1
|
Benagiano V, Rizzi A, Sannace C, Alessio G, Ribatti D, Dammacco R. Aqueous humor as eye lymph: A crossroad between venous and lymphatic system. Exp Eye Res 2024; 243:109904. [PMID: 38642600 DOI: 10.1016/j.exer.2024.109904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/18/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Aqueous humor (AQH) is a transparent fluid with characteristics similar to those of the interstitial fluid, which fills the eyeball posterior and anterior chambers and circulates in them from the sites of production to those of drainage. The AQH volume and pressure homeostasis is essential for the trophism of the ocular avascular tissues and their normal structure and function. Different AQH outflow pathways exist, including a main pathway, quite well defined anatomically and referred to as the conventional pathway, and some accessory pathways, more recently described and still not fully morphofunctionally understood, generically referred to as unconventional pathways. The conventional pathway is based on the existence of a series of conduits starting with the trabecular meshwork and Schlemm's Canal and continuing with a system of intrascleral and episcleral venules, which are tributaries to veins of the anterior segment of the eyeball. The unconventional pathways are mainly represented by the uveoscleral pathway, in which AQH flows through clefts, interstitial conduits located in the ciliary body and sclera, and then merges into the aforementioned intrascleral and episcleral venules. A further unconventional pathway, the lymphatic pathway, has been supported by the demonstration of lymphatic microvessels in the limbal sclera and, possibly, in the uvea (ciliary body, choroid) as well as by the ocular glymphatic channels, present in the neural retina and optic nerve. It follows that AQH may be drained from the eyeball through blood vessels (TM-SC pathway, US pathway) or lymphatic vessels (lymphatic pathway), and the different pathways may integrate or compensate for each other, optimizing the AQH drainage. The present review aims to define the state-of-the-art concerning the structural organization and the functional anatomy of all the AQH outflow pathways. Particular attention is paid to examining the regulatory mechanisms active in each of them. The new data on the anatomy and physiology of AQH outflow pathways is the key to understanding the pathophysiology of AQH outflow disorders and could open the way for novel approaches to their treatment.
Collapse
Affiliation(s)
- Vincenzo Benagiano
- Department of Translational Biomedicine and Neuroscience, University of Bari 'Aldo Moro', Bari, Italy.
| | - Anna Rizzi
- Department of Translational Biomedicine and Neuroscience, University of Bari 'Aldo Moro', Bari, Italy
| | - Carmela Sannace
- Azienda Sanitaria Locale Bari, Ophthalmology Day Service Triggiano-Gioia del Colle, Bari, Italy
| | - Giovanni Alessio
- Department of Translational Biomedicine and Neuroscience, University of Bari 'Aldo Moro', Bari, Italy
| | - Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, University of Bari 'Aldo Moro', Bari, Italy
| | - Rosanna Dammacco
- Department of Translational Biomedicine and Neuroscience, University of Bari 'Aldo Moro', Bari, Italy
| |
Collapse
|
2
|
Naaman E, Qarawani A, Ben-Zvi Elimelech R, Harel M, Sigal-Dror S, Safuri S, Smirnovas V, Baronaite I, Romanova NV, Morozova-Roche LA, Zayit-Soudry S. The Surprising Nonlinear Effects of S100A9 Proteins in the Retina. ACS Chem Neurosci 2024; 15:735-744. [PMID: 38324770 DOI: 10.1021/acschemneuro.3c00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024] Open
Abstract
Age-related macular degeneration (AMD) is a complex disease in which inflammation is implicated as a key factor but the precise molecular mechanisms are poorly understood. AMD lesions contain an excess of the pro-inflammatory S100A9 protein, but its retinal significance was yet unexplored. S100A9 was shown to be intrinsically amyloidogenic in vitro and in vivo. Here, we hypothesized that the retinal effects of S100A9 are related to its supramolecular conformation. ARPE-19 cultures were treated with native dimeric and fibrillar S100A9 preparations, and cell viability was determined. Wild-type rats were treated intravitreally with the S100A9 solutions in the right eye and with the vehicle in the left. Retinal function was assessed longitudinally by electroretinography (ERG), comparing the amplitudes and configurations for each intervention. Native S100A9 had no impact on cellular viability in vitro or on the retinal function in vivo. Despite dispersed intracellular uptake, fibrillar S100A9 did not decrease ARPE-19 cell viability. In contrast, S100A9 fibrils impaired retinal function in vivo following intravitreal injection in rats. Intriguingly, low-dose fibrillar S100A9 induced contrasting in vivo effects, significantly increasing the ERG responses, particularly over 14 days postinjection. The retinal effects of S100A9 were further characterized by glial and microglial cell activation. We provide the first indication for the retinal effects of S100A9, showing that its fibrils inflicted retinal dysfunction and glial activation in vivo, while low dose of the same assemblies resulted in an unpredicted enhancement of the ERG amplitudes. These nonlinear responses highlight the consequences of self-assembly of S100A9 and provide insight into its pathophysiological and possibly physiological roles in the retina.
Collapse
Affiliation(s)
- Efrat Naaman
- Department of Ophthalmology, Rambam Health Care Campus, Haifa 3109601, Israel
- Clinical Research Institute, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Amanda Qarawani
- Clinical Research Institute, Rambam Health Care Campus, Haifa 3109601, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa 69094, Israel
| | - Rony Ben-Zvi Elimelech
- Clinical Research Institute, Rambam Health Care Campus, Haifa 3109601, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa 69094, Israel
| | - Michal Harel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa 69094, Israel
| | - Shahaf Sigal-Dror
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa 69094, Israel
| | - Shadi Safuri
- Department of Ophthalmology, Rambam Health Care Campus, Haifa 3109601, Israel
- Clinical Research Institute, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Vytautas Smirnovas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
| | - Ieva Baronaite
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius LT-10257, Lithuania
| | - Nina V Romanova
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå SE-90781, Sweden
| | | | - Shiri Zayit-Soudry
- Department of Ophthalmology, Rambam Health Care Campus, Haifa 3109601, Israel
- Clinical Research Institute, Rambam Health Care Campus, Haifa 3109601, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa 69094, Israel
| |
Collapse
|
3
|
Araujo VG, Dias MS, Hauswirth WW, Linden R, Petrs-Silva H. rAAV-compatible human mini promoters enhance transgene expression in rat retinal ganglion cells. Exp Eye Res 2024; 239:109758. [PMID: 38123011 DOI: 10.1016/j.exer.2023.109758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/14/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Recombinant adeno-associated viral vectors (rAAV) are the safest and most effective gene delivery platform to drive the treatment of many inherited eye disorders in well-characterized animal models. The use in rAAV of ubiquitous promoters derived from viral sequences such as CMV/CBA (chicken β-actin promoter with cytomegalovirus enhancer) can lead to unwanted side effects such as pro-inflammatory immune responses and retinal cytotoxicity, thus reducing therapy efficacy. Thus, an advance in gene therapy is the availability of small promoters, that potentiate and direct gene expression to the cell type of interest, with higher safety and efficacy. In this study, we used six human mini-promoters packaged in rAAV2 quadruple mutant (Y-F) to test for transduction of the rat retina after intravitreal injection. After four weeks, immunohistochemical analysis detected GFP-labeled cells in the ganglion cell layer (GCL) for all constructs tested. Among them, Ple25sh1, Ple25sh2 and Ple53 promoted a widespread reporter-transgene expression in the GCL, with an increased number of GFP-expressing retinal ganglion cells when compared with the CMV/CBA vector. Moreover, Ple53 provided the strongest levels of GFP fluorescence in both cell soma and axons of retinal ganglion cells (RGCs) without any detectable adverse effects in retina function. Remarkably, a nearly 50-fold reduction in the number of intravitreally injected vector particles containing Ple53 promoter, still attained levels of transgene expression similar to CMV/CBA. Thus, the tested MiniPs show great potential for protocols of retinal gene therapy in therapeutic applications for retinal degenerations, especially those involving RGC-related disorders such as glaucoma.
Collapse
Affiliation(s)
- Victor G Araujo
- Laboratory of Gene Therapy and Viral Vector, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana S Dias
- Laboratory of Gene Therapy and Viral Vector, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - William W Hauswirth
- Retinal Gene Therapy Group, Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Rafael Linden
- Laboratory of Neurogenesis, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hilda Petrs-Silva
- Laboratory of Gene Therapy and Viral Vector, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
4
|
Nieuwenhuis B, Laperrousaz E, Tribble JR, Verhaagen J, Fawcett JW, Martin KR, Williams PA, Osborne A. Improving adeno-associated viral (AAV) vector-mediated transgene expression in retinal ganglion cells: comparison of five promoters. Gene Ther 2023:10.1038/s41434-022-00380-z. [PMID: 36635457 DOI: 10.1038/s41434-022-00380-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 12/02/2022] [Accepted: 12/21/2022] [Indexed: 01/14/2023]
Abstract
Recombinant adeno-associated viral vectors (AAVs) are an effective system for gene transfer. AAV serotype 2 (AAV2) is commonly used to deliver transgenes to retinal ganglion cells (RGCs) via intravitreal injection. The AAV serotype however is not the only factor contributing to the effectiveness of gene therapies. Promoters influence the strength and cell-selectivity of transgene expression. This study compares five promoters designed to maximise AAV2 cargo space for gene delivery: chicken β-actin (CBA), cytomegalovirus (CMV), short CMV early enhancer/chicken β-actin/short β-globulin intron (sCAG), mouse phosphoglycerate kinase (PGK), and human synapsin (SYN). The promoters driving enhanced green fluorescent protein (eGFP) were examined in adult C57BL/6J mice eyes and tissues of the visual system. eGFP expression was strongest in the retina, optic nerves and brain when driven by the sCAG and SYN promoters. CBA, CMV, and PGK had moderate expression by comparison. The SYN promoter had almost exclusive transgene expression in RGCs. The PGK promoter had predominant expression in both RGCs and AII amacrine cells. The ubiquitous CBA, CMV, and sCAG promoters expressed eGFP in a variety of cell types across multiple retinal layers including Müller glia and astrocytes. We also found that these promoters could transduce human retina ex vivo, although expression was predominantly in glial cells due to low RGC viability. Taken together, this promoter comparison study contributes to optimising AAV-mediated transduction in the retina, and could be valuable for research in ocular disorders, particularly those with large or complex genetic cargos.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK. .,Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK.
| | - Elise Laperrousaz
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, The Netherlands.,Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - James W Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Centre of Reconstructive Neuroscience, Institute of Experimental Medicine, Prague, Czech Republic
| | - Keith R Martin
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, VIC, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Andrew Osborne
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK. .,Ikarovec Ltd, The Norwich Research Park Innovation Centre, Norwich, UK.
| |
Collapse
|
5
|
Safi SZ, Saeed L, Shah H, Latif Z, Ali A, Imran M, Muhammad N, Emran TB, Subramaniyan V, Ismail ISB. Mechanisms of β-adrenergic receptors agonists in mediating pro and anti-apoptotic pathways in hyperglycemic Müller cells. Mol Biol Rep 2022; 49:9473-9480. [PMID: 35925485 DOI: 10.1007/s11033-022-07816-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/20/2022] [Indexed: 10/16/2022]
Abstract
BACKGROUND The current study aimed to investigate the stimulatory effect of beta-adrenergic receptors (β-ARs) on brain derived neurotropic factor (BDNF) and cAMP response element binding protein (CREB). METHODS Human Müller cells were cultured in low and high glucose conditions. Cells were treated with xamoterol (selective agonist for β1-AR), salmeterol (selective agonist for β2-AR), isoproterenol (β-ARs agonist) and propranolol (β-ARs antagonist), at 20 µM concentration for 24 h. Western Blotting assay was performed for the gene expression analysis. DNA damage was evaluated by TUNEL assay. DCFH-DA assay was used to check the level of reactive oxygen species (ROS). Cytochrome C release was measured by ELISA. RESULTS Xamoterol, salmeterol and isoproterenol showed no effect on Caspase-8 but it reduced the apoptosis and increased the expression of BDNF in Müller cells. A significant change in the expression of caspase-3 was observed in cells treated with xamoterol and salmeterol as compared to isoproterenol. Xamoterol, salmeterol and isoproterenol significantly decreased the reactive oxygen species (ROS) when treated for 24 hours. Glucose-induced cytochrome c release was disrupted in Müller cells. CONCLUSION β-ARs, stimulated by agonist play a protective role in hyperglycemic Müller cells, with the suppression of glucose-induced caspase-3 and cytochrome c release. B-Ars may directly mediate the gene expression of BDNF.
Collapse
Affiliation(s)
- Sher Zaman Safi
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, 42610, Jenjarom, Selangor, Malaysia.
- IRCBM, COMSATS University Islamabad, Lahore Campus, Lahore, Pakistan.
| | - Laiba Saeed
- Department of Zoology, Lahore College for Women University, Lahore, Pakistan
| | - Humaira Shah
- KRSS University of Management and Technology, Lahore, Pakistan
| | - Zahina Latif
- Regional Blood Center Peshawar, Peshawar, Pakistan
| | - Abid Ali
- Department of Zoology, Abdul Wali Khan University, Mardan, Mardan, Pakistan
| | - Muhammad Imran
- Department of Microbiology, University of Health Sciences, Lahore, Lahore, Pakistan
| | - Nawshad Muhammad
- Department of Microbiology, University of Health Sciences, Lahore, Lahore, Pakistan
- Department of Dental Materials, Institute of Basic Medical Sciences, Khyber Medical University Peshawar, Peshawar, Pakistan
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Vetriselvan Subramaniyan
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, 42610, Jenjarom, Selangor, Malaysia
| | - Ikram Shah Bin Ismail
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, 42610, Jenjarom, Selangor, Malaysia
| |
Collapse
|
6
|
Balaratnasingam C, An D, Hein M, Yu P, Yu DY. Studies of the retinal microcirculation using human donor eyes and high-resolution clinical imaging: Insights gained to guide future research in diabetic retinopathy. Prog Retin Eye Res 2022; 94:101134. [PMID: 37154065 DOI: 10.1016/j.preteyeres.2022.101134] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/18/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
The microcirculation plays a key role in delivering oxygen to and removing metabolic wastes from energy-intensive retinal neurons. Microvascular changes are a hallmark feature of diabetic retinopathy (DR), a major cause of irreversible vision loss globally. Early investigators have performed landmark studies characterising the pathologic manifestations of DR. Previous works have collectively informed us of the clinical stages of DR and the retinal manifestations associated with devastating vision loss. Since these reports, major advancements in histologic techniques coupled with three-dimensional image processing has facilitated a deeper understanding of the structural characteristics in the healthy and diseased retinal circulation. Furthermore, breakthroughs in high-resolution retinal imaging have facilitated clinical translation of histologic knowledge to detect and monitor progression of microcirculatory disturbances with greater precision. Isolated perfusion techniques have been applied to human donor eyes to further our understanding of the cytoarchitectural characteristics of the normal human retinal circulation as well as provide novel insights into the pathophysiology of DR. Histology has been used to validate emerging in vivo retinal imaging techniques such as optical coherence tomography angiography. This report provides an overview of our research on the human retinal microcirculation in the context of the current ophthalmic literature. We commence by proposing a standardised histologic lexicon for characterising the human retinal microcirculation and subsequently discuss the pathophysiologic mechanisms underlying key manifestations of DR, with a focus on microaneurysms and retinal ischaemia. The advantages and limitations of current retinal imaging modalities as determined using histologic validation are also presented. We conclude with an overview of the implications of our research and provide a perspective on future directions in DR research.
Collapse
Affiliation(s)
- Chandrakumar Balaratnasingam
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia; Department of Ophthalmology, Sir Charles Gairdner Hospital, Western Australia, Australia.
| | - Dong An
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Martin Hein
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Paula Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| | - Dao-Yi Yu
- Lions Eye Institute, Nedlands, Western Australia, Australia; Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, Australia
| |
Collapse
|
7
|
Le N, Appel H, Pannullo N, Hoang T, Blackshaw S. Ectopic insert-dependent neuronal expression of GFAP promoter-driven AAV constructs in adult mouse retina. Front Cell Dev Biol 2022; 10:914386. [PMID: 36200040 PMCID: PMC9527291 DOI: 10.3389/fcell.2022.914386] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Direct reprogramming of retinal Müller glia is a promising avenue for replacing photoreceptors and retinal ganglion cells lost to retinal dystrophies. However, questions have recently been raised about the accuracy of studies claiming efficient glia-to-neuron reprogramming in retina that were conducted using GFAP mini promoter-driven adeno-associated virus (AAV) vectors. In this study, we have addressed these questions using GFAP mini promoter-driven AAV constructs to simultaneously overexpress the mCherry reporter and candidate transcription factors predicted to induce glia-to-neuron conversion, in combination with prospective genetic labeling of retinal Müller glia using inducible Cre-dependent GFP reporters. We find that, while control GFAP-mCherry constructs express faithfully in Müller glia, 5 out of 7 transcription factor overexpression constructs tested are predominantly expressed in amacrine and retinal ganglion cells. These findings demonstrate strong insert-dependent effects on AAV-based GFAP mini promoter specificity that preclude its use in inferring cell lineage relationships when studying glia-to-neuron conversion in retina.
Collapse
Affiliation(s)
- Nguyet Le
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Haley Appel
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nicole Pannullo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Thanh Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
8
|
Liu G, Li H, Cull G, Wilsey L, Yang H, Reemmer J, Shen HY, Wang F, Fortune B, Bui BV, Wang L. Downregulation of Retinal Connexin 43 in GFAP-Expressing Cells Modifies Vasoreactivity Induced by Perfusion Ocular Pressure Changes. Invest Ophthalmol Vis Sci 2021; 62:26. [PMID: 33502459 PMCID: PMC7846954 DOI: 10.1167/iovs.62.1.26] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Purpose Glia and their communication via connexin 43 (Cx43) gap junctions are known to mediate neurovascular coupling, a process driven by metabolic demand. However, it is unclear whether Cx43 mediated glial communication intermediates classical autoregulation. Here we used viral transfection and a glial fibrillary acidic protein (GFAP) promoter to downregulate glial Cx43 to evaluate its role in retinal vascular autoregulation to ocular perfusion pressure (OPP) reduction. Methods Adult rats were intravitreally injected with the viral active construct or a control. Three weeks after the injection, eyes were imaged using confocal scanning laser ophthalmoscopy before and during a period of OPP decrease induced by blood draw to lower blood pressure or by manometric IOP elevation. Vessel diameter responses to the OPP decrease were compared between Cx43-downregulated and control-injected eyes. The extent of Cx43 downregulation was evaluated by Western blot and immunohistochemistry. Results In control eyes, the OPP decrease induced dilatation of arterioles, but not venules. In Cx43-downregulated eyes, Cx43 expression in whole retina was decreased by approximately 40%. In these eyes, the resting diameter of the venules increased significantly, but there was no effect on arterioles. In Cx43-downregulated eyes, vasoreactivity evoked by blood pressure lowering was significantly compromised in both arterioles (P = 0.005) and venules (P = 0.001). Cx43 downregulation did not affect the arteriole responses to IOP elevation, whereas the responses of the venules showed a significantly greater decrease in diameter (P < 0.001). Conclusions The downregulation of retinal Cx43 in GFAP–expressing cells compromises vasoreactivity of both arterioles and venules in response to an OPP decrease achieved via blood pressure lowering or IOP elevation. The results also suggest that Cx43-mediated glial communication actively regulates resting venular diameter.
Collapse
Affiliation(s)
- Guodong Liu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Shanghai, China.,Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Hui Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Shanghai, China.,Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Grant Cull
- Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Laura Wilsey
- Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Hongli Yang
- Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Jesica Reemmer
- RS Dow Neurobiology, Department of Translational Neuroscience, Legacy Research Institute, Portland, Oregon, United States
| | - Hai-Ying Shen
- RS Dow Neurobiology, Department of Translational Neuroscience, Legacy Research Institute, Portland, Oregon, United States
| | - Fang Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Shanghai, China
| | - Brad Fortune
- Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| | - Bang V Bui
- Department of Optometry and Vision Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Lin Wang
- Devers Eye Institute, Legacy Research Institute, Portland, Oregon, United States
| |
Collapse
|
9
|
Buck TM, Wijnholds J. Recombinant Adeno-Associated Viral Vectors (rAAV)-Vector Elements in Ocular Gene Therapy Clinical Trials and Transgene Expression and Bioactivity Assays. Int J Mol Sci 2020; 21:E4197. [PMID: 32545533 PMCID: PMC7352801 DOI: 10.3390/ijms21124197] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Inherited retinal dystrophies and optic neuropathies cause chronic disabling loss of visual function. The development of recombinant adeno-associated viral vectors (rAAV) gene therapies in all disease fields have been promising, but the translation to the clinic has been slow. The safety and efficacy profiles of rAAV are linked to the dose of applied vectors. DNA changes in the rAAV gene cassette affect potency, the expression pattern (cell-specificity), and the production yield. Here, we present a library of rAAV vectors and elements that provide a workflow to design novel vectors. We first performed a meta-analysis on recombinant rAAV elements in clinical trials (2007-2020) for ocular gene therapies. We analyzed 33 unique rAAV gene cassettes used in 57 ocular clinical trials. The rAAV gene therapy vectors used six unique capsid variants, 16 different promoters, and six unique polyadenylation sequences. Further, we compiled a list of promoters, enhancers, and other sequences used in current rAAV gene cassettes in preclinical studies. Then, we give an update on pro-viral plasmid backbones used to produce the gene therapy vectors, inverted terminal repeats, production yield, and rAAV safety considerations. Finally, we assess rAAV transgene and bioactivity assays applied to cells or organoids in vitro, explants ex vivo, and clinical studies.
Collapse
Affiliation(s)
- Thilo M. Buck
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
- Netherlands Institute of Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
10
|
Touahri Y, Dixit R, Kofoed RH, Mikloska K, Park E, Raeisossadati R, Markham-Coultes K, David LA, Rijal H, Zhao J, Lynch M, Hynynen K, Aubert I, Schuurmans C. Focused ultrasound as a novel strategy for noninvasive gene delivery to retinal Müller glia. Theranostics 2020; 10:2982-2999. [PMID: 32194850 PMCID: PMC7053200 DOI: 10.7150/thno.42611] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
Müller glia are specialized retinal cells with stem cell properties in fish and frogs but not in mammals. Current efforts to develop gene therapies to activate mammalian Müller glia for retinal repair will require safe and effective delivery strategies for recombinant adeno-associated viruses (AAVs), vectors of choice for clinical translation. Intravitreal and subretinal injections are currently used for AAV gene delivery in the eye, but less invasive methods efficiently targeting Müller glia have yet to be developed. Methods: As gene delivery strategies have been more extensively studied in the brain, to validate our vectors, we initially compared the glial tropism of AAV-PHP.eB, an AAV9 that crosses the blood-brain and blood-retinal barriers, for its ability to drive fluorescent protein expression in glial cells in both the brain and retina. We then tested the glial transduction of AAV2/8-GFAP-mCherry, a virus that does not cross blood-brain and blood-retinal barriers, for its effectiveness in transducing Müller glia in murine retinal explants ex vivo. For in vivo assays we used larger rat eyes, performing invasive intravitreal injections, and non-invasive intravenous delivery using focused ultrasound (FUS) (pressure amplitude: 0.360 - 0.84 MPa) and microbubbles (Definity, 0.2 ml/kg). Results: We showed that AAV-PHP.eB carrying a ubiquitous promoter (CAG) and green fluorescent protein (GFP) reporter, readily crossed the blood-brain and blood-retinal barriers after intravenous delivery in mice. However, murine Müller glia did not express GFP, suggesting that they were not transduced by AAV-PHP.eB. We thus tested an AAV2/8 variant, which was selected based on its safety record in multiple clinical trials, adding a glial fibrillary acidic protein (GFAP) promoter and mCherry (red fluorescent protein) reporter. We confirmed the glial specificity of AAV2/8-GFAP-mCherry, showing effective expression of mCherry in astrocytes after intracranial injection in the mouse brain, and of Müller glia in murine retinal explants. For in vivo experiments we switched to rats because of their larger size, injecting AAV2/8-GFAP-mCherry intravitreally, an invasive procedure, demonstrating passage across the inner limiting membrane, leading to Müller glia transduction. We then tested an alternative non-invasive delivery approach targeting a different barrier - the inner blood-retinal-barrier, applying focused ultrasound (FUS) to the retina after intravenous injection of AAV2/8 and microbubbles in rats, using magnetic resonance imaging (MRI) for FUS targeting. FUS permeabilized the rat blood-retinal-barrier and allowed the passage of macromolecules to the retina (Evans blue, IgG, IgM), with minimal extravasation of platelets and red blood cells. Intravenous injection of microbubbles and AAV2/8-GFAP-mCherry followed by FUS resulted in mCherry expression in rat Müller glia. However, systemic delivery of AAV2/8 also had off-target effects, transducing several murine peripheral organs, particularly the liver. Conclusions: Retinal permeabilisation via FUS in the presence of microbubbles is effective for delivering AAV2/8 across the inner blood-retinal-barrier, targeting Müller glia, which is less invasive than intravitreal injections that bypass the inner limiting membrane. However, implementing FUS in the clinic will require a comprehensive consideration of any off-target tropism of the AAV in peripheral organs, combined ideally, with the development of Müller glia-specific promoters.
Collapse
Affiliation(s)
- Yacine Touahri
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Rajiv Dixit
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Rikke Hahn Kofoed
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Kristina Mikloska
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - EunJee Park
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Reza Raeisossadati
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Kelly Markham-Coultes
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Luke Ajay David
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Hibo Rijal
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Jiayi Zhao
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Madelaine Lynch
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Isabelle Aubert
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Carol Schuurmans
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
11
|
Structure comparison of the chimeric AAV2.7m8 vector with parental AAV2. J Struct Biol 2019; 209:107433. [PMID: 31859208 DOI: 10.1016/j.jsb.2019.107433] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/28/2019] [Accepted: 12/03/2019] [Indexed: 12/26/2022]
Abstract
The AAV2.7m8 vector is an engineered capsid with a 10-amino acid insertion in adeno-associated virus (AAV) surface variable region VIII (VR-VIII) resulting in the alteration of an antigenic region of AAV2 and the ability to efficiently transduce retina cells following intravitreal administration. Directed evolution and in vivo screening in the mouse retina isolated this vector. In the present study, we sought to identify the structural differences between a recombinant AAV2.7m8 (rAAV2.7m8) vector packaging a GFP genome and its parental serotype, AAV2, by cryo-electron microscopy (cryo-EM) and image reconstruction. The structures of rAAV2.7m8 and AAV2 were determined to 2.91 and 3.02 Å resolution, respectively. The rAAV2.7m8 amino acid side-chains for residues 219-745 (the last C-terminal residue) were interpretable in the density map with the exception of the 10 inserted amino acids. While observable in a low sigma threshold density, side-chains were only resolved at the base of the insertion, likely due to flexibility at the top of the loop. A comparison to parental AAV2 (ordered from residues 217-735) showed the structures to be similar, except at some side-chains that had different orientations and, in VR-VIII containing the 10 amino acid insertion. VR-VIII is part of an AAV2 antigenic epitope, and the difference is consistent with rAAV2.7m8's escape from a known AAV2 monoclonal antibody, C37-B. The observations provide valuable insight into the configuration of inserted surface peptides on the AAV capsid and structural differences to be leveraged for future AAV vector rational design, especially for retargeted tropism and antibody escape.
Collapse
|
12
|
Cao J, Liu X, Yuan Y, Wang F, Kong W, Shi G, Li W, Zhang C. A rAAV2/6 Mutant with Enhanced Targeting for Mouse Retinal Müller Cells. Curr Eye Res 2019; 45:64-71. [PMID: 31294618 DOI: 10.1080/02713683.2019.1639768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose: Adeno-associated virus vector (AAV) is the most accepted gene delivery vector for retinal gene therapy. Müller cells play an important role in maintaining homeostasis and neuronal structural integrity, stability and it has been found to be involved in many retinopathies. The aim of this study is to identify a rAAV2/6 mutant which has increased tropism for Müller cell of the mouse retina.Materials and Methods: Using amino acid mutagenesis, we created a rAAV2/6 capsid mutant, rAAV2/6-S663L. In vivo imaging and retinal flat mount were employed to analyze the gene expression of rAAV2/6-S663L and wt rAAV2/6 in mouse retinal tissue. Retinal tissue cryosection, immunohistochemistry (IHC), Müller cell-specific promoter-controlled gene expression, and double AAV fluorescent protein co-expression were performed to determine the targeting of rAAV2/6-S663L for mouse retinal Müller cells.Results: In vivo imaging, retinal flat mount and retinal tissue cryosection results showed that rAAV2/6-S663L and wt rAAV2/6 have different specific tropisms in mouse retina and rAAV2/6-S663L is more preferentially targeting Müller cells. Müller cell-specific promoter-controlled gene expression experiments and IHC test confirmed that rAAV2/6-S663L has a higher tendency to infect Müller cells than wt rAAV2/6. Co-infection of the mouse retina with one rAAV2/6-S663L expressing EGFP under the control of GFAP promoter and the other one expressing mCherry under the control of CMV promoter revealed co-expression of the two fluorescent proteins in Müller cells.Conclusions: The results confirmed that rAAV2/6-S663L has a higher tropism for Müller cells than wt rAAV2/6. Our findings could add a new useful tool for retinal disease gene therapy.
Collapse
Affiliation(s)
- Jinjing Cao
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| | - Xiaomei Liu
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| | - Yun Yuan
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| | - Feifei Wang
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| | - Wen Kong
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| | - Guohua Shi
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China.,The Innovation Center of Excellence on Brain Science, Chinese Academy of Sciences, Shanghai, China
| | - Wensheng Li
- Aier School of Ophthalmology, Central South University, Changsha, China.,Ophthalmology, Shanghai Aier Eye Hospital, Shanghai, China
| | - Chun Zhang
- Suzhou Institute of Biomedical Engineering and Technology, University of Science and Technology of China, Suzhou, China
| |
Collapse
|
13
|
Jin J, Smith MD, Kersbergen CJ, Kam TI, Viswanathan M, Martin K, Dawson TM, Dawson VL, Zack DJ, Whartenby K, Calabresi PA. Glial pathology and retinal neurotoxicity in the anterior visual pathway in experimental autoimmune encephalomyelitis. Acta Neuropathol Commun 2019; 7:125. [PMID: 31366377 PMCID: PMC6670238 DOI: 10.1186/s40478-019-0767-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/06/2019] [Indexed: 12/23/2022] Open
Abstract
The animal model experimental autoimmune encephalomyelitis (EAE) has been used extensively in the past to test mechanisms that target peripheral immune cells for treatment of multiple sclerosis (MS). While there have been some notable successes in relapsing MS, the development of therapies for progressive multiple sclerosis (MS) has been hampered by lack of an appropriate animal model. Further, the mechanisms underlying CNS inflammation and neuronal injury remain incompletely elucidated. It is known that the MOG 35-55 EAE mouse model does not have insidious behavioral progression as occurs in people with MS, but there is significant neuronal and axonal injury in EAE, as a result of the inflammation. In the present study, we describe the time course of glial activation and retinal neurodegeneration in the EAE model, and highlight the utility of studying the anterior visual pathway for modeling mechanisms of neuronal injury that may recapitulate critical aspects of the pathology described in people with MS following optic neuritis and subclinical optic neuropathy. We show that A1 neurotoxic astrocytes are prevalent in optic nerve tissue and retina, and are associated with subsequent RGC loss in the most commonly used form of the EAE model induced by MOG 35-55 peptide in C57/B6 mice. We developed a semi-automatic method to quantify retinal ganglion cells (RGC) and show that RGCs remain intact at peak EAE (PID 16) but are significantly reduced in late EAE (PID 42). Postsynaptic proteins and neurites were also compromised in the retina of late EAE mice. The retinal pathology manifests weeks after the microglial and astrocyte activation, which were prominent in optic nerve tissues at PID 16. Microglia expressed iNOS and had increased gene expression of C1q, TNF-α, and IL-1α. Astrocytes expressed high levels of complement component 3 and other genes associated with A1 neurotoxic astrocytes. Our data suggest that EAE can be used to study the pathobiology of optic neuropathy and to examine the preclinical neuroprotective effects of drugs that target activation of neurotoxic A1 astrocytes.
Collapse
|
14
|
Quinn PM, Buck TM, Mulder AA, Ohonin C, Alves CH, Vos RM, Bialecka M, van Herwaarden T, van Dijk EHC, Talib M, Freund C, Mikkers HMM, Hoeben RC, Goumans MJ, Boon CJF, Koster AJ, Chuva de Sousa Lopes SM, Jost CR, Wijnholds J. Human iPSC-Derived Retinas Recapitulate the Fetal CRB1 CRB2 Complex Formation and Demonstrate that Photoreceptors and Müller Glia Are Targets of AAV5. Stem Cell Reports 2019; 12:906-919. [PMID: 30956116 PMCID: PMC6522954 DOI: 10.1016/j.stemcr.2019.03.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/13/2022] Open
Abstract
Human retinal organoids from induced pluripotent stem cells (hiPSCs) can be used to confirm the localization of proteins in retinal cell types and to test transduction and expression patterns of gene therapy vectors. Here, we compared the onset of CRB protein expression in human fetal retina with human iPSC-derived retinal organoids. We show that CRB2 protein precedes the expression of CRB1 in the developing human retina. Our data suggest the presence of CRB1 and CRB2 in human photoreceptors and Müller glial cells. Thus the fetal CRB complex formation is replicated in hiPSC-derived retina. CRB1 patient iPSC retinal organoids showed disruptions at the outer limiting membrane as found in Crb1 mutant mice. Furthermore, AAV serotype 5 (AAV5) is potent in infecting human Müller glial cells and photoreceptors in hiPSC-derived retinas and retinal explants. Our data suggest that human photoreceptors can be efficiently transduced by AAVs in the presence of photoreceptor segments.
Collapse
Affiliation(s)
- Peter M Quinn
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Thilo M Buck
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Aat A Mulder
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Charlotte Ohonin
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - C Henrique Alves
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Rogier M Vos
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands
| | - Monika Bialecka
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Tessa van Herwaarden
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Elon H C van Dijk
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Mays Talib
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Christian Freund
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Harald M M Mikkers
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Rob C Hoeben
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Marie-José Goumans
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Camiel J F Boon
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands; Department of Ophthalmology, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, 1000 AE Amsterdam, The Netherlands
| | - Abraham J Koster
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | | | - Carolina R Jost
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands; Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands.
| |
Collapse
|
15
|
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
|
16
|
Peynshaert K, Devoldere J, Minnaert AK, De Smedt SC, Remaut K. Morphology and Composition of the Inner Limiting Membrane: Species-Specific Variations and Relevance toward Drug Delivery Research. Curr Eye Res 2019; 44:465-475. [PMID: 30638413 DOI: 10.1080/02713683.2019.1565890] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The inner limiting membrane (ILM) represents the structural boundary between the vitreous and the retina, and is suggested to act as a barrier for a wide range of retinal therapies. While it is widely acknowledged that the morphology of the human ILM exhibits regional variations and undergoes age-related changes, insight into its structure in laboratory animals is very limited. Besides presenting a detailed overview of the morphology and composition of the human ILM, this review specifically reflects on the species-specific differences in ILM structure. With these differences in mind, we furthermore summarize the most relevant reports on the barrier role of the ILM with regard to viral vectors, nanoparticles, anti-VEGF medication and stem cells. Overall, this review aims to deliberate on the impact of species-specific ILM variations on drug delivery research as well as to pinpoint knowledge gaps which future basic research should resolve.
Collapse
Affiliation(s)
- Karen Peynshaert
- a Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium.,b Ghent Research Group on Nanomedicines , Ghent University , Ghent , Belgium
| | - Joke Devoldere
- a Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium.,b Ghent Research Group on Nanomedicines , Ghent University , Ghent , Belgium
| | - An-Katrien Minnaert
- a Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium.,b Ghent Research Group on Nanomedicines , Ghent University , Ghent , Belgium
| | - Stefaan C De Smedt
- a Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium.,b Ghent Research Group on Nanomedicines , Ghent University , Ghent , Belgium
| | - Katrien Remaut
- a Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences , Ghent University , Ghent , Belgium.,b Ghent Research Group on Nanomedicines , Ghent University , Ghent , Belgium
| |
Collapse
|
17
|
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: 148] [Impact Index Per Article: 21.1] [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
|
18
|
Schultz R, Krug M, Precht M, Wohl SG, Witte OW, Schmeer C. Frataxin overexpression in Müller cells protects retinal ganglion cells in a mouse model of ischemia/reperfusion injury in vivo. Sci Rep 2018; 8:4846. [PMID: 29555919 PMCID: PMC5859167 DOI: 10.1038/s41598-018-22887-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/02/2018] [Indexed: 01/28/2023] Open
Abstract
Müller cells are critical for retinal function and neuronal survival but can become detrimental in response to retinal ischemia and increased oxidative stress. Elevated oxidative stress increases expression of the mitochondrial enzyme frataxin in the retina, and its overexpression is neuroprotective after ischemia. Whether frataxin expression in Müller cells might improve their function and protect neurons after ischemia is unknown. The aim of this study was to evaluate the effect of frataxin overexpression in Müller cells on neuronal survival after retinal ischemia/reperfusion in the mouse in vivo. Retinal ischemia/reperfusion was induced in mice overexpressing frataxin in Müller cells by transient elevation of intraocular pressure. Retinal ganglion cells survival was determined 14 days after lesion. Expression of frataxin, antioxidant enzymes, growth factors and inflammation markers was determined with qRT-PCR, Western blotting and immunohistochemistry 24 hours after lesion. Following lesion, there was a 65% increase in the number of surviving RGCs in frataxin overexpressing mice. Improved survival was associated with increased expression of the antioxidant enzymes Gpx1 and Sod1 as well as the growth factors Cntf and Lif. Additionally, microglial activation was decreased in these mice. Therefore, support of Müller cell function constitutes a feasible approach to reduce neuronal degeneration after ischemia.
Collapse
Affiliation(s)
- Rowena Schultz
- Department of Ophthalmology, Jena University Hospital, Jena, Germany
| | - Melanie Krug
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Michel Precht
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Stefanie G Wohl
- Department of Biological Structure, University of Washington Seattle, Seattle, United States
| | - Otto W Witte
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Christian Schmeer
- Hans-Berger Department of Neurology, Jena University Hospital, Jena, Germany.
| |
Collapse
|
19
|
Retinal Angiogenesis Regulates Astrocytic Differentiation in Neonatal Mouse Retinas by Oxygen Dependent Mechanisms. Sci Rep 2017; 7:17608. [PMID: 29242645 PMCID: PMC5730567 DOI: 10.1038/s41598-017-17962-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 12/01/2017] [Indexed: 11/13/2022] Open
Abstract
In mice, retinal vascular and astrocyte networks begin to develop at birth, expanding radially from the optic nerve head (ONH) towards the retinal periphery. The retinal vasculature grows towards the periphery ahead of differentiated astrocytes, but behind astrocytic progenitor cells (APCs) and immature astrocytes. Endothelial cell specific Vegfr-2 disruption in newborn mice not only blocked retinal vascular development but also suppressed astrocytic differentiation, reducing the abundance of differentiated astrocytes while causing the accumulation of precursors. By contrast, retinal astrocytic differentiation was accelerated by the exposure of wild-type newborn mice to hyperoxia for 24 hours, or by APC specific deficiency in hypoxia inducible factor (HIF)−2α, an oxygen labile transcription factor. These findings reveal a novel function of the retinal vasculature, and imply that in normal neonatal mice, oxygen from the retinal circulation may promote astrocytic differentiation, in part by triggering oxygen dependent HIF-2α degradation in astrocytic precursors.
Collapse
|
20
|
Peyer SM, Heath-Heckman EAC, McFall-Ngai MJ. Characterization of the cell polarity gene crumbs during the early development and maintenance of the squid-vibrio light organ symbiosis. Dev Genes Evol 2017; 227:375-387. [PMID: 28105525 PMCID: PMC5519459 DOI: 10.1007/s00427-017-0576-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/13/2017] [Indexed: 11/26/2022]
Abstract
The protein Crumbs is a determinant of apical-basal cell polarity and plays a role in apoptosis of epithelial cells and their protection against photodamage. Using the squid-vibrio system, a model for development of symbiotic partnerships, we examined the modulation of the crumbs gene in host epithelial tissues during initiation and maintenance of the association. The extracellular luminous symbiont Vibrio fischeri colonizes the apical surfaces of polarized epithelia in deep crypts of the Euprymna scolopes light organ. During initial colonization each generation, symbiont harvesting is potentiated by the biochemical and biophysical activity of superficial ciliated epithelia, which are several cell layers from the crypt epithelia where the symbionts reside. Within hours of crypt colonization, the symbionts induce the cell death mediated regression of the remote superficial ciliated fields. However, the crypt cells directly interacting with the symbiont are protected from death. In the squid host, we characterized the gene and encoded protein during light organ morphogenesis and in response to symbiosis. Features of the protein sequence and structure, phylogenetic relationships, and localization patterns in the eye supported assignment of the squid protein to the Crumbs family. In situ hybridization revealed that the crumbs transcript shows opposite expression at the onset of symbiosis in the two different regions of the light organ: elevated levels in the superficial epithelia were attenuated whereas low levels in the crypt epithelia were turned up. Although a rhythmic association in which the host controls the symbiont population over the day-night cycle begins in the juvenile upon colonization, cycling of crumbs was evident only in the adult organ with peak expression coincident with maximum symbiont population and luminescence. Our results provide evidence that crumbs responds to symbiont cues that induce developmental apoptosis and to symbiont population dynamics correlating with luminescence-based stress throughout the duration of the host-microbe association.
Collapse
Affiliation(s)
- Suzanne M Peyer
- School of Medicine and Public Health, Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, 53706, USA.
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI, 53706, USA.
| | - Elizabeth A C Heath-Heckman
- School of Medicine and Public Health, Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, 53706, USA
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA, 94720, USA
| | - Margaret J McFall-Ngai
- School of Medicine and Public Health, Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, 53706, USA.
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI, 53706, USA.
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
21
|
Destination Brain: the Past, Present, and Future of Therapeutic Gene Delivery. J Neuroimmune Pharmacol 2017; 12:51-83. [PMID: 28160121 DOI: 10.1007/s11481-016-9724-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/12/2016] [Indexed: 12/20/2022]
Abstract
Neurological diseases and disorders (NDDs) present a significant societal burden and currently available drug- and biological-based therapeutic strategies have proven inadequate to alleviate it. Gene therapy is a suitable alternative to treat NDDs compared to conventional systems since it can be tailored to specifically alter select gene expression, reverse disease phenotype and restore normal function. The scope of gene therapy has broadened over the years with the advent of RNA interference and genome editing technologies. Consequently, encouraging results from central nervous system (CNS)-targeted gene delivery studies have led to their transition from preclinical to clinical trials. As we shift to an exciting gene therapy era, a retrospective of available literature on CNS-associated gene delivery is in order. This review is timely in this regard, since it analyzes key challenges and major findings from the last two decades and evaluates future prospects of brain gene delivery. We emphasize major areas consisting of physiological and pharmacological challenges in gene therapy, function-based selection of a ideal cellular target(s), available therapy modalities, and diversity of viral vectors and nanoparticles as vehicle systems. Further, we present plausible answers to key questions such as strategies to circumvent low blood-brain barrier permeability and most suitable CNS cell types for targeting. We compare and contrast pros and cons of the tested viral vectors in the context of delivery systems used in past and current clinical trials. Gene vector design challenges are also evaluated in the context of cell-specific promoters. Key challenges and findings reported for recent gene therapy clinical trials, assessing viral vectors and nanoparticles are discussed from the perspective of bench to bedside gene therapy translation. We conclude this review by tying together gene delivery challenges, available vehicle systems and comprehensive analyses of neuropathogenesis to outline future prospects of CNS-targeted gene therapies.
Collapse
|
22
|
Heparan Sulfate Binding Promotes Accumulation of Intravitreally Delivered Adeno-associated Viral Vectors at the Retina for Enhanced Transduction but Weakly Influences Tropism. J Virol 2016; 90:9878-9888. [PMID: 27558418 DOI: 10.1128/jvi.01568-16] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 08/15/2016] [Indexed: 12/20/2022] Open
Abstract
Many adeno-associated virus (AAV) serotypes efficiently transduce the retina when delivered to the subretinal space but show limited success when delivered to the vitreous due to the inner limiting membrane (ILM). Subretinal delivery of AAV serotype 2 (AAV2) and its heparan sulfate (HS)-binding-deficient capsid led to similar expression, indicating transduction of the outer retina occurred by HS-independent mechanisms. However, intravitreal delivery of HS-ablated recombinant AAV2 (rAAV2) led to a 300-fold decrease in transduction compared to AAV2. Fluorescence in situ hybridization of AAV transgenes was used to identify differences in retinal trafficking and revealed that HS binding was responsible for AAV2 accumulation at the ILM. This mechanism was tested on human ex vivo retinas and showed similar accumulation with HS-binding AAV2 capsid only. To evaluate if HS binding could be applied to other AAV serotypes to enhance their transduction, AAV1 and AAV8 were modified to bind HS with a single-amino-acid mutation and tested in mice. Both HS-binding mutants of AAV1 and AAV8 had higher intravitreal transduction than their non-HS-binding parent capsid due to increased retinal accumulation. To understand the influence that HS binding has on tropism, chimeric AAV2 capsids with dual-glycan usage were tested intravitreally in mice. Compared to HS binding alone, these chimeric capsids displayed enhanced transduction that was correlated with a change in tropism. Taken together, these data indicate that HS binding serves to sequester AAV capsids from the vitreous to the ILM but does not influence retinal tropism. The enhanced retinal transduction of HS-binding capsids provides a rational design strategy for engineering capsids for intravitreal delivery. IMPORTANCE Adeno-associated virus (AAV) has become the vector of choice for viral gene transfer and has shown great promise in clinical trials. The need for development of an easy, less invasive injection route for ocular gene therapy is met by intravitreal delivery, but delivery of AAV by this route results in poor transduction outcomes. The inner limiting membrane (ILM) creates a barrier separating the vitreous and the retina. Binding of AAV to heparan sulfate proteoglycan (HSPG) at the ILM may allow the virus to traverse this barrier for better retinal transduction. We show that HSPG binding is correlated with greater accumulation and penetration of AAV in the retina. We demonstrated that this accumulation is conserved across mouse and human retinas and that the addition of HSPG binding to other AAV capsids can increase the number of vectors accumulating at the ILM without dictating tropism.
Collapse
|
23
|
El-Shamayleh Y, Ni AM, Horwitz GD. Strategies for targeting primate neural circuits with viral vectors. J Neurophysiol 2016; 116:122-34. [PMID: 27052579 PMCID: PMC4961743 DOI: 10.1152/jn.00087.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/05/2016] [Indexed: 11/22/2022] Open
Abstract
Understanding how the brain works requires understanding how different types of neurons contribute to circuit function and organism behavior. Progress on this front has been accelerated by optogenetics and chemogenetics, which provide an unprecedented level of control over distinct neuronal types in small animals. In primates, however, targeting specific types of neurons with these tools remains challenging. In this review, we discuss existing and emerging strategies for directing genetic manipulations to targeted neurons in the adult primate central nervous system. We review the literature on viral vectors for gene delivery to neurons, focusing on adeno-associated viral vectors and lentiviral vectors, their tropism for different cell types, and prospects for new variants with improved efficacy and selectivity. We discuss two projection targeting approaches for probing neural circuits: anterograde projection targeting and retrograde transport of viral vectors. We conclude with an analysis of cell type-specific promoters and other nucleotide sequences that can be used in viral vectors to target neuronal types at the transcriptional level.
Collapse
Affiliation(s)
- Yasmine El-Shamayleh
- Department of Physiology and Biophysics and Washington National Primate Research Center, University of Washington, Seattle, Washington; and
| | - Amy M Ni
- Department of Neuroscience and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gregory D Horwitz
- Department of Physiology and Biophysics and Washington National Primate Research Center, University of Washington, Seattle, Washington; and
| |
Collapse
|
24
|
Boyd RF, Sledge DG, Boye SL, Boye SE, Hauswirth WW, Komáromy AM, Petersen-Jones SM, Bartoe JT. Photoreceptor-targeted gene delivery using intravitreally administered AAV vectors in dogs. Gene Ther 2016; 23:223-30. [PMID: 26467396 PMCID: PMC4840844 DOI: 10.1038/gt.2015.96] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/03/2015] [Accepted: 08/24/2015] [Indexed: 12/11/2022]
Abstract
Delivery of therapeutic transgenes to retinal photoreceptors using adeno-associated virus (AAV) vectors has traditionally required subretinal injection. Recently, photoreceptor transduction efficiency following intravitreal injection (IVT) has improved in rodent models through use of capsid-mutant AAV vectors; but remains limited in large animal models. Thickness of the inner limiting membrane (ILM) in large animals is thought to impair retinal penetration by AAV. Our study compared two newly developed AAV vectors containing multiple capsid amino acid substitutions following IVT in dogs. The ability of two promoter constructs to restrict reporter transgene expression to photoreceptors was also evaluated. AAV vectors containing the interphotoreceptor-binding protein (IRBP) promoter drove expression exclusively in rod and cone photoreceptors, with transduction efficiencies of ~4% of cones and 2% of rods. Notably, in the central region containing the cone-rich visual streak, 15.6% of cones were transduced. Significant regional variation existed, with lower transduction efficiencies in the temporal regions of all eyes. This variation did not correlate with ILM thickness. Vectors carrying a cone-specific promoter failed to transduce a quantifiable percentage of cone photoreceptors. The newly developed AAV vectors containing the IRBP promoter were capable of producing photoreceptor-specific transgene expression following IVT in the dog.
Collapse
Affiliation(s)
- RF Boyd
- Small Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - DG Sledge
- Diagnostic Center for Population and Animal Health, Michigan State University, East Lansing, MI, USA
| | - SL Boye
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, FL, USA
| | - SE Boye
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, FL, USA
| | - WW Hauswirth
- Department of Ophthalmology, University of Florida College of Medicine, Gainesville, FL, USA
| | - AM Komáromy
- Small Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - SM Petersen-Jones
- Small Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - JT Bartoe
- Small Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
25
|
Retinal gene delivery by adeno-associated virus (AAV) vectors: Strategies and applications. Eur J Pharm Biopharm 2015; 95:343-52. [DOI: 10.1016/j.ejpb.2015.01.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 01/10/2015] [Accepted: 01/12/2015] [Indexed: 11/20/2022]
|
26
|
Pellissier LP, Quinn PM, Alves CH, Vos RM, Klooster J, Flannery JG, Heimel JA, Wijnholds J. Gene therapy into photoreceptors and Müller glial cells restores retinal structure and function in CRB1 retinitis pigmentosa mouse models. Hum Mol Genet 2015; 24:3104-18. [PMID: 25701872 DOI: 10.1093/hmg/ddv062] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/13/2015] [Indexed: 11/14/2022] Open
Abstract
Mutations in the Crumbs-homologue-1 (CRB1) gene lead to severe recessive inherited retinal dystrophies. Gene transfer therapy is the most promising cure for retinal dystrophies and has primarily been applied for recessive null conditions via a viral gene expression vector transferring a cDNA encoding an enzyme or channel protein, and targeting expression to one cell type. Therapy for the human CRB1 disease will be more complex, as CRB1 is a structural and signaling transmembrane protein present in three cell classes: Müller glia, cone and rod photoreceptors. In this study, we applied CRB1 and CRB2 gene therapy vectors in Crb1-retinitis pigmentosa mouse models at mid-stage disease. We tested if CRB expression restricted to Müller glial cells or photoreceptors or co-expression in both is required to recover retinal function. We show that targeting both Müller glial cells and photoreceptors with CRB2 ameliorated retinal function and structure in Crb1 mouse models. Surprisingly, targeting a single cell type or all cell types with CRB1 reduced retinal function. We show here the first pre-clinical studies for CRB1-related eye disorders using CRB2 vectors and initial elucidation of the cellular mechanisms underlying CRB1 function.
Collapse
Affiliation(s)
| | | | | | | | | | - John G Flannery
- Department of Molecular and Cellular Biology, The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA and
| | - J Alexander Heimel
- Department of Cortical Structure & Function, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands
| | - Jan Wijnholds
- Department of Neuromedical Genetics Department of Ophthalmology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
27
|
Woappi YL, Jangiti R, Singh OV. Synthetic immunosurveillance systems: nanodevices to monitor physiological events. Biosens Bioelectron 2014; 61:152-64. [PMID: 24874659 PMCID: PMC7065416 DOI: 10.1016/j.bios.2014.04.056] [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: 01/16/2014] [Revised: 04/24/2014] [Accepted: 04/29/2014] [Indexed: 11/23/2022]
Abstract
The field of nanotechnology has recently seen vast advancements in its applications for therapeutic strategy. This technological revolution has led way to nanomedicine, which spurred the development of clever drug delivery designs and ingenious nanovehicles for the monitoring of cellular events in vivo. The clinical implementations of this technology are innumerable and have demonstrated utility as diagnostic tools and fortifying machineries for the mammalian immune system. Recently engineered viral vectors and multi-subunit packaging RNAs have verified stable enough for long-term existence in the physiological environment and therefore reveal unique potential as artificial immunosurveillance devices. Physiological and pathological events recorded by nanodevices could help develop "biocatalogs" of patients' infection history, frequency of disease, and much more. In this article, we introduce a novel design concept for a multilayer synthetic immune network parallel to the natural immune system; an artificial network of continuously patrolling nanodevices incorporated in the blood and lymphatic systems, and adapted for molecular event recording, anomaly detection, drug delivery, and gene silencing. We also aim to discuss the approaches and advances recently reported in nanomedicine, especially as it pertains to promising viral and RNA-based nanovehicles and their prospective applications for the development of a synthetic immunosurveillance system (SIS). Alternative suggestions and limitations of these technologies are also discussed.
Collapse
Affiliation(s)
- Yvon L Woappi
- Division of Biological and Health Sciences, 300 Campus Drive, University of Pittsburgh, Bradford, PA 16701, USA
| | - Rahul Jangiti
- Division of Physical and Computational Sciences, 300 Campus Drive, University of Pittsburgh, Bradford, PA 16701, USA
| | - Om V Singh
- Division of Biological and Health Sciences, 300 Campus Drive, University of Pittsburgh, Bradford, PA 16701, USA.
| |
Collapse
|
28
|
Pellissier LP, Hoek RM, Vos RM, Aartsen WM, Klimczak RR, Hoyng SA, Flannery JG, Wijnholds J. Specific tools for targeting and expression in Müller glial cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14009. [PMID: 26015954 PMCID: PMC4362388 DOI: 10.1038/mtm.2014.9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/12/2014] [Indexed: 12/17/2022]
Abstract
Despite their physiological roles, Müller glial cells are involved directly or indirectly in retinal disease pathogenesis and are an interesting target for therapeutic approaches for retinal diseases and regeneration such as CRB1 inherited retinal dystrophies. In this study, we characterized the efficiency of adeno-associated virus (AAV) capsid variants and different promoters to drive protein expression in Müller glial cells. ShH10Y and AAV9 were the most powerful capsids to infect mouse Müller glial cells. Retinaldehyde-binding protein 1 (RLBP1) promoter was the most powerful promoter to transduce Müller glial cells. ShH10Y capsids and RLBP1 promoter targeted human Müller glial cells in vitro. We also developed and tested smaller promoters to express the large CRB1 gene via AAV vectors. Minimal cytomegalovirus (CMV) promoter allowed expression of full-length CRB1 protein in Müller glial cells. In summary, ShH10Y and AAV9 capsids, and RLBP1 or minimal CMV promoters are of interest as specific tools to target and express in mouse or human Müller glial cells.
Collapse
Affiliation(s)
- Lucie P Pellissier
- Department of Neuromedical Genetics, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| | - Robert M Hoek
- Department of Neuromedical Genetics, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| | - Rogier M Vos
- Department of Neuromedical Genetics, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| | - Wendy M Aartsen
- Department of Neuromedical Genetics, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| | - Ryan R Klimczak
- Department of Molecular and Cellular Biology and The Helen Wills Neuroscience Institute, University of California , Berkeley, California, USA
| | - Stefan A Hoyng
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| | - John G Flannery
- Department of Molecular and Cellular Biology and The Helen Wills Neuroscience Institute, University of California , Berkeley, California, USA
| | - Jan Wijnholds
- Department of Neuromedical Genetics, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences , Amsterdam, The Netherlands
| |
Collapse
|
29
|
Vacca O, Darche M, Schaffer DV, Flannery JG, Sahel JA, Rendon A, Dalkara D. AAV-mediated gene delivery in Dp71-null mouse model with compromised barriers. Glia 2013; 62:468-76. [PMID: 24382652 DOI: 10.1002/glia.22617] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 11/21/2013] [Accepted: 11/22/2013] [Indexed: 12/28/2022]
Abstract
Formation and maintenance of the blood-retinal barrier (BRB) is required for proper vision and breaching of this barrier contributes to the pathology in a wide variety of retinal conditions such as retinal detachment and diabetic retinopathy. Dystrophin Dp71 being a key membrane cytoskeletal protein, expressed mainly in Müller cells, its absence has been related to BRB permeability through delocalization and down-regulation of the AQP4 and Kir4.1 channels. Dp71-null mouse is thus an excellent model to approach the study of retinal pathologies showing blood-retinal barrier permeability. We aimed to investigate the participation of Müller cells in the BRB and in the inner limiting membrane of Dp71-null mice compared with wild-type mice in order to understand how these barriers work in this model of permeable BRB. To this aim, we used an Adeno-associated virus (AAV) variant, ShH10-GFP, engineered to target Müller cells specifically. ShH10 coding GFP was introduced by intravitreal injection and Müller cell transduction was studied in Dp71-null mice in comparison to wild-type animals. We show that Müller cell transduction follows a significantly different pattern in Dp71-null mice indicating changes in viral cell-surface receptors as well as differences in the permeability of the inner limiting membrane in this mouse line. However, the compromised BRB of the Dp71-null mice does not lead to virus leakage into the bloodstream when the virus is injected intravitreally - an important consideration for AAV-mediated retinal gene therapy.
Collapse
Affiliation(s)
- Ophélie Vacca
- Institut de la Vision/INSERM/UPMC, Univ Paris 06/CNRS/CHNO des Quinze-Vingts, Paris, France
| | | | | | | | | | | | | |
Collapse
|
30
|
Cai X, Seal S, McGinnis JF. Sustained inhibition of neovascularization in vldlr-/- mice following intravitreal injection of cerium oxide nanoparticles and the role of the ASK1-P38/JNK-NF-κB pathway. Biomaterials 2013; 35:249-58. [PMID: 24140045 DOI: 10.1016/j.biomaterials.2013.10.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 10/04/2013] [Indexed: 02/06/2023]
Abstract
Cerium oxide nanoparticles (nanoceria) are direct antioxidants; they inhibit pathological neovascularization following a single intravitreal injection into new born very low density lipoprotein receptor knockout (vldlr(-/-)) mice. However, the long-term therapeutic effects and mechanisms of nanoceria action on regression of the existing pathologic neovascularization in the eyes are unknown. We intravitreally injected P28 vldlr(-/-) mice and extended the endpoint for analysis until P70. The data demonstrate that nanoceria sustained their therapeutic function up to 6 weeks. Multiple parameters for nanoceria effects were examined including: regression of existing abnormal blood vessels, reduction of vascular leakage, down-regulation of the expression of vascular endothelial growth factor (VEGF), acrolein, glial fibrillary acidic protein (GFAP) and caspase 3 as well as up-regulation of the expression of rod- and cone-opsin genes. Regulation of ASK1-P38/JNK-NF-κB signaling pathway by nanoceria was investigated. Our data demonstrated that a single intravitreal injection of nanoceria in P28 vldlr(-/-) mice produced sustained regression of existing oxidative stress-induced neovascularizations, prevented blood vessel leakage and inhibited apoptosis via down-regulation of the ASK1-P38/JNK-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xue Cai
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | | | | |
Collapse
|
31
|
Bemelmans AP, Duqué S, Rivière C, Astord S, Desrosiers M, Marais T, Sahel JA, Voit T, Barkats M. A single intravenous AAV9 injection mediates bilateral gene transfer to the adult mouse retina. PLoS One 2013; 8:e61618. [PMID: 23613884 PMCID: PMC3626698 DOI: 10.1371/journal.pone.0061618] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 03/15/2013] [Indexed: 11/25/2022] Open
Abstract
Widespread gene delivery to the retina is an important challenge for the treatment of retinal diseases, such as retinal dystrophies. We and others have recently shown that the intravenous injection of a self-complementary (sc) AAV9 vector can direct efficient cell transduction in the central nervous system, in both neonatal and adult animals. We show here that the intravenous injection of scAAV9 encoding green fluorescent protein (GFP) resulted in gene transfer to all layers of the retina in adult mice, despite the presence of a mature blood-eye barrier. Cell morphology studies and double-labeling with retinal cell-specific markers showed that GFP was expressed in retinal pigment epithelium cells, photoreceptors, bipolar cells, Müller cells and retinal ganglion cells. The cells on the inner side of the retina, including retinal ganglion cells in particular, were transduced with the highest efficiency. Quantification of the cell population co-expressing GFP and Brn-3a showed that 45% of the retinal ganglion cells were efficiently transduced after intravenous scAAV9-GFP injection in adult mice. This study provides the first demonstration that a single intravenous scAAV9 injection can deliver transgenes to the retinas of both eyes in adult mice, suggesting that this vector serotype is able to cross mature blood-eye barriers. This intravascular gene transfer approach, by eliminating the potential invasiveness of ocular surgery, could constitute an alternative when fragility of the retina precludes subretinal or intravitreal injections of viral vectors, opening up new possibilities for gene therapy for retinal diseases.
Collapse
Affiliation(s)
- Alexis-Pierre Bemelmans
- INSERM, U968, Paris, France
- Institut de la Vision, Université Pierre et Marie Curie Paris 6 - UM80, Paris, France
- UMR_7210, CNRS, Paris, France
- Molecular Imaging Research Center (MIRCen) and CNRS URA2210, Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Département des Sciences du Vivant (DSV), Institut d’Imagerie Biomédicale (I2BM), Fontenay-aux-Roses, France
| | - Sandra Duqué
- UM76 UPMC-AIM UMR S974, INSERM U 974, CNRS UMR 7215, Institut de Myologie, Université Pierre et Marie Curie Paris 6, Paris, France
| | | | - Stéphanie Astord
- UM76 UPMC-AIM UMR S974, INSERM U 974, CNRS UMR 7215, Institut de Myologie, Université Pierre et Marie Curie Paris 6, Paris, France
| | - Mélissa Desrosiers
- INSERM, U968, Paris, France
- Institut de la Vision, Université Pierre et Marie Curie Paris 6 - UM80, Paris, France
- UMR_7210, CNRS, Paris, France
| | - Thibault Marais
- UM76 UPMC-AIM UMR S974, INSERM U 974, CNRS UMR 7215, Institut de Myologie, Université Pierre et Marie Curie Paris 6, Paris, France
| | - José-Alain Sahel
- INSERM, U968, Paris, France
- Institut de la Vision, Université Pierre et Marie Curie Paris 6 - UM80, Paris, France
- UMR_7210, CNRS, Paris, France
- INSERM-DHOS CIC 503, Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, Paris, France
| | - Thomas Voit
- UM76 UPMC-AIM UMR S974, INSERM U 974, CNRS UMR 7215, Institut de Myologie, Université Pierre et Marie Curie Paris 6, Paris, France
| | - Martine Barkats
- UM76 UPMC-AIM UMR S974, INSERM U 974, CNRS UMR 7215, Institut de Myologie, Université Pierre et Marie Curie Paris 6, Paris, France
- * E-mail:
| |
Collapse
|
32
|
Autoantibody against transient receptor potential M1 cation channels of retinal ON bipolar cells in paraneoplastic vitelliform retinopathy. BMC Ophthalmol 2012; 12:56. [PMID: 23148706 PMCID: PMC3514129 DOI: 10.1186/1471-2415-12-56] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 11/07/2012] [Indexed: 01/09/2023] Open
Abstract
Background Paraneoplastic retinopathy is caused by the cross-reaction of neoplasm-directed autoantibodies against retinal antigens and results in retinal damage. Paraneoplastic vitelliform retinopathy, a presumed paraneoplastic retinopathy with features of atypical melanoma-associated retinopathy, has recently been reported in patients with metastatic melanoma. Ocular ultrastructure and its autoantibody localization of paraneoplastic vitelliform retinopathy are still indefinable. This is the first report of anti-transient receptor potential M1 antibody directly against human retinal bipolar dendritic tips in a melanoma patient with paraneoplastic vitelliform retinopathy. Case presentation We present a pair of postmortem eyes of an 80-year-old male with metastatic cutaneous melanoma, who developed paraneoplastic vitelliform retinopathy. The autopsied eyes were examined with light microscopy, immunohistochemistry, and transmission electron microscopy. Microscopically, the inner nuclear layer and outer plexiform layer were the most affected retinal structures, with local thinning. The lesions extended to the outer nuclear layer, resulting in focal retinal degeneration, edema, and atrophy. No active inflammation or melanoma cells were observed. Immunohistochemistry showed tightly compact bipolar cell nuclei (protein kinase C alpha/calbindin positive) with blur/loss of ON bipolar cell dendritic tips (transient receptor potential M1 positive) in diffusely condensed outer plexiform layer. The metastatic melanoma cells in his lung also showed immunoreactivity against transient receptor potential M1 antibody. Transmission electron microscopy illustrated degenerated inner nuclear layer with disintegration of cells and loss of cytoplasmic organelles. These cells contained many lysosomal and autophagous bodies and damaged mitochondria. Their nuclei appeared pyknotic and fragmentary. The synapses in the outer plexiform layer were extensively degenerated and replaced with empty vacuoles and disintegrated organelles. Conclusion This case provides a convincing histological evidence of melanoma-associated autoantibodies directly against transient receptor potential M1 channels that target the ON bipolar cell structures in the inner nuclear and outer plexiform layers in paraneoplastic vitelliform retinopathy.
Collapse
|
33
|
Hoyng SA, Tannemaat MR, De Winter F, Verhaagen J, Malessy MJA. Nerve surgery and gene therapy: a neurobiological and clinical perspective. J Hand Surg Eur Vol 2011; 36:735-46. [PMID: 21914696 DOI: 10.1177/1753193411420348] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Despite major microsurgical improvements the clinical outcome of peripheral nerve surgery is still regarded as suboptimal. Over the past decade several innovative techniques have been developed to extend the armamentarium of the nerve surgeon. This review evaluates the potential of gene therapy in the context of peripheral nerve repair. First the main challenges impeding peripheral nerve regeneration are presented. This is followed by a short introduction to gene therapy and an overview of its most important advantages over the classical delivery of therapeutic proteins. Next, this review focuses on the most promising viral vectors capable of targeting the peripheral nervous system and their first application in animal models. In addition, the challenges of translating these experimental results to the clinic, the limitations of current vectors and the further developments needed, are discussed. Finally, four strategies are presented on how gene therapy could help patients that have to undergo reconstructive nerve surgery in the future.
Collapse
Affiliation(s)
- S A Hoyng
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Royal Academy of Arts and Sciences, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
34
|
Vandenberghe LH, Auricchio A. Novel adeno-associated viral vectors for retinal gene therapy. Gene Ther 2011; 19:162-8. [PMID: 21993172 DOI: 10.1038/gt.2011.151] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vectors derived from adeno-associated virus (AAV) are currently the most promising vehicles for therapeutic gene delivery to the retina. Recently, subretinal administration of AAV2 has been demonstrated to be safe and effective in patients with a rare form of inherited childhood blindness, suggesting that AAV-mediated retinal gene therapy may be successfully extended to other blinding conditions. This is further supported by the great versatility of AAV as a vector platform as there are a large number of AAV variants and many of these have unique transduction characteristics useful for targeting different cell types in the retina including glia, epithelium and many types of neurons. Naturally occurring, rationally designed or in vitro evolved AAV vectors are currently being utilized to transduce several different cell types in the retina and to treat a variety of animal models of retinal disease. The continuous and creative development of AAV vectors provides opportunities to overcome existing challenges in retinal gene therapy such as efficient transfer of genes exceeding AAV's cargo capacity, or the targeting of specific cells within the retina or transduction of photoreceptors following routinely used intravitreal injections. Such developments should ultimately advance the treatment of a wide range of blinding retinal conditions.
Collapse
Affiliation(s)
- L H Vandenberghe
- Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA.
| | | |
Collapse
|
35
|
Calame M, Cachafeiro M, Philippe S, Schouwey K, Tekaya M, Wanner D, Sarkis C, Kostic C, Arsenijevic Y. Retinal degeneration progression changes lentiviral vector cell targeting in the retina. PLoS One 2011; 6:e23782. [PMID: 21901134 PMCID: PMC3161995 DOI: 10.1371/journal.pone.0023782] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 07/27/2011] [Indexed: 11/19/2022] Open
Abstract
In normal mice, the lentiviral vector (LV) is very efficient to target the RPE cells, but transduces retinal neurons well only during development. In the present study, the tropism of LV has been investigated in the degenerating retina of mice, knowing that the retina structure changes during degeneration. We postulated that the viral transduction would be increased by the alteration of the outer limiting membrane (OLM). Two different LV pseudotypes were tested using the VSVG and the Mokola envelopes, as well as two animal models of retinal degeneration: light-damaged Balb-C and Rhodopsin knockout (Rho-/-) mice. After light damage, the OLM is altered and no significant increase of the number of transduced photoreceptors can be obtained with a LV-VSVG-Rhop-GFP vector. In the Rho-/- mice, an alteration of the OLM was also observed, but the possibility of transducing photoreceptors was decreased, probably by ongoing gliosis. The use of a ubiquitous promoter allows better photoreceptor transduction, suggesting that photoreceptor-specific promoter activity changes during late stages of photoreceptor degeneration. However, the number of targeted photoreceptors remains low. In contrast, LV pseudotyped with the Mokola envelope allows a wide dispersion of the vector into the retina (corresponding to the injection bleb) with preferential targeting of Müller cells, a situation which does not occur in the wild-type retina. Mokola-pseudotyped lentiviral vectors may serve to engineer these glial cells to deliver secreted therapeutic factors to a diseased area of the retina.
Collapse
Affiliation(s)
- Maritza Calame
- Unit of Gene Therapy and Stem Cell Biology, Service of Ophthalmology, Jules-Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | - Maité Cachafeiro
- Unit of Gene Therapy and Stem Cell Biology, Service of Ophthalmology, Jules-Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | - Stéphanie Philippe
- Unit of Gene Therapy and Stem Cell Biology, Service of Ophthalmology, Jules-Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | - Karine Schouwey
- Unit of Gene Therapy and Stem Cell Biology, Service of Ophthalmology, Jules-Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | - Meriem Tekaya
- Unit of Gene Therapy and Stem Cell Biology, Service of Ophthalmology, Jules-Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | - Dana Wanner
- Unit of Gene Therapy and Stem Cell Biology, Service of Ophthalmology, Jules-Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | - Chamsy Sarkis
- NewVectys SAS, Paris, France
- Team of Biotherapy and Biotechnology, CRICM, UPMC-Paris6 UMR_S 975, INSERM U975, CNRS UMR 7225, Paris, France
| | - Corinne Kostic
- Unit of Gene Therapy and Stem Cell Biology, Service of Ophthalmology, Jules-Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | - Yvan Arsenijevic
- Unit of Gene Therapy and Stem Cell Biology, Service of Ophthalmology, Jules-Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|