (VEGFR2) pathway contributes to CNV pathogenesis. Previous gene editing research indicated that disrupting these genes in retinal pigment epithelial cells could have a preventive effect on CNV progression. However, no studies have yet been conducted using gene editing to disrupt VEGF signalling after CNV induction for therapeutic validation, which is critical to the clinical application of wet AMD gene editing therapies. METHOD Here, we employed the single-adeno-associated virus-mediated Nme2 Cas9 to disrupt key molecules in VEGF signalling, Hif1α, Vegfa and Vegfr2 after inducing CNV and estimated their therapeutic effects. RESULTS We found that Nme2 Cas9 made efficient editing in target genes up to 71.8% post 11 days in vivo. And only Nme2 Cas9-Vegfa treatment during the early stage of CNV development reduced the CNV lesion area by 49.5%, compared to the negative control, while Nme2 Cas9-Hif1α or Nme2 Cas9-Vegfr2 treatment did not show therapeutic effect. Besides, no off-target effects were observed in Nme2 Cas9-mediated gene editing in vivo. CONCLUSIONS This study provides proof-of-concept possibility of employing Nme2 Cas9 for potential anti-angiogenesis therapy in wet AMD.
Collapse
Affiliation(s)
- Sihui Hu
- MOE Key Laboratory of Gene Function and RegulationState Key Laboratory of BiocontrolSchool of Life SciencesSun Yat‐sen UniversityGuangzhouChina
| | - Yuxi Chen
- MOE Key Laboratory of Gene Function and RegulationState Key Laboratory of BiocontrolSchool of Life SciencesSun Yat‐sen UniversityGuangzhouChina
| | - Dongchun Xie
- MOE Key Laboratory of Gene Function and RegulationState Key Laboratory of BiocontrolSchool of Life SciencesSun Yat‐sen UniversityGuangzhouChina
| | - Kan Xu
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Yunzhao Fu
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Wei Chi
- The State Key Laboratory of Ophthalmology Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Haiying Liu
- MOE Key Laboratory of Gene Function and RegulationState Key Laboratory of BiocontrolSchool of Life SciencesSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Reproductive Medicine of Guangdong ProvinceSchool of Life Sciences and the First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Junjiu Huang
- MOE Key Laboratory of Gene Function and RegulationState Key Laboratory of BiocontrolSchool of Life SciencesSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Reproductive Medicine of Guangdong ProvinceSchool of Life Sciences and the First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
4
|
Chirco KR, Martinez C, Lamba DA. Advancements in pre-clinical development of gene editing-based therapies to treat inherited retinal diseases. Vision Res 2023; 209:108257. [PMID: 37210864 PMCID: PMC10524382 DOI: 10.1016/j.visres.2023.108257] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/23/2023]
Abstract
One of the major goals in the inherited retinal disease (IRD) field is to develop an effective therapy that can be applied to as many patients as possible. Significant progress has already been made toward this end, with gene editing at the forefront. The advancement of gene editing-based tools has been a recent focus of many research groups around the world. Here, we provide an update on the status of CRISPR/Cas-derived gene editors, promising options for delivery of these editing systems to the retina, and animal models that aid in pre-clinical testing of new IRD therapeutics.
Collapse
Affiliation(s)
- Kathleen R Chirco
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States.
| | - Cassandra Martinez
- Department of Ophthalmology, University of California San Francisco, CA, United States; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, CA, United States
| | - Deepak A Lamba
- Department of Ophthalmology, University of California San Francisco, CA, United States; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, CA, United States
| |
Collapse
|
5
|
Chan YC, Kienle E, Oti M, Di Liddo A, Mendez-Lago M, Aschauer DF, Peter M, Pagani M, Arnold C, Vonderheit A, Schön C, Kreuz S, Stark A, Rumpel S. An unbiased AAV-STARR-seq screen revealing the enhancer activity map of genomic regions in the mouse brain in vivo. Sci Rep 2023; 13:6745. [PMID: 37185990 PMCID: PMC10130037 DOI: 10.1038/s41598-023-33448-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Enhancers are important cis-regulatory elements controlling cell-type specific expression patterns of genes. Furthermore, combinations of enhancers and minimal promoters are utilized to construct small, artificial promoters for gene delivery vectors. Large-scale functional screening methodology to construct genomic maps of enhancer activities has been successfully established in cultured cell lines, however, not yet applied to terminally differentiated cells and tissues in a living animal. Here, we transposed the Self-Transcribing Active Regulatory Region Sequencing (STARR-seq) technique to the mouse brain using adeno-associated-viruses (AAV) for the delivery of a highly complex screening library tiling entire genomic regions and covering in total 3 Mb of the mouse genome. We identified 483 sequences with enhancer activity, including sequences that were not predicted by DNA accessibility or histone marks. Characterizing the expression patterns of fluorescent reporters controlled by nine candidate sequences, we observed differential expression patterns also in sparse cell types. Together, our study provides an entry point for the unbiased study of enhancer activities in organisms during health and disease.
Collapse
Affiliation(s)
- Ya-Chien Chan
- Institute of Physiology, Focus Program Translational Neurosciences, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Eike Kienle
- Institute of Physiology, Focus Program Translational Neurosciences, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Martin Oti
- Institute of Molecular Biology GmbH (IMB), Mainz, Germany
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an Der Riß, Germany
| | | | | | - Dominik F Aschauer
- Institute of Physiology, Focus Program Translational Neurosciences, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Manuel Peter
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Michaela Pagani
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Cosmas Arnold
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | | | - Christian Schön
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an Der Riß, Germany
| | - Sebastian Kreuz
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an Der Riß, Germany
| | - Alexander Stark
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
- Medical University of Vienna, Vienna BioCenter (VBC), 1030, Vienna, Austria
| | - Simon Rumpel
- Institute of Physiology, Focus Program Translational Neurosciences, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
6
|
Altay HY, Ozdemir F, Afghah F, Kilinc Z, Ahmadian M, Tschopp M, Agca C. Gene regulatory and gene editing tools and their applications for retinal diseases and neuroprotection: From proof-of-concept to clinical trial. Front Neurosci 2022; 16:924917. [PMID: 36340792 PMCID: PMC9630553 DOI: 10.3389/fnins.2022.924917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/26/2022] [Indexed: 09/11/2023] Open
Abstract
Gene editing and gene regulatory fields are continuously developing new and safer tools that move beyond the initial CRISPR/Cas9 technology. As more advanced applications are emerging, it becomes crucial to understand and establish more complex gene regulatory and editing tools for efficient gene therapy applications. Ophthalmology is one of the leading fields in gene therapy applications with more than 90 clinical trials and numerous proof-of-concept studies. The majority of clinical trials are gene replacement therapies that are ideal for monogenic diseases. Despite Luxturna's clinical success, there are still several limitations to gene replacement therapies including the size of the target gene, the choice of the promoter as well as the pathogenic alleles. Therefore, further attempts to employ novel gene regulatory and gene editing applications are crucial to targeting retinal diseases that have not been possible with the existing approaches. CRISPR-Cas9 technology opened up the door for corrective gene therapies with its gene editing properties. Advancements in CRISPR-Cas9-associated tools including base modifiers and prime editing already improved the efficiency and safety profile of base editing approaches. While base editing is a highly promising effort, gene regulatory approaches that do not interfere with genomic changes are also becoming available as safer alternatives. Antisense oligonucleotides are one of the most commonly used approaches for correcting splicing defects or eliminating mutant mRNA. More complex gene regulatory methodologies like artificial transcription factors are also another developing field that allows targeting haploinsufficiency conditions, functionally equivalent genes, and multiplex gene regulation. In this review, we summarized the novel gene editing and gene regulatory technologies and highlighted recent translational progress, potential applications, and limitations with a focus on retinal diseases.
Collapse
Affiliation(s)
- Halit Yusuf Altay
- Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, Turkey
| | - Fatma Ozdemir
- Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, Turkey
| | - Ferdows Afghah
- Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, Turkey
| | - Zeynep Kilinc
- Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, Turkey
| | - Mehri Ahmadian
- Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, Turkey
| | - Markus Tschopp
- Department of Ophthalmology, Cantonal Hospital Aarau, Aarau, Switzerland
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Cavit Agca
- Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, Turkey
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul, Turkey
| |
Collapse
|
7
|
Ramamurthy RM, Atala A, Porada CD, Almeida-Porada G. Organoids and microphysiological systems: Promising models for accelerating AAV gene therapy studies. Front Immunol 2022; 13:1011143. [PMID: 36225917 PMCID: PMC9549755 DOI: 10.3389/fimmu.2022.1011143] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/01/2022] [Indexed: 11/24/2022] Open
Abstract
The FDA has predicted that at least 10-20 gene therapy products will be approved by 2025. The surge in the development of such therapies can be attributed to the advent of safe and effective gene delivery vectors such as adeno-associated virus (AAV). The enormous potential of AAV has been demonstrated by its use in over 100 clinical trials and the FDA’s approval of two AAV-based gene therapy products. Despite its demonstrated success in some clinical settings, AAV-based gene therapy is still plagued by issues related to host immunity, and recent studies have suggested that AAV vectors may actually integrate into the host cell genome, raising concerns over the potential for genotoxicity. To better understand these issues and develop means to overcome them, preclinical model systems that accurately recapitulate human physiology are needed. The objective of this review is to provide a brief overview of AAV gene therapy and its current hurdles, to discuss how 3D organoids, microphysiological systems, and body-on-a-chip platforms could serve as powerful models that could be adopted in the preclinical stage, and to provide some examples of the successful application of these models to answer critical questions regarding AAV biology and toxicity that could not have been answered using current animal models. Finally, technical considerations while adopting these models to study AAV gene therapy are also discussed.
Collapse
|
8
|
Bartsch U, Storch S. Experimental Therapeutic Approaches for the Treatment of Retinal Pathology in Neuronal Ceroid Lipofuscinoses. Front Neurol 2022; 13:866983. [PMID: 35509995 PMCID: PMC9058077 DOI: 10.3389/fneur.2022.866983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are a group of childhood-onset neurodegenerative lysosomal storage disorders mainly affecting the brain and the retina. In the NCLs, disease-causing mutations in 13 different ceroid lipofuscinoses genes (CLN) have been identified. The clinical symptoms include seizures, progressive neurological decline, deterioration of motor and language skills, and dementia resulting in premature death. In addition, the deterioration and loss of vision caused by progressive retinal degeneration is another major hallmark of NCLs. To date, there is no curative therapy for the treatment of retinal degeneration and vision loss in patients with NCL. In this review, the key findings of different experimental approaches in NCL animal models aimed at attenuating progressive retinal degeneration and the decline in retinal function are discussed. Different approaches, including experimental enzyme replacement therapy, gene therapy, cell-based therapy, and immunomodulation therapy were evaluated and showed encouraging therapeutic benefits. Recent experimental ocular gene therapies in NCL animal models with soluble lysosomal enzyme deficiencies and transmembrane protein deficiencies have shown the strong potential of gene-based approaches to treat retinal dystrophies in NCLs. In CLN3 and CLN6 mouse models, an adeno-associated virus (AAV) vector-mediated delivery of CLN3 and CLN6 to bipolar cells has been shown to attenuate the retinal dysfunction. Therapeutic benefits of ocular enzyme replacement therapies were evaluated in CLN2 and CLN10 animal models. Since brain-targeted gene or enzyme replacement therapies will most likely not attenuate retinal neurodegeneration, there is an unmet need for treatment options additionally targeting the retina in patients with NCL. The long-term benefits of these therapeutic interventions aimed at attenuating retinal degeneration and vision loss in patients with NCL remain to be investigated in future clinical studies.
Collapse
Affiliation(s)
- Udo Bartsch
- Department of Ophthalmology, Experimental Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Storch
- University Children's Research@Kinder-UKE, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- *Correspondence: Stephan Storch
| |
Collapse
|
9
|
Moraru AD, Costin D, Iorga RE, Munteanu M, Moraru RL, Branisteanu DC. Current trends in gene therapy for retinal diseases (Review). Exp Ther Med 2021; 23:26. [PMID: 34815778 PMCID: PMC8593927 DOI: 10.3892/etm.2021.10948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/10/2021] [Indexed: 11/17/2022] Open
Abstract
The eye is considered an effective target for genetic therapy, as it has a privileged immune status, it is easily accessed for medication delivery and it is affected by a number of inherited disorders. In particular, the retina is considered for gene therapy due to the fact that it can be visualized with ease, it does not have lymphatic vessels, nor a direct blood network for the outer layers and its cells do not divide after birth, and thus transgene expression is not affected. As gene therapy is currently on a continuously progressive development trend, this emerging field of gene manipulation techniques has yielded promising results. This involves the development of treatments for a number of debilitating and blinding diseases, which were to date considered intractable. However, numerous unanswered questions remain as regards the long-term efficacy and safety profile of these treatments. The present review article discusses the current research status regarding genetic manipulation techniques aimed at addressing visual impairment related to retinal disorders, both inherited and degenerative.
Collapse
Affiliation(s)
- Andreea Dana Moraru
- Department of Ophthalmology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iași, Romania.,Department of Ophthalmology, 'N. Oblu' Clinical Hospital, 700309 Iași, Romania
| | - Dănuț Costin
- Department of Ophthalmology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iași, Romania.,Department of Ophthalmology, 'N. Oblu' Clinical Hospital, 700309 Iași, Romania
| | - Raluca Eugenia Iorga
- Department of Ophthalmology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iași, Romania.,Department of Ophthalmology, 'N. Oblu' Clinical Hospital, 700309 Iași, Romania
| | - Mihnea Munteanu
- Department of Ophthalmology, 'Victor Babeș' University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Radu Lucian Moraru
- Department of Otorhinolaryngology, 'Transmed Expert' Medical Center, 700011 Iași, Romania
| | - Daniel Constantin Branisteanu
- Department of Ophthalmology, 'Grigore T. Popa' University of Medicine and Pharmacy, 700115 Iași, Romania.,Department of Ophthalmology, 'Retina Center' Eye Clinic, 700126 Iași, Romania
| |
Collapse
|
10
|
Weigelt CM, Fuchs H, Schönberger T, Stierstorfer B, Strobel B, Lamla T, Ciossek T, Bakker RA, Redemann NH. AAV-Mediated Expression of Human VEGF, TNF-α, and IL-6 Induces Retinal Pathology in Mice. Transl Vis Sci Technol 2021; 10:15. [PMID: 34520511 PMCID: PMC8444492 DOI: 10.1167/tvst.10.11.15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Retinopathies display complex pathologies, including vasculopathies, inflammation, and fibrosis, leading ultimately to visual impairment. However, animal models accurately reflecting these pathologies are lacking. In this study, we evaluate the suitability of using Adeno-associated virus (AAV)-mediated long-term expression of cytokines to establish retinal pathology in the murine retina. Methods We administered recombinant, Müller-glia targeted AAV-ShH10 into the mouse vitreous to induce retinal expression of either human vascular endothelial growth factor (VEGF)-A165, tumor necrosis factor alpha (TNF-α), or interleukin-6 (IL-6) and evaluated consequent effects by optical coherence tomography, fluorescein angiography, and histology. Results Intravitreal injection of AAVs resulted in rapid and stable expression of the transgenes within 1 to 6 weeks. Akin to the role of VEGF-A in wet age-related macular degeneration, expression of VEGF-A led to several vasculopathies in mice, including neovascularization and vascular leakage. In contrast, the expression of the proinflammatory cytokines TNF-α or IL-6 induced retinal inflammation, as indicated by microglial activation. Furthermore, the expression of TNF-α, but not of IL-6, induced immune cell infiltration into the vitreous as well as vasculitis, and subsequently induced the development of fibrosis and epiretinal membranes. Conclusions In summary, the long-term expression of human VEGF-A165, TNF-α, or IL-6 in the mouse eye induced specific pathologies within 6 weeks that mimic different aspects of human retinopathies. Translational Relevance AAV-mediated expression of human genes in mice is an attractive approach to provide valuable insights into the underlying molecular mechanisms causing retinopathies and is easily adaptable to other genes and preclinical species supporting drug discovery for retinal diseases.
Collapse
Affiliation(s)
- Carina M Weigelt
- Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Holger Fuchs
- Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Tanja Schönberger
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Birgit Stierstorfer
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Benjamin Strobel
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Thorsten Lamla
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Thomas Ciossek
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Remko A Bakker
- Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Norbert H Redemann
- Cardiometabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| |
Collapse
|
11
|
Development of a stable lyophilized adeno-associated virus gene therapy formulation. Int J Pharm 2021; 606:120912. [PMID: 34298099 DOI: 10.1016/j.ijpharm.2021.120912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/13/2021] [Accepted: 07/18/2021] [Indexed: 11/21/2022]
Abstract
Adeno-associated viruses (AAV) are among the most actively investigated vectors for gene therapy. Supply of early clinical studies with frozen drug product (DP) can accelerate timelines and minimize degradation risks. In the long-term, logistical challenges of frozen DP may limit patient access. In this work, we developed a lyophilized (freeze-dried) formulation of AAV. The mass concentration of AAV is typically low, and AAV also requires a minimum ionic strength to inhibit aggregation. These factors result in a low collapse temperature, which is limiting to lyophilization. Mannitol crystallization was found to cause extensive degradation and potency loss of AAV during the freezing step. With further development, we determined that AAV could be lyophilized in a sucrose and citrate formulation with a more desirable high glass transition temperature of the dried cake. An optimal residual moisture range (1-3%) was found to be critical to maintaining AAV8 stability. Glycerol was found to protect AAV8 from over-drying by preventing capsid damage and genome DNA release. A lyophilized formulation was identified that maintained potency for 24 months at 2-8 °C, indicating the feasibility of a dried formulation for AAV gene therapy.
Collapse
|
12
|
Croze RH, Kotterman M, Burns CH, Schmitt CE, Quezada M, Schaffer D, Kirn D, Francis P. Viral Vector Technologies and Strategies: Improving on Nature. Int Ophthalmol Clin 2021; 61:59-89. [PMID: 34196318 PMCID: PMC8253506 DOI: 10.1097/iio.0000000000000361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
13
|
Murray SJ, Russell KN, Melzer TR, Gray SJ, Heap SJ, Palmer DN, Mitchell NL. Intravitreal gene therapy protects against retinal dysfunction and degeneration in sheep with CLN5 Batten disease. Exp Eye Res 2021; 207:108600. [PMID: 33930398 DOI: 10.1016/j.exer.2021.108600] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 04/12/2021] [Accepted: 04/21/2021] [Indexed: 01/01/2023]
Abstract
Neuronal ceroid lipofuscinoses (NCL; Batten disease) are a group of inherited neurodegenerative diseases primarily affecting children. A common feature across most NCLs is the progressive loss of vision. We performed intravitreal injections of self-complementary AAV9 vectors packaged with either ovine CLN5 or CLN6 into one eye of 3-month-old CLN5-/- or CLN6-/- animals, respectively. Electroretinography (ERG) was performed every month following treatment, and retinal histology was assessed post-mortem in the treated compared to untreated eye. In CLN5-/- animals, ERG amplitudes were normalised in the treated eye whilst the untreated eye declined in a similar manner to CLN5 affected controls. In CLN6-/- animals, ERG amplitudes in both eyes declined over time although the treated eye showed a slower decline. Post-mortem examination revealed significant attenuation of retinal atrophy and lysosomal storage body accumulation in the treated eye compared with the untreated eye in CLN5-/- animals. This proof-of-concept study provides the first observation of efficacious intravitreal gene therapy in a large animal model of NCL. In particular, the single administration of AAV9-mediated intravitreal gene therapy can successfully ameliorate retinal deficits in CLN5-/- sheep. Combining ocular gene therapy with brain-directed therapy presents a promising treatment strategy to be used in future sheep trials aiming to halt neurological and retinal disease in CLN5 Batten disease.
Collapse
Affiliation(s)
- Samantha J Murray
- Faculty of Agricultural and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand
| | - Katharina N Russell
- Faculty of Agricultural and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand
| | - Tracy R Melzer
- Department of Medicine, University of Otago, Christchurch and the New Zealand Brain Research Institute, Christchurch, 8011, New Zealand
| | - Steven J Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Stephen J Heap
- McMaster & Heap Veterinary Practice, Christchurch, 8025, New Zealand
| | - David N Palmer
- Faculty of Agricultural and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand; Department of Radiology, University of Otago, Christchurch, 8140, New Zealand
| | - Nadia L Mitchell
- Faculty of Agricultural and Life Sciences, Lincoln University, Lincoln, 7647, New Zealand; Department of Radiology, University of Otago, Christchurch, 8140, New Zealand.
| |
Collapse
|
14
|
Rieser R, Koch J, Faccioli G, Richter K, Menzen T, Biel M, Winter G, Michalakis S. Comparison of Different Liquid Chromatography-Based Purification Strategies for Adeno-Associated Virus Vectors. Pharmaceutics 2021; 13:pharmaceutics13050748. [PMID: 34070226 PMCID: PMC8158740 DOI: 10.3390/pharmaceutics13050748] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 01/01/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) vectors have evolved as one of the most promising technologies for gene therapy due to their good safety profile, high transduction efficacy, and long-term gene expression in nondividing cells. rAAV-based gene therapy holds great promise for treating genetic disorders like inherited blindness, muscular atrophy, or bleeding disorders. There is a high demand for efficient and scalable production and purification methods for rAAVs. This is particularly true for the downstream purification methods. The current standard methods are based on multiple steps of gradient ultracentrifugation, which allow for the purification and enrichment of full rAAV particles, but the scale up of this method is challenging. Here, we explored fast, scalable, and universal liquid chromatography-based strategies for the purification of rAAVs. In contrast to the hydrophobic interaction chromatography (HIC), where a substantial amount of AAV was lost, the cation exchange chromatography (CEX) was performed robustly for multiple tested serotypes and resulted in a mixture of full and empty rAAVs with a good purity profile. For the used affinity chromatography (AC), a serotype dependence was observed. Anion exchange chromatography (AEX) worked well for the AAV8 serotype and achieved high levels of purification and a baseline separation of full and empty rAAVs. Depending on the AAV serotype, a combination of CEX and AEX or AC and AEX is recommended and holds promise for future translational projects that require highly pure and full particle-enriched rAAVs.
Collapse
Affiliation(s)
- Ruth Rieser
- Department of Pharmacy—Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany; (R.R.); (J.K.); (G.F.); (M.B.)
| | - Johanna Koch
- Department of Pharmacy—Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany; (R.R.); (J.K.); (G.F.); (M.B.)
| | - Greta Faccioli
- Department of Pharmacy—Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany; (R.R.); (J.K.); (G.F.); (M.B.)
| | - Klaus Richter
- Coriolis Pharma, Fraunhoferstr. 18 b, 82152 Martinsried, Germany; (K.R.); (T.M.)
| | - Tim Menzen
- Coriolis Pharma, Fraunhoferstr. 18 b, 82152 Martinsried, Germany; (K.R.); (T.M.)
| | - Martin Biel
- Department of Pharmacy—Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany; (R.R.); (J.K.); (G.F.); (M.B.)
| | - Gerhard Winter
- Department of Pharmacy—Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany; (R.R.); (J.K.); (G.F.); (M.B.)
- Correspondence: (G.W.); (S.M.)
| | - Stylianos Michalakis
- Department of Pharmacy—Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, Germany; (R.R.); (J.K.); (G.F.); (M.B.)
- Department of Ophthalmology, University Hospital, LMU Munich, Mathildenstr. 8, 80336 Munich, Germany
- Correspondence: (G.W.); (S.M.)
| |
Collapse
|
15
|
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
|
16
|
Korecki AJ, Cueva-Vargas JL, Fornes O, Agostinone J, Farkas RA, Hickmott JW, Lam SL, Mathelier A, Zhou M, Wasserman WW, Di Polo A, Simpson EM. Human MiniPromoters for ocular-rAAV expression in ON bipolar, cone, corneal, endothelial, Müller glial, and PAX6 cells. Gene Ther 2021; 28:351-372. [PMID: 33531684 PMCID: PMC8222000 DOI: 10.1038/s41434-021-00227-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/17/2020] [Accepted: 01/15/2021] [Indexed: 02/06/2023]
Abstract
Small and cell-type restricted promoters are important tools for basic and preclinical research, and clinical delivery of gene therapies. In clinical gene therapy, ophthalmic trials have been leading the field, with over 50% of ocular clinical trials using promoters that restrict expression based on cell type. Here, 19 human DNA MiniPromoters were bioinformatically designed for rAAV, tested by neonatal intravenous delivery in mouse, and successful MiniPromoters went on to be tested by intravitreal, subretinal, intrastromal, and/or intravenous delivery in adult mouse. We present promoter development as an overview for each cell type, but only show results in detail for the recommended MiniPromoters: Ple265 and Ple341 (PCP2) ON bipolar, Ple349 (PDE6H) cone, Ple253 (PITX3) corneal stroma, Ple32 (CLDN5) endothelial cells of the blood-retina barrier, Ple316 (NR2E1) Müller glia, and Ple331 (PAX6) PAX6 positive. Overall, we present a resource of new, redesigned, and improved MiniPromoters for ocular gene therapy that range in size from 784 to 2484 bp, and from weaker, equal, or stronger in strength relative to the ubiquitous control promoter smCBA. All MiniPromoters will be useful for therapies involving small regulatory RNA and DNA, and proteins ranging from 517 to 1084 amino acids, representing 62.9-90.2% of human proteins.
Collapse
Affiliation(s)
- Andrea J. Korecki
- grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics at BC Children’s Hospital, University of British Columbia, Vancouver, BC Canada
| | - Jorge L. Cueva-Vargas
- grid.14848.310000 0001 2292 3357Department of Neuroscience, University of Montreal Hospital Research Centre, University of Montreal, Montreal, QC Canada
| | - Oriol Fornes
- grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics at BC Children’s Hospital, University of British Columbia, Vancouver, BC Canada
| | - Jessica Agostinone
- grid.14848.310000 0001 2292 3357Department of Neuroscience, University of Montreal Hospital Research Centre, University of Montreal, Montreal, QC Canada
| | - Rachelle A. Farkas
- grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics at BC Children’s Hospital, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC Canada
| | - Jack W. Hickmott
- grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics at BC Children’s Hospital, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC Canada
| | - Siu Ling Lam
- grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics at BC Children’s Hospital, University of British Columbia, Vancouver, BC Canada
| | - Anthony Mathelier
- grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics at BC Children’s Hospital, University of British Columbia, Vancouver, BC Canada
| | - Michelle Zhou
- grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics at BC Children’s Hospital, University of British Columbia, Vancouver, BC Canada
| | - Wyeth W. Wasserman
- grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics at BC Children’s Hospital, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC Canada
| | - Adriana Di Polo
- grid.14848.310000 0001 2292 3357Department of Neuroscience, University of Montreal Hospital Research Centre, University of Montreal, Montreal, QC Canada
| | - Elizabeth M. Simpson
- grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics at BC Children’s Hospital, University of British Columbia, Vancouver, BC Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC Canada
| |
Collapse
|
17
|
Gao M, Liu H, Xiao Y, Guo Y, Wan X, Li X, Li M, Liang J, Zhai Y, Liu W, Jiang M, Luo X, Sun X. xCT regulates redox homeostasis and promotes photoreceptor survival after retinal detachment. Free Radic Biol Med 2020; 158:32-43. [PMID: 32679366 DOI: 10.1016/j.freeradbiomed.2020.06.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/17/2020] [Accepted: 06/08/2020] [Indexed: 01/20/2023]
Abstract
BACKGROUNDS Photoreceptor degeneration underlies various retinal disorders that lead to vision impairment. Currently, no effective medication is available to rescue photoreceptors under disease conditions. Elucidation of the molecular pathways involved in photoreceptor degeneration is a prerequisite for the rational design of therapeutic interventions. Photoreceptors are among the most energy-demanding tissues that require highly active oxidative phosphorylation. Therefore, disruption of metabolic support to photoreceptors results in a redox imbalance and subsequent cell death. We hypothesize that the redox regulatory pathway could be a potential therapeutic target to rescue photoreceptors under disease conditions. METHODS Experimental retinal detachment was induced in mice. A murine photoreceptor-derived 661w cell line treated with H2O2 was employed as an in vitro model to study the cellular response to oxidative stress. The expression and functional role of xCT, an upstream regulator of redox homeostasis, was assessed in vivo and in vitro. An xCT expression vector was constructed for an in vivo study to evaluate the therapeutic potential of this molecule. RESULTS xCT expression was upregulated in detached retina and H2O2-stimulated 661w cells compared to the control cells. Pharmacological inhibition of xCT by sulfasalazine (SAS) promoted photoreceptor degeneration after retinal detachment and 661w cell death upon H2O2 treatment. Additionally, SAS treatment induced reactive oxidative species (ROS) accumulation, glutathione (GSH) depletion, and glutamate release in 661w cells. In contrast, xCT overexpression via viral infection protected photoreceptors from degeneration after retinal detachment. CONCLUSION We conclude that xCT expression is upregulated in photoreceptors after retinal detachment and plays a neuroprotective role in preserving photoreceptors. Mechanistically, xCT promotes cellular homeostasis by regulating intracellular ROS and GSH levels, which are critical to photoreceptor survival after retinal detachment. Collectively, our findings identify xCT as a potential therapeutic target for protection of photoreceptors under disease conditions.
Collapse
Affiliation(s)
- Min Gao
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, 200080, Shanghai, China
| | - Haiyun Liu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, 200080, Shanghai, China
| | - Yushu Xiao
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, 200080, Shanghai, China
| | - Yinong Guo
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, 200080, Shanghai, China
| | - Xiaoling Wan
- Shanghai Key Laboratory of Fundus Diseases, 200080, Shanghai, China
| | - Xiaomeng Li
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, 200080, Shanghai, China
| | - Min Li
- Shanghai Key Laboratory of Fundus Diseases, 200080, Shanghai, China
| | - Jian Liang
- Shanghai Key Laboratory of Fundus Diseases, 200080, Shanghai, China
| | - Yuanqi Zhai
- Shanghai Key Laboratory of Fundus Diseases, 200080, Shanghai, China
| | - Wenjia Liu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, 200080, Shanghai, China
| | - Mei Jiang
- Shanghai Key Laboratory of Fundus Diseases, 200080, Shanghai, China
| | - Xueting Luo
- Shanghai Key Laboratory of Fundus Diseases, 200080, Shanghai, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, School of Medicine, 200080, Shanghai, China; Shanghai Key Laboratory of Fundus Diseases, 200080, Shanghai, China; Shanghai Engineering Center for Visual Science and Photomedicine, 200080, Shanghai, China.
| |
Collapse
|
18
|
Liu Y, Hegarty S, Winter C, Wang F, He Z. Viral vectors for neuronal cell type-specific visualization and manipulations. Curr Opin Neurobiol 2020; 63:67-76. [PMID: 32344323 DOI: 10.1016/j.conb.2020.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/13/2020] [Accepted: 03/24/2020] [Indexed: 12/19/2022]
Abstract
Characterizing neuronal cell types demands efficient strategies for specific labeling and manipulation of individual subtypes to dissect their connectivity and functions. Recombinant viral technology offers a powerful toolbox for targeted transgene expression in specific neuronal populations. In order to achieve cell type-specific targeting, exciting progress has been made to: alter viral tropisms, design rational delivery strategies, and drive selective expression patterns with engineered DNA sequences in viral genomes. For the latter case, emerging single-cell genomic analyses provide rich databases. In this review, we will summarize current status, and point out challenges, of using viral vectors for neuronal cell type-specific visualization and manipulations. With concerted efforts, progress will continue to be made toward developing viral vectors for the vast array of neuronal subtypes in the mammalian nervous system.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Somatosensation and Pain Unit, National Institute of Dental and Craniofacial Research (NIDCR), National Center for Complementary and Integrative Health (NCCIH), National Institutes of Health (NIH), MD, USA
| | - Shane Hegarty
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Carla Winter
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA; PhD Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Fan Wang
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Zhigang He
- F. M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Mouse γ-Synuclein Promoter-Mediated Gene Expression and Editing in Mammalian Retinal Ganglion Cells. J Neurosci 2020; 40:3896-3914. [PMID: 32300046 DOI: 10.1523/jneurosci.0102-20.2020] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/28/2020] [Accepted: 04/02/2020] [Indexed: 12/21/2022] Open
Abstract
Optic neuropathies are a group of optic nerve (ON) diseases caused by various insults including glaucoma, inflammation, ischemia, trauma, and genetic deficits, which are characterized by retinal ganglion cell (RGC) death and ON degeneration. An increasing number of genes involved in RGC intrinsic signaling have been found to be promising neural repair targets that can potentially be modulated directly by gene therapy, if we can achieve RGC specific gene targeting. To address this challenge, we first used adeno-associated virus (AAV)-mediated gene transfer to perform a low-throughput in vivo screening in both male and female mouse eyes and identified the mouse γ-synuclein (mSncg) promoter, which specifically and potently sustained transgene expression in mouse RGCs and also works in human RGCs. We further demonstrated that gene therapy that combines AAV-mSncg promoter with clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing can knock down pro-degenerative genes in RGCs and provide effective neuroprotection in optic neuropathies.SIGNIFICANCE STATEMENT Here, we present an RGC-specific promoter, mouse γ-synuclein (mSncg) promoter, and perform extensive characterization and proof-of-concept studies of mSncg promoter-mediated gene expression and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing in RGCs in vivo To our knowledge, this is the first report demonstrating in vivo neuroprotection of injured RGCs and optic nerve (ON) by AAV-mediated CRISPR/Cas9 inhibition of genes that are critical for neurodegeneration. It represents a powerful tool to achieve RGC-specific gene modulation, and also opens up a promising gene therapy strategy for optic neuropathies, the most common form of eye diseases that cause irreversible blindness.
Collapse
|
20
|
Liu YF, Huang S, Ng TK, Liang JJ, Xu Y, Chen SL, Xu C, Zhang M, Pang CP, Cen LP. Longitudinal evaluation of immediate inflammatory responses after intravitreal AAV2 injection in rats by optical coherence tomography. Exp Eye Res 2020; 193:107955. [PMID: 32017940 DOI: 10.1016/j.exer.2020.107955] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/22/2020] [Accepted: 01/31/2020] [Indexed: 02/05/2023]
Abstract
Gene therapy has been proposed as a feasible strategy for RGC survival and optic nerve regeneration. Some preclinical and clinical studies revealed intraocular inflammation after intravitreal injection of adeno-associated virus (AAV) by slit-lamp or indirect ophthalmoscope. Here we evaluate the longitudinal profile of immediate inflammatory responses after AAV2 injection in rat retina and vitreous body by optical coherence tomography (OCT). Adult Fischer F344 rats were intravitreally injected once with saline, AAV2 or zymosan. Retinal thickness and cell infiltration were recorded by OCT longitudinally for 2 months and verified by histological analysis. The transduction rate of single intravitreal AAV2 injection was 21.3 ± 4.9% of whole retina, and the transduction efficiency on RGCs was 91.5 ± 2.5% in the transduced area. Significant increase in cell infiltration was observed from Day 1-3 after AAV2 injection, compared to very few infiltrating cells observed in the saline-injected group. The infiltrating cells ceased at Day 5 after intravitreal injection and remained absent at 2 months. The thicknesses of total and inner retina were increased along Day 1-3 after AAV2 injection, but reverted to normal afterwards. The surviving RGCs in the AAV2-injected groups at Day 14 showed no significant difference compared to saline-injected group. In summary, this study revealed the immediate inflammatory responses and retinal edema after intravitreal AAV2 injection in normal rats, without influencing long-term retinal thickness and RGC survival. OCT can be implemented for the time-lapse in vivo evaluation of inflammatory response after AAV-mediated gene therapy through intravitreal injection.
Collapse
Affiliation(s)
- Yu-Fen Liu
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China; Shantou University Medical College, Shantou, Guangdong, China
| | - Shaofen Huang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China; Shantou University Medical College, Shantou, Guangdong, China; Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Jia-Jian Liang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Yanxuan Xu
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Shao-Lang Chen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Ciyan Xu
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Mingzhi Zhang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
| | - Chi Pui Pang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China; Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Ling-Ping Cen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China.
| |
Collapse
|
21
|
Rieser R, Penaud-Budloo M, Bouzelha M, Rossi A, Menzen T, Biel M, Büning H, Ayuso E, Winter G, Michalakis S. Intrinsic Differential Scanning Fluorimetry for Fast and Easy Identification of Adeno-Associated Virus Serotypes. J Pharm Sci 2020; 109:854-862. [DOI: 10.1016/j.xphs.2019.10.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/23/2019] [Accepted: 10/11/2019] [Indexed: 12/14/2022]
|
22
|
Augustine J, Troendle EP, Barabas P, McAleese CA, Friedel T, Stitt AW, Curtis TM. The Role of Lipoxidation in the Pathogenesis of Diabetic Retinopathy. Front Endocrinol (Lausanne) 2020; 11:621938. [PMID: 33679605 PMCID: PMC7935543 DOI: 10.3389/fendo.2020.621938] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/21/2020] [Indexed: 12/31/2022] Open
Abstract
Lipids can undergo modification as a result of interaction with reactive oxygen species (ROS). For example, lipid peroxidation results in the production of a wide variety of highly reactive aldehyde species which can drive a range of disease-relevant responses in cells and tissues. Such lipid aldehydes react with nucleophilic groups on macromolecules including phospholipids, nucleic acids, and proteins which, in turn, leads to the formation of reversible or irreversible adducts known as advanced lipoxidation end products (ALEs). In the setting of diabetes, lipid peroxidation and ALE formation has been implicated in the pathogenesis of macro- and microvascular complications. As the most common diabetic complication, retinopathy is one of the leading causes of vision loss and blindness worldwide. Herein, we discuss diabetic retinopathy (DR) as a disease entity and review the current knowledge and experimental data supporting a role for lipid peroxidation and ALE formation in the onset and development of this condition. Potential therapeutic approaches to prevent lipid peroxidation and lipoxidation reactions in the diabetic retina are also considered, including the use of antioxidants, lipid aldehyde scavenging agents and pharmacological and gene therapy approaches for boosting endogenous aldehyde detoxification systems. It is concluded that further research in this area could lead to new strategies to halt the progression of DR before irreversible retinal damage and sight-threatening complications occur.
Collapse
Affiliation(s)
- Josy Augustine
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University of Belfast, Belfast, United Kingdom
| | - Evan P. Troendle
- Department of Chemistry, King’s College London, London, United Kingdom
| | - Peter Barabas
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University of Belfast, Belfast, United Kingdom
| | - Corey A. McAleese
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University of Belfast, Belfast, United Kingdom
| | - Thomas Friedel
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University of Belfast, Belfast, United Kingdom
| | - Alan W. Stitt
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University of Belfast, Belfast, United Kingdom
| | - Tim M. Curtis
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen’s University of Belfast, Belfast, United Kingdom
- *Correspondence: Tim M. Curtis,
| |
Collapse
|
23
|
Vázquez-Domínguez I, Garanto A, Collin RWJ. Molecular Therapies for Inherited Retinal Diseases-Current Standing, Opportunities and Challenges. Genes (Basel) 2019; 10:genes10090654. [PMID: 31466352 PMCID: PMC6770110 DOI: 10.3390/genes10090654] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 12/15/2022] Open
Abstract
Inherited retinal diseases (IRDs) are both genetically and clinically highly heterogeneous and have long been considered incurable. Following the successful development of a gene augmentation therapy for biallelic RPE65-associated IRD, this view has changed. As a result, many different therapeutic approaches are currently being developed, in particular a large variety of molecular therapies. These are depending on the severity of the retinal degeneration, knowledge of the pathophysiological mechanism underlying each subtype of IRD, and the therapeutic target molecule. DNA therapies include approaches such as gene augmentation therapy, genome editing and optogenetics. For some genetic subtypes of IRD, RNA therapies and compound therapies have also shown considerable therapeutic potential. In this review, we summarize the current state-of-the-art of various therapeutic approaches, including the pros and cons of each strategy, and outline the future challenges that lie ahead in the combat against IRDs.
Collapse
Affiliation(s)
- Irene Vázquez-Domínguez
- Department of Human Genetics and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands
| | - Alejandro Garanto
- Department of Human Genetics and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands.
| | - Rob W J Collin
- Department of Human Genetics and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands.
| |
Collapse
|
24
|
Völkner M, Kurth T, Karl MO. The Mouse Retinal Organoid Trisection Recipe: Efficient Generation of 3D Retinal Tissue from Mouse Embryonic Stem Cells. Methods Mol Biol 2019; 1834:119-141. [PMID: 30324441 DOI: 10.1007/978-1-4939-8669-9_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
The introduction of stem cell-based technologies for the derivation of three-dimensional retinal tissues, the so-called retinal organoids, offers many new possibilities for vision research: Organoids facilitate studies on retinal development and in vitro retinal disease modeling, as well as being valuable for drug testing. Further, retinal organoids also provide an unlimited cell source for cell replacement therapies. Here, we describe our protocol for efficiently differentiating large, stratified retinal organoids from mouse embryonic stem cells: unbiased manual dissection of the developing retinal organoid at an early stage into three evenly sized neuroepithelial portions (trisection step) doubles the yield of high-quality organoids. We also describe some useful applications of the protocol, e.g., generation of rod- or cone-enriched retinal organoids, AAV transfection, and cell birth dating. In addition, we provide details of how to process retinal organoids for single organoid gene expression analysis, immunohistochemistry, and electron microscopy.
Collapse
Affiliation(s)
- Manuela Völkner
- German Center for Neurodegenerative Diseases Dresden (DZNE), Dresden, Germany
| | - Thomas Kurth
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Mike O Karl
- German Center for Neurodegenerative Diseases Dresden (DZNE), Dresden, Germany. .,Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany.
| |
Collapse
|
25
|
Devoldere J, Peynshaert K, De Smedt SC, Remaut K. Müller cells as a target for retinal therapy. Drug Discov Today 2019; 24:1483-1498. [PMID: 30731239 DOI: 10.1016/j.drudis.2019.01.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/20/2018] [Accepted: 01/30/2019] [Indexed: 12/28/2022]
Abstract
Müller cells are specialized glial cells that span the entire retina from the vitreous cavity to the subretinal space. Their functional diversity and unique radial morphology render them particularly interesting targets for new therapeutic approaches. In this review, we reflect on various possibilities for selective Müller cell targeting and describe how some of their cellular mechanisms can be used for retinal neuroprotection. Intriguingly, cross-species investigation of their properties has revealed that Müller cells also have an essential role in retinal regeneration. Although many questions regarding this subject remain, it is clear that Müller cells have unique characteristics that make them suitable targets for the prevention and treatment of numerous 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
| | - 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
|
26
|
Wagner JE, Schön C, Becirovic E, Biel M, Michalakis S. In Vitro Evaluation of AAV Vectors for Retinal Gene Therapy. Methods Mol Biol 2019; 1834:383-390. [PMID: 30324456 DOI: 10.1007/978-1-4939-8669-9_24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Gene therapy holds promise for treating previously untreatable retinal disorders. The most promising approaches use gene transfer vectors derived from adeno-associated virus (AAV) to supplement a gene function in the affected cell type. One example is gene therapy for achromatopsia which affects daylight vision. In this case, recombinant AAV (rAAV) vectors are being developed to specifically target cone photoreceptors. Development of rAAV vectors could be facilitated by the use of in vitro models. In this chapter we provide a protocol which utilizes mouse 661W cells, an in vitro model of cone photoreceptors for evaluation of the transduction efficacy of rAAV vectors.
Collapse
Affiliation(s)
- Johanna E Wagner
- Department of Pharmacy-Center for Drug Research, Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Schön
- Department of Pharmacy-Center for Drug Research, Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elvir Becirovic
- Department of Pharmacy-Center for Drug Research, Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Biel
- Department of Pharmacy-Center for Drug Research, Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stylianos Michalakis
- Department of Pharmacy-Center for Drug Research, Center for Integrated Protein Science Munich (CiPSM), Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
27
|
Mühlfriedel R, Michalakis S, Garrido MG, Sothilingam V, Schön C, Biel M, Seeliger MW. Optimized Subretinal Injection Technique for Gene Therapy Approaches. Methods Mol Biol 2019; 1834:405-412. [PMID: 30324458 DOI: 10.1007/978-1-4939-8669-9_26] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Gene therapy for inherited eye diseases requires local viral vector delivery by intraocular injection. Since large animal models are lacking for most of these diseases, genetically modified mouse models are commonly used in preclinical proof-of-concept studies. However, because of the relatively small mouse eye, adverse effects of the subretinal delivery procedure itself may interfere with the therapeutic outcome. The method described here aims to provide the details relevant to perform a transscleral pars plana virus-mediated gene transfer to achieve an optimized therapeutic effect in the small mouse eye.
Collapse
Affiliation(s)
- Regine Mühlfriedel
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, Eberhard Karls Universität Tübingen, Tübingen, Germany.
| | - Stylianos Michalakis
- Department of Pharmacy-Center for Drug Research, Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marina Garcia Garrido
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Vithiyanjali Sothilingam
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Christian Schön
- Department of Pharmacy-Center for Drug Research, Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Biel
- Department of Pharmacy-Center for Drug Research, Center for Integrated Protein Science Munich (CIPSM), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mathias W Seeliger
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| |
Collapse
|
28
|
Rodrigues GA, Shalaev E, Karami TK, Cunningham J, Slater NKH, Rivers HM. Pharmaceutical Development of AAV-Based Gene Therapy Products for the Eye. Pharm Res 2018; 36:29. [PMID: 30591984 PMCID: PMC6308217 DOI: 10.1007/s11095-018-2554-7] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/30/2018] [Indexed: 12/18/2022]
Abstract
A resurgence of interest and investment in the field of gene therapy, driven in large part by advances in viral vector technology, has recently culminated in United States Food and Drug Administration approval of the first gene therapy product targeting a disease caused by mutations in a single gene. This product, LUXTURNA™ (voretigene neparvovec-rzyl; Spark Therapeutics, Inc., Philadelphia, PA), delivers a normal copy of the RPE65 gene to retinal cells for the treatment of biallelic RPE65 mutation–associated retinal dystrophy, a blinding disease. Many additional gene therapy programs targeting both inherited retinal diseases and other ocular diseases are in development, owing to an improved understanding of the genetic basis of ocular disease and the unique properties of the ocular compartment that make it amenable to local gene therapy. Here we review the growing body of literature that describes both the design and development of ocular gene therapy products, with a particular emphasis on target and vector selection, and chemistry, manufacturing, and controls.
Collapse
Affiliation(s)
| | - Evgenyi Shalaev
- Pharmaceutical Research and Development, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612-1531, USA
| | - Thomas K Karami
- Pharmaceutical Research and Development, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612-1531, USA
| | - James Cunningham
- Pharmaceutical Research and Development, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612-1531, USA
| | - Nigel K H Slater
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, CB3 0AS, UK
| | - Hongwen M Rivers
- Pharmaceutical Research and Development, Allergan plc, 2525 Dupont Drive, Irvine, California, 92612-1531, USA.
| |
Collapse
|
29
|
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
|
30
|
Gao YG, Alam U, Ding AX, Tang Q, Tan ZL, Shi YD, Lu ZL, Qian AR. [12]aneN3-based lipid with naphthalimide moiety for enhanced gene transfection efficiency. Bioorg Chem 2018; 79:334-340. [DOI: 10.1016/j.bioorg.2018.04.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/16/2018] [Accepted: 04/24/2018] [Indexed: 01/02/2023]
|
31
|
Ezra-Elia R, Obolensky A, Ejzenberg A, Ross M, Mintz D, Banin E, Ofri R. Can an in vivo imaging system be used to determine localization and biodistribution of AAV5-mediated gene expression following subretinal and intravitreal delivery in mice? Exp Eye Res 2018; 176:227-234. [PMID: 30171858 DOI: 10.1016/j.exer.2018.08.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/24/2018] [Accepted: 08/28/2018] [Indexed: 02/07/2023]
Abstract
Recombinant adeno associated viruses (AAV) are the most commonly used vectors in animal model studies of gene therapy for retinal diseases. The ability of a vector to localize and remain in the target tissue, and in this manner to avoid off-target effects beyond the site of delivery, is critical to the efficacy and safety of the treatment. The in vivo imaging system (IVIS) is a non-invasive imaging tool used for detection and quantification of bioluminescence activity in rodents. Our aim was to investigate whether IVIS can detect localization and biodistribution of AAV5 vector in mice following subretinal (SR) and intravitreal (IVT) injections. AAV5 carrying firefly luciferase DNA under control of the ubiquitous cytomegalovirus (CMV) promoter was injected unilaterally IVT or SR (in the central or peripheral retina) of forty-one mice. Luciferase activity was tracked for up to 60 weeks in the longest surviving animals, using repeated (up to 12 times) IVIS bioluminescence imaging. Luciferase presence was also confirmed immunohistochemically (IHC) and by PCR in representative animals. In the SR group, IVIS readings demonstrated luciferase activity in all (32/32) eyes, and luciferase presence was confirmed by IHC (4/4 eyes) and PCR (12/12 eyes). In the IVT group, IVIS readings demonstrated luciferase activity in 7/9 eyes, and luciferase presence was confirmed by PCR in 5/5 eyes and by IHC (2/2 eyes). In two SR-injected animals (one each from the central and peripheral injection sites), PCR detected luciferase presence in the ipsilateral optic nerves, a finding that was not detected by IVIS or IHC. Our results show that when evaluating SR delivery, IVIS has a sensitivity and specificity of 100% compared with the gold standard PCR. When evaluating IVT delivery, IVIS has a sensitivity of 78% and specificity of 100%. These finding confirm the ability of IVIS to detect in-vivo localized expression of AAV following SR delivery in the retina up to 60 weeks post-treatment, using repeated imaging for longitudinal evaluation, without fading of the biological signal, thereby replacing the need for post mortem processing in order to confirm vector expression. However, IVIS is probably not sensitive enough, compared with genome detection, to demonstrate biodistribution to the optic nerve, as it could not detect luciferase activity in ipsilateral optic nerves following SR delivery in mice.
Collapse
Affiliation(s)
- Raaya Ezra-Elia
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Alexey Obolensky
- Center for Retinal and Macular Degenerations (CRMD), Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ayala Ejzenberg
- Center for Retinal and Macular Degenerations (CRMD), Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Maya Ross
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Dvir Mintz
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Eyal Banin
- Center for Retinal and Macular Degenerations (CRMD), Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ron Ofri
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel.
| |
Collapse
|
32
|
Bosco A, Anderson SR, Breen KT, Romero CO, Steele MR, Chiodo VA, Boye SL, Hauswirth WW, Tomlinson S, Vetter ML. Complement C3-Targeted Gene Therapy Restricts Onset and Progression of Neurodegeneration in Chronic Mouse Glaucoma. Mol Ther 2018; 26:2379-2396. [PMID: 30217731 DOI: 10.1016/j.ymthe.2018.08.017] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 08/02/2018] [Accepted: 08/19/2018] [Indexed: 12/16/2022] Open
Abstract
Dysregulation of the complement system is implicated in neurodegeneration, including human and animal glaucoma. Optic nerve and retinal damage in glaucoma is preceded by local complement upregulation and activation, but whether targeting this early innate immune response could have therapeutic benefit remains undefined. Because complement signals through three pathways that intersect at complement C3 activation, here we targeted this step to restore complement balance in the glaucomatous retina and to determine its contribution to degeneration onset and/or progression. To achieve this, we combined adeno-associated virus retinal gene therapy with the targeted C3 inhibitor CR2-Crry. We show that intravitreal injection of AAV2.CR2-Crry produced sustained Crry overexpression in the retina and reduced deposition of the activation product complement C3d on retinal ganglion cells and the inner retina of DBA/2J mice. This resulted in neuroprotection of retinal ganglion cell axons and somata despite continued intraocular pressure elevation, suggesting a direct restriction of neurodegeneration onset and progression and significant delay to terminal disease stages. Our study uncovers a damaging effect of complement C3 or downstream complement activation in glaucoma, and it establishes AAV2.CR2-Crry as a viable therapeutic strategy to target pathogenic C3-mediated complement activation in the glaucomatous retina.
Collapse
Affiliation(s)
- Alejandra Bosco
- Department of Neurobiology and Anatomy, School of Medicine, University of Utah, Salt Lake City, UT, USA.
| | - Sarah R Anderson
- Department of Neurobiology and Anatomy, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Kevin T Breen
- Department of Neurobiology and Anatomy, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Cesar O Romero
- Department of Neurobiology and Anatomy, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Michael R Steele
- Department of Neurobiology and Anatomy, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Vince A Chiodo
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | | | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Monica L Vetter
- Department of Neurobiology and Anatomy, School of Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
33
|
Cell-specific gene therapy driven by an optimized hypoxia-regulated vector reduces choroidal neovascularization. J Mol Med (Berl) 2018; 96:1107-1118. [PMID: 30105447 DOI: 10.1007/s00109-018-1683-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 07/31/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022]
Abstract
Aberrant growth of blood vessels in the choroid layer of the eye, termed choroidal neovascularization (CNV), is the pathological hallmark of exudative age-related macular degeneration (AMD), causing irreversible blindness among the elderly. Co-localization of proangiogenic factors and hypoxia inducible factors (HIF) in neovascular membranes from AMD eyes suggests the role of hypoxia in pathogenesis of CNV. In order to utilize hypoxic conditions in RPE for therapeutic purposes, we developed an optimized hypoxia regulated, RPE cell-specific gene therapy to inhibit choroidal neovascularization. An adeno-associated virus (AAV2) vector comprising a RPE-specific promoter and HIF-1 response elements (HRE) was designed to regulate production of human endostatin (a powerful angiostatic protein) in RPE. The vector was tested in a mouse model of laser-induced CNV using subretinal delivery. Spectral domain optical coherence tomography (SD-OCT) images from live mice and confocal images from lectin stained RPE flat mount sections demonstrated reduction in CNV areas by 80% compared to untreated eyes. Quantitative real-time polymerase chain reaction (qPCR) confirmed exogenous endostatin mRNA expression from the regulated vector that was significantly elevated 3, 7, and 14 days following laser treatment, but its expression was completely shut off after 45 days. Thus, RPE-specific, hypoxia-regulated delivery of anti-angiogenic proteins could be a valuable therapeutic approach to treat neovascular AMD at the time and in the ocular space where it arises. KEY POINTS An optimized gene therapy vector targeting hypoxia and tissue-specific expression has been designed. The inhibitory role of gene therapy vector was tested in a mouse model of laser-induced CNV. An 80% reduction in choroidal neovascularization was achieved by the optimized vector. The expression of endostatin was limited to retinal pigment epithelium and regulated by hypoxia.
Collapse
|
34
|
Teo KYC, Lee SY, Barathi AV, Tun SBB, Tan L, Constable IJ. Surgical Removal of Internal Limiting Membrane and Layering of AAV Vector on the Retina Under Air Enhances Gene Transfection in a Nonhuman Primate. ACTA ACUST UNITED AC 2018; 59:3574-3583. [DOI: 10.1167/iovs.18-24333] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Kelvin Yi Chong Teo
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Shu Yen Lee
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Amutha Veluchamy Barathi
- Translational Pre-clinical Model Platform, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sai Bo Bo Tun
- Translational Pre-clinical Model Platform, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Licia Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Ian Jeffery Constable
- Centre of Ophthalmology and Visual Science (incorporating Lions Eye Institute), The University of Western Australia, Perth, Western Australia
- Department of Ophthalmology, Sir Charles Gairdner Hospital, Perth, Western Australia
| |
Collapse
|
35
|
Sun S, Schaffer DV. Engineered viral vectors for functional interrogation, deconvolution, and manipulation of neural circuits. Curr Opin Neurobiol 2018; 50:163-170. [PMID: 29614429 PMCID: PMC5984719 DOI: 10.1016/j.conb.2017.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/27/2017] [Accepted: 12/16/2017] [Indexed: 12/19/2022]
Abstract
Optimization of traditional replication-competent viral tracers has granted access to immediate synaptic partners of target neuronal populations, enabling the dissection of complex brain circuits into functional neural pathways. The excessive virulence of most conventional tracers, however, impedes their utility in revealing and genetically perturbing cellular function on long time scales. As a promising alternative, the natural capacity of adeno-associated viral (AAV) vectors to safely mediate persistent and robust gene expression has stimulated strong interest in adapting them for sparse neuronal labeling and physiological studies. Furthermore, increasingly refined engineering strategies have yielded novel AAV variants with enhanced target specificity, transduction, and retrograde trafficking in the CNS. These potent vectors offer new opportunities for characterizing the identity and connectivity of single neurons within immense networks and modulating their activity via robust delivery of functional genetic tools.
Collapse
Affiliation(s)
- Sabrina Sun
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA; Department of Bioengineering, University of California, Berkeley, CA, USA; The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| |
Collapse
|
36
|
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: 21] [Impact Index Per Article: 3.0] [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
|
37
|
Carvalho LS, Xiao R, Wassmer SJ, Langsdorf A, Zinn E, Pacouret S, Shah S, Comander JI, Kim LA, Lim L, Vandenberghe LH. Synthetic Adeno-Associated Viral Vector Efficiently Targets Mouse and Nonhuman Primate Retina In Vivo. Hum Gene Ther 2018; 29:771-784. [PMID: 29325457 DOI: 10.1089/hum.2017.154] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Gene therapy is a promising approach in the treatment of inherited and common complex disorders of the retina. Preclinical and clinical studies have validated the use of adeno-associated viral vectors (AAV) as a safe and efficient delivery vehicle for gene transfer. Retinal pigment epithelium and rods-and to a lesser extent, cone photoreceptors-can be efficiently targeted with AAV. Other retinal cell types however are more challenging targets. The aim of this study was to characterize the transduction profile and efficiency of in silico designed, synthetic Anc80 AAVs for retinal gene transfer. Three Anc80 variants were evaluated for retinal targeting in mice and primates following subretinal delivery. In the murine retina Anc80L65 demonstrated high level of retinal pigment epithelium and photoreceptor targeting with comparable cone photoreceptor affinity compared to other AAVs. Remarkably, Anc80L65 enhanced transduction kinetics with visible expression as early as day 1 and steady state mRNA levels at day 3. Inner retinal tropism of Anc80 variants demonstrated distinct transduction patterns of Müller glia, retinal ganglion cells and inner nuclear layer neurons. Finally, murine findings with Anc80L65 qualitatively translated to the Rhesus macaque in terms of cell targets, levels and onset of expression. Our findings support the use of Anc80L65 for therapeutic subretinal gene delivery.
Collapse
Affiliation(s)
- Livia S Carvalho
- 1 Grousbeck Gene Therapy Center, Boston, Massachusetts.,2 Ocular Genomics Institute , Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.,3 Schepens Eye Research Institute, Boston, Massachusetts.,4 Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Ru Xiao
- 1 Grousbeck Gene Therapy Center, Boston, Massachusetts.,2 Ocular Genomics Institute , Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.,3 Schepens Eye Research Institute, Boston, Massachusetts.,4 Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Sarah J Wassmer
- 1 Grousbeck Gene Therapy Center, Boston, Massachusetts.,2 Ocular Genomics Institute , Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.,3 Schepens Eye Research Institute, Boston, Massachusetts.,4 Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Aliete Langsdorf
- 2 Ocular Genomics Institute , Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.,4 Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Eric Zinn
- 1 Grousbeck Gene Therapy Center, Boston, Massachusetts.,2 Ocular Genomics Institute , Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.,3 Schepens Eye Research Institute, Boston, Massachusetts.,4 Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Simon Pacouret
- 1 Grousbeck Gene Therapy Center, Boston, Massachusetts.,2 Ocular Genomics Institute , Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.,3 Schepens Eye Research Institute, Boston, Massachusetts.,4 Massachusetts Eye and Ear Infirmary, Boston, Massachusetts.,6 INSERM UMR 1089, University of Nantes, Nantes University Hospital , Nantes, France
| | - Samiksha Shah
- 1 Grousbeck Gene Therapy Center, Boston, Massachusetts.,2 Ocular Genomics Institute , Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.,3 Schepens Eye Research Institute, Boston, Massachusetts.,4 Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Jason I Comander
- 2 Ocular Genomics Institute , Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.,4 Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Leo A Kim
- 3 Schepens Eye Research Institute, Boston, Massachusetts.,4 Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Laurence Lim
- 4 Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Luk H Vandenberghe
- 1 Grousbeck Gene Therapy Center, Boston, Massachusetts.,2 Ocular Genomics Institute , Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.,3 Schepens Eye Research Institute, Boston, Massachusetts.,4 Massachusetts Eye and Ear Infirmary, Boston, Massachusetts.,5 Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
38
|
Abstract
The first step in vision is the absorption of photons by the photopigments in cone and rod photoreceptors. After initial amplification within the phototransduction cascade the signal is translated into an electrical signal by the action of cyclic nucleotide-gated (CNG) channels. CNG channels are ligand-gated ion channels that are activated by the binding of cyclic guanosine monophosphate (cGMP) or cyclic adenosine monophosphate (cAMP). Retinal CNG channels transduce changes in intracellular concentrations of cGMP into changes of the membrane potential and the Ca2+ concentration. Structurally, the CNG channels belong to the superfamily of pore-loop cation channels and share a common gross structure with hyperpolarization-activated cyclic nucleotide-gated (HCN) channels and voltage-gated potassium channels (KCN). In this review, we provide an overview on the molecular properties of CNG channels and describe their physiological role in the phototransduction pathways. We also discuss insights into the pathophysiological role of CNG channel proteins that have emerged from the analysis of CNG channel-deficient animal models and human CNG channelopathies. Finally, we summarize recent gene therapy activities and provide an outlook for future clinical application.
Collapse
Affiliation(s)
- Stylianos Michalakis
- Center for Integrated Protein Science Munich (CIPSM), Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr, 5-13, 81377 Munich, Germany.
| | - Elvir Becirovic
- Center for Integrated Protein Science Munich (CIPSM), Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr, 5-13, 81377 Munich, Germany.
| | - Martin Biel
- Center for Integrated Protein Science Munich (CIPSM), Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr, 5-13, 81377 Munich, Germany.
| |
Collapse
|
39
|
Zhang C, Zhou X, Yao T, Tian Z, Zhou D. Precision Fluorescent Labeling of an Adeno-Associated Virus Vector to Monitor the Viral Infection Pathway. Biotechnol J 2018; 13:e1700374. [DOI: 10.1002/biot.201700374] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 12/04/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Chuanling Zhang
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University; Beijing 100191 China
| | - Xueying Zhou
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University; Beijing 100191 China
| | - Tianzhuo Yao
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University; Beijing 100191 China
| | - Zhenyu Tian
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University; Beijing 100191 China
| | - Demin Zhou
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University; Beijing 100191 China
| |
Collapse
|
40
|
Awwad S, Mohamed Ahmed AHA, Sharma G, Heng JS, Khaw PT, Brocchini S, Lockwood A. Principles of pharmacology in the eye. Br J Pharmacol 2017; 174:4205-4223. [PMID: 28865239 PMCID: PMC5715579 DOI: 10.1111/bph.14024] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/14/2017] [Accepted: 08/17/2017] [Indexed: 12/18/2022] Open
Abstract
The eye is a highly specialized organ that is subject to a huge range of pathology. Both local and systemic disease may affect different anatomical regions of the eye. The least invasive routes for ocular drug administration are topical (e.g. eye drops) and systemic (e.g. tablets) formulations. Barriers that subserve as protection against pathogen entry also restrict drug permeation. Topically administered drugs often display limited bioavailability due to many physical and biochemical barriers including the pre-corneal tear film, the structure and biophysiological properties of the cornea, the limited volume that can be accommodated by the cul-de-sac, the lacrimal drainage system and reflex tearing. The tissue layers of the cornea and conjunctiva are further key factors that act to restrict drug delivery. Using carriers that enhance viscosity or bind to the ocular surface increases bioavailability. Matching the pH and polarity of drug molecules to the tissue layers allows greater penetration. Drug delivery to the posterior segment is a greater challenge and, currently, the standard route is via intravitreal injection, notwithstanding the risks of endophthalmitis and retinal detachment with frequent injections. Intraocular implants that allow sustained drug release are at different stages of development. Novel exciting therapeutic approaches include methods for promoting transscleral delivery, sustained release devices, nanotechnology and gene therapy.
Collapse
Affiliation(s)
- Sahar Awwad
- UCL School of PharmacyLondonUK
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of OphthalmologyLondonUK
| | - Abeer H A Mohamed Ahmed
- UCL School of PharmacyLondonUK
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of OphthalmologyLondonUK
| | - Garima Sharma
- UCL School of PharmacyLondonUK
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of OphthalmologyLondonUK
| | - Jacob S Heng
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of OphthalmologyLondonUK
| | - Peng T Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of OphthalmologyLondonUK
| | - Steve Brocchini
- UCL School of PharmacyLondonUK
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of OphthalmologyLondonUK
| | | |
Collapse
|
41
|
Letelier J, Bovolenta P, Martínez-Morales JR. The pigmented epithelium, a bright partner against photoreceptor degeneration. J Neurogenet 2017; 31:203-215. [PMID: 29113536 DOI: 10.1080/01677063.2017.1395876] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sight depends on the intimate association between photoreceptors and pigment epithelial cells. The evolutionary origin of this cellular tandem can be traced back to the emergence of bilateral animals, at least 450 million years ago, as they define the minimal unit of the ancestral prototypic eye. Phototransduction is a demanding process from the energetic and homeostatic points of view, and not surprisingly photoreceptive cells are particularly susceptible to damage and degeneration. Here, we will examine the different ancillary roles that the pigmented cells play in the physiology and homeostasis of photoreceptors, linking each one of these processes to the most common hereditary retinal diseases. We will discuss the challenges and opportunities of recent therapeutic advances based on cell and gene replacement. The transition from animal models to clinical trials will be addressed for each one of the different therapeutic strategies with a special focus on those depending on retinal-pigmented epithelial cells. Finally, we will discuss the potential impact of combining CRISPR technologies with gene and cell therapy approaches, which - in the frame of the personalized medicine revolution - may constitute a leap forward in the treatment of retinal dystrophies.
Collapse
Affiliation(s)
- Joaquín Letelier
- a Centro Andaluz de Biología del Desarrollo (CSIC/UPO/JA) , Seville , Spain
| | - Paola Bovolenta
- b Centro de Biología Molecular "Severo Ochoa," (CSIC/UAM) and CIBERER, ISCIII , Madrid , Spain
| | | |
Collapse
|
42
|
Haenraets K, Foster E, Johannssen H, Kandra V, Frezel N, Steffen T, Jaramillo V, Paterna JC, Zeilhofer HU, Wildner H. Spinal nociceptive circuit analysis with recombinant adeno-associated viruses: the impact of serotypes and promoters. J Neurochem 2017; 142:721-733. [PMID: 28700081 DOI: 10.1111/jnc.14124] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 01/21/2023]
Abstract
Recombinant adeno-associated virus (rAAV) vector-mediated gene transfer into genetically defined neuron subtypes has become a powerful tool to study the neuroanatomy of neuronal circuits in the brain and to unravel their functions. More recently, this methodology has also become popular for the analysis of spinal cord circuits. To date, a variety of naturally occurring AAV serotypes and genetically modified capsid variants are available but transduction efficiency in spinal neurons, target selectivity, and the ability for retrograde tracing are only incompletely characterized. Here, we have compared the transduction efficiency of seven commonly used AAV serotypes after intraspinal injection. We specifically analyzed local transduction of different types of dorsal horn neurons, and retrograde transduction of dorsal root ganglia (DRG) neurons and of neurons in the rostral ventromedial medulla (RVM) and the somatosensory cortex (S1). Our results show that most of the tested rAAV vectors have similar transduction efficiency in spinal neurons. All serotypes analyzed were also able to transduce DRG neurons and descending RVM and S1 neurons via their spinal axon terminals. When comparing the commonly used rAAV serotypes to the recently developed serotype 2 capsid variant rAAV2retro, a > 20-fold increase in transduction efficiency of descending supraspinal neurons was observed. Conversely, transgene expression in retrogradely transduced neurons was strongly reduced when the human synapsin 1 (hSyn1) promoter was used instead of the strong ubiquitous hybrid cytomegalovirus enhancer/chicken β-actin promoter (CAG) or cytomegalovirus (CMV) promoter fragments. We conclude that the use of AAV2retro greatly increases transduction of neurons connected to the spinal cord via their axon terminals, while the hSyn1 promoter can be used to minimize transgene expression in retrogradely connected neurons of the DRG or brainstem. Cover Image for this issue: doi. 10.1111/jnc.13813.
Collapse
Affiliation(s)
- Karen Haenraets
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Institute of Pharmaceutical Sciences, Swiss Federal Institute (ETH) Zurich, Zurich, Switzerland
| | - Edmund Foster
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Helge Johannssen
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Vinnie Kandra
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Noémie Frezel
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Timothy Steffen
- Institute of Pharmaceutical Sciences, Swiss Federal Institute (ETH) Zurich, Zurich, Switzerland
| | - Valeria Jaramillo
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Jean-Charles Paterna
- Viral Vector Facility, University of Zurich and Swiss Federal Institute (ETH) Zurich, Zurich, Switzerland
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Institute of Pharmaceutical Sciences, Swiss Federal Institute (ETH) Zurich, Zurich, Switzerland
| | - Hendrik Wildner
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
43
|
Thakur SS, Ward MS, Popat A, Flemming NB, Parat MO, Barnett NL, Parekh HS. Stably engineered nanobubbles and ultrasound - An effective platform for enhanced macromolecular delivery to representative cells of the retina. PLoS One 2017; 12:e0178305. [PMID: 28542473 PMCID: PMC5444814 DOI: 10.1371/journal.pone.0178305] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/10/2017] [Indexed: 01/10/2023] Open
Abstract
Herein we showcase the potential of ultrasound-responsive nanobubbles in enhancing macromolecular permeation through layers of the retina, ultimately leading to significant and direct intracellular delivery; this being effectively demonstrated across three relevant and distinct retinal cell lines. Stably engineered nanobubbles of a highly homogenous and echogenic nature were fully characterised using dynamic light scattering, B-scan ultrasound and transmission electron microscopy (TEM). The nanobubbles appeared as spherical liposome-like structures under TEM, accompanied by an opaque luminal core and darkened corona around their periphery, with both features indicative of efficient gas entrapment and adsorption, respectively. A nanobubble +/- ultrasound sweeping study was conducted next, which determined the maximum tolerated dose for each cell line. Detection of underlying cellular stress was verified using the biomarker heat shock protein 70, measured before and after treatment with optimised ultrasound. Next, with safety to nanobubbles and optimised ultrasound demonstrated, each human or mouse-derived cell population was incubated with biotinylated rabbit-IgG in the presence and absence of ultrasound +/- nanobubbles. Intracellular delivery of antibody in each cell type was then quantified using Cy3-streptavidin. Nanobubbles and optimised ultrasound were found to be negligibly toxic across all cell lines tested. Macromolecular internalisation was achieved to significant, yet varying degrees in all three cell lines. The results of this study pave the way towards better understanding mechanisms underlying cellular responsiveness to ultrasound-triggered drug delivery in future ex vivo and in vivo models of the posterior eye.
Collapse
Affiliation(s)
- Sachin S. Thakur
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Micheal S. Ward
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
- School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Nicole B. Flemming
- Mater Research Institute, Translational Research Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Marie-Odile Parat
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Nigel L. Barnett
- Queensland Eye Institute, South Brisbane, Queensland, Australia
- UQ Centre for Clinical Research, The University of Queensland, Herston, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane Queensland, Australia
| | - Harendra S. Parekh
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
- * E-mail:
| |
Collapse
|
44
|
Broadgate S, Yu J, Downes SM, Halford S. Unravelling the genetics of inherited retinal dystrophies: Past, present and future. Prog Retin Eye Res 2017; 59:53-96. [PMID: 28363849 DOI: 10.1016/j.preteyeres.2017.03.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 02/07/2023]
Abstract
The identification of the genes underlying monogenic diseases has been of interest to clinicians and scientists for many years. Using inherited retinal dystrophies as an example of monogenic disease we describe the history of molecular genetic techniques that have been pivotal in the discovery of disease causing genes. The methods that were developed in the 1970's and 80's are still in use today but have been refined and improved. These techniques enabled the concept of the Human Genome Project to be envisaged and ultimately realised. When the successful conclusion of the project was announced in 2003 many new tools and, as importantly, many collaborations had been developed that facilitated a rapid identification of disease genes. In the post-human genome project era advances in computing power and the clever use of the properties of DNA replication has allowed the development of next-generation sequencing technologies. These methods have revolutionised the identification of disease genes because for the first time there is no need to define the position of the gene in the genome. The use of next generation sequencing in a diagnostic setting has allowed many more patients with an inherited retinal dystrophy to obtain a molecular diagnosis for their disease. The identification of novel genes that have a role in the development or maintenance of retinal function is opening up avenues of research which will lead to the development of new pharmacological and gene therapy approaches. Neither of which can be used unless the defective gene and protein is known. The continued development of sequencing technologies also holds great promise for the advent of truly personalised medicine.
Collapse
Affiliation(s)
- Suzanne Broadgate
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Levels 5 and 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| | - Jing Yu
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Levels 5 and 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| | - Susan M Downes
- Oxford Eye Hospital, Oxford University Hospitals NHS Trust, Oxford, OX3 9DU, UK
| | - Stephanie Halford
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Levels 5 and 6 West Wing, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK.
| |
Collapse
|
45
|
Suñé-Pou M, Prieto-Sánchez S, Boyero-Corral S, Moreno-Castro C, El Yousfi Y, Suñé-Negre JM, Hernández-Munain C, Suñé C. Targeting Splicing in the Treatment of Human Disease. Genes (Basel) 2017; 8:genes8030087. [PMID: 28245575 PMCID: PMC5368691 DOI: 10.3390/genes8030087] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/14/2017] [Accepted: 02/17/2017] [Indexed: 02/07/2023] Open
Abstract
The tightly regulated process of precursor messenger RNA (pre-mRNA) alternative splicing (AS) is a key mechanism in the regulation of gene expression. Defects in this regulatory process affect cellular functions and are the cause of many human diseases. Recent advances in our understanding of splicing regulation have led to the development of new tools for manipulating splicing for therapeutic purposes. Several tools, including antisense oligonucleotides and trans-splicing, have been developed to target and alter splicing to correct misregulated gene expression or to modulate transcript isoform levels. At present, deregulated AS is recognized as an important area for therapeutic intervention. Here, we summarize the major hallmarks of the splicing process, the clinical implications that arise from alterations in this process, and the current tools that can be used to deliver, target, and correct deficiencies of this key pre-mRNA processing event.
Collapse
Affiliation(s)
- Marc Suñé-Pou
- Department of Molecular Biology, Institute of Parasitology and Biomedicine "López Neyra" (IPBLN-CSIC), PTS, Granada 18016, Spain.
- Drug Development Service, Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Barcelona, Avda. Joan XXIII, s/n 08028 Barcelona, Spain.
| | - Silvia Prieto-Sánchez
- Department of Molecular Biology, Institute of Parasitology and Biomedicine "López Neyra" (IPBLN-CSIC), PTS, Granada 18016, Spain.
| | - Sofía Boyero-Corral
- Department of Molecular Biology, Institute of Parasitology and Biomedicine "López Neyra" (IPBLN-CSIC), PTS, Granada 18016, Spain.
| | - Cristina Moreno-Castro
- Department of Molecular Biology, Institute of Parasitology and Biomedicine "López Neyra" (IPBLN-CSIC), PTS, Granada 18016, Spain.
| | - Younes El Yousfi
- Department of Molecular Biology, Institute of Parasitology and Biomedicine "López Neyra" (IPBLN-CSIC), PTS, Granada 18016, Spain.
| | - Josep Mª Suñé-Negre
- Drug Development Service, Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Barcelona, Avda. Joan XXIII, s/n 08028 Barcelona, Spain.
| | - Cristina Hernández-Munain
- Department of Cell Biology and Immunology, Institute of Parasitology and Biomedicine "López Neyra" (IPBLN-CSIC), PTS, Granada 18016, Spain.
| | - Carlos Suñé
- Department of Molecular Biology, Institute of Parasitology and Biomedicine "López Neyra" (IPBLN-CSIC), PTS, Granada 18016, Spain.
| |
Collapse
|
46
|
Del Amo EM, Rimpelä AK, Heikkinen E, Kari OK, Ramsay E, Lajunen T, Schmitt M, Pelkonen L, Bhattacharya M, Richardson D, Subrizi A, Turunen T, Reinisalo M, Itkonen J, Toropainen E, Casteleijn M, Kidron H, Antopolsky M, Vellonen KS, Ruponen M, Urtti A. Pharmacokinetic aspects of retinal drug delivery. Prog Retin Eye Res 2016; 57:134-185. [PMID: 28028001 DOI: 10.1016/j.preteyeres.2016.12.001] [Citation(s) in RCA: 419] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/25/2016] [Accepted: 12/01/2016] [Indexed: 12/14/2022]
Abstract
Drug delivery to the posterior eye segment is an important challenge in ophthalmology, because many diseases affect the retina and choroid leading to impaired vision or blindness. Currently, intravitreal injections are the method of choice to administer drugs to the retina, but this approach is applicable only in selected cases (e.g. anti-VEGF antibodies and soluble receptors). There are two basic approaches that can be adopted to improve retinal drug delivery: prolonged and/or retina targeted delivery of intravitreal drugs and use of other routes of drug administration, such as periocular, suprachoroidal, sub-retinal, systemic, or topical. Properties of the administration route, drug and delivery system determine the efficacy and safety of these approaches. Pharmacokinetic and pharmacodynamic factors determine the required dosing rates and doses that are needed for drug action. In addition, tolerability factors limit the use of many materials in ocular drug delivery. This review article provides a critical discussion of retinal drug delivery, particularly from the pharmacokinetic point of view. This article does not include an extensive review of drug delivery technologies, because they have already been reviewed several times recently. Instead, we aim to provide a systematic and quantitative view on the pharmacokinetic factors in drug delivery to the posterior eye segment. This review is based on the literature and unpublished data from the authors' laboratory.
Collapse
Affiliation(s)
- Eva M Del Amo
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Anna-Kaisa Rimpelä
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Emma Heikkinen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Otto K Kari
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Eva Ramsay
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Tatu Lajunen
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Mechthild Schmitt
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Laura Pelkonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Madhushree Bhattacharya
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Dominique Richardson
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Astrid Subrizi
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Tiina Turunen
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Mika Reinisalo
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Jaakko Itkonen
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Elisa Toropainen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Marco Casteleijn
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | - Maxim Antopolsky
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland
| | | | - Marika Ruponen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Arto Urtti
- Centre for Drug Research, Division of Pharmaceutical Biosciences, University of Helsinki, Helsinki, Finland; School of Pharmacy, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
47
|
Choudhury S, Strang CE, Alexander JJ, Scalabrino ML, Lynch Hill J, Kasuga DT, Witherspoon CD, Boye SL, Gamlin PD, Boye SE. Novel Methodology for Creating Macaque Retinas with Sortable Photoreceptors and Ganglion Cells. Front Neurosci 2016; 10:551. [PMID: 27990105 PMCID: PMC5131003 DOI: 10.3389/fnins.2016.00551] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/16/2016] [Indexed: 01/08/2023] Open
Abstract
Purpose: The ability to generate macaque retinas with sortable cell populations would be of great benefit to both basic and translational studies of the primate retina. The purpose of our study was therefore to develop methods to achieve this goal by selectively labeling, in life, photoreceptors (PRs) and retinal ganglion cells (RGCs) with separate fluorescent markers. Methods: Labeling of macaque (Macaca fascicularis) PRs and RGCs was accomplished by subretinal delivery of AAV5-hGRK1-GFP, and retrograde transport of micro-ruby™ from the lateral geniculate nucleus, respectively. Retinas were anatomically separated into different regions. Dissociation conditions were optimized, and cells from each region underwent fluorescent activated cell sorting (FACS). Expression of retinal cell type- specific genes was assessed by quantitative real-time PCR to characterize isolated cell populations. Results: We show that macaque PRs and RGCs can be simultaneously labeled in-life and enriched populations isolated by FACS. Recovery from different retinal regions indicated efficient isolation/enrichment for PRs and RGCs, with the macula being particularly amendable to this technique. Conclusions: The methods and materials presented here allow for the identification of novel reagents designed to target RGCs and/or photoreceptors in a species that is phylogenetically and anatomically similar to human. These techniques will enable screening of intravitreally-delivered AAV capsid libraries for variants with increased tropism for PRs and/or RGCs and the evaluation of vector tropism and/or cellular promoter activity of gene therapy vectors in a clinically relevant species.
Collapse
Affiliation(s)
| | - Christianne E Strang
- Department of Psychology, University of Alabama at Birmingham Birmingham, AL, USA
| | | | | | - Julie Lynch Hill
- Department of Ophthalmology, University of Alabama at Birmingham Birmingham, AL, USA
| | - Daniel T Kasuga
- Department of Ophthalmology, University of Alabama at Birmingham Birmingham, AL, USA
| | - C Douglas Witherspoon
- Department of Ophthalmology, University of Alabama at Birmingham Birmingham, AL, USA
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida Gainesville, FL, USA
| | - Paul D Gamlin
- Department of Human Genetics, Emory University Atlanta, GA, USA
| | - Shannon E Boye
- Department of Ophthalmology, University of Florida Gainesville, FL, USA
| |
Collapse
|
48
|
Abstract
Glaucoma is a chronic optic neuropathy characterized by progressive damage to the optic nerve, death of retinal ganglion cells and ultimately visual field loss. It is one of the leading causes of irreversible loss of vision worldwide. The most important trigger of glaucomatous damage is elevated eye pressure, and the current standard approach in glaucoma therapy is reduction of intraocular pressure (IOP). However, despite the use of effective medications or surgical treatment leading to lowering of IOP, progression of glaucomatous changes and loss of vision among patients with glaucoma is common. Therefore, it is critical to prevent vision loss through additional treatment. To implement such treatment(s), it is imperative to identify pathophysiological changes in glaucoma and develop therapeutic methods taking into account neuroprotection. Currently, there is no method of neuroprotection with long-term proven effectiveness in the treatment of glaucoma. Among the most promising molecules shown to protect the retina and optic nerve are neurotrophic factors. Thus, the current focus is on the development of safe and non-invasive methods for the long-term elevation of the intraocular level of neurotrophins through advanced gene therapy and topical eye treatment and on the search for selective agonists of neurotrophin receptors affording more efficient neuroprotection.
Collapse
Affiliation(s)
- Anna Wójcik-Gryciuk
- Department of Ophthalmology, MSW Hospital, Warsaw, Poland
- Department of Neurophysiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Małgorzata Skup
- Department of Neurophysiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | | |
Collapse
|
49
|
Mitra RN, Zheng M, Han Z. Nanoparticle-motivated gene delivery for ophthalmic application. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 8:160-74. [PMID: 26109528 PMCID: PMC4688250 DOI: 10.1002/wnan.1356] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/21/2015] [Accepted: 05/23/2015] [Indexed: 12/24/2022]
Abstract
Ophthalmic gene therapy is an intellectual and intentional manipulation of desired gene expression into the specific cells of an eye for the treatment of ophthalmic (ocular) genetic dystrophies and pathological conditions. Exogenous nucleic acids such as DNA, small interfering RNA, micro RNA, and so on, are used for the purpose of managing expression of the desired therapeutic proteins in ocular tissues. The delivery of unprotected nucleic acids into the cells is limited because of exogenous and endogenous degradation modalities. Nanotechnology, a promising and sophisticated cutting edge tool, works as a protective shelter for these therapeutic nucleic acids. They can be safely delivered to the required cells in order to modulate anticipated protein expression. To this end, nanotechnology is seen as a potential and promising strategy in the field of ocular gene delivery. This review focused on current nanotechnology modalities and other promising nonviral strategies being used to deliver therapeutic genes in order to treat various devastating ocular diseases.
Collapse
Affiliation(s)
| | - Min Zheng
- Department of Ophthalmology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Zongchao Han
- Department of Ophthalmology, University of North Carolina, Chapel Hill, NC 27599, USA
- Carolina Institute for NanoMedicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
50
|
Giblin JP, Comes N, Strauss O, Gasull X. Ion Channels in the Eye: Involvement in Ocular Pathologies. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 104:157-231. [PMID: 27038375 DOI: 10.1016/bs.apcsb.2015.11.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The eye is the sensory organ of vision. There, the retina transforms photons into electrical signals that are sent to higher brain areas to produce visual sensations. In the light path to the retina, different types of cells and tissues are involved in maintaining the transparency of avascular structures like the cornea or lens, while others, like the retinal pigment epithelium, have a critical role in the maintenance of photoreceptor function by regenerating the visual pigment. Here, we have reviewed the roles of different ion channels expressed in ocular tissues (cornea, conjunctiva and neurons innervating the ocular surface, lens, retina, retinal pigment epithelium, and the inflow and outflow systems of the aqueous humor) that are involved in ocular disease pathophysiologies and those whose deletion or pharmacological modulation leads to specific diseases of the eye. These include pathologies such as retinitis pigmentosa, macular degeneration, achromatopsia, glaucoma, cataracts, dry eye, or keratoconjunctivitis among others. Several disease-associated ion channels are potential targets for pharmacological intervention or other therapeutic approaches, thus highlighting the importance of these channels in ocular physiology and pathophysiology.
Collapse
Affiliation(s)
- Jonathan P Giblin
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Nuria Comes
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Xavier Gasull
- Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|