98 results on '"Deniz Dalkara"'
Search Results
2. Supplementary Fig. S3 from Suppression of cervical carcinoma cell growth by intracytoplasmic codelivery of anti-oncoprotein E6 antibody and small interfering RNA
- Author
-
Etienne Weiss, Guy Zuber, Mustapha Oulad-Abdelghani, Deniz Dalkara, Gabrielle Zeder-Lutz, Annie-Paule Sibler, and Jérôme Courtête
- Abstract
Supplementary Fig. S3 from Suppression of cervical carcinoma cell growth by intracytoplasmic codelivery of anti-oncoprotein E6 antibody and small interfering RNA
- Published
- 2023
3. Supplementary Table from Suppression of cervical carcinoma cell growth by intracytoplasmic codelivery of anti-oncoprotein E6 antibody and small interfering RNA
- Author
-
Etienne Weiss, Guy Zuber, Mustapha Oulad-Abdelghani, Deniz Dalkara, Gabrielle Zeder-Lutz, Annie-Paule Sibler, and Jérôme Courtête
- Abstract
Supplementary Table from Suppression of cervical carcinoma cell growth by intracytoplasmic codelivery of anti-oncoprotein E6 antibody and small interfering RNA
- Published
- 2023
4. Data from Suppression of cervical carcinoma cell growth by intracytoplasmic codelivery of anti-oncoprotein E6 antibody and small interfering RNA
- Author
-
Etienne Weiss, Guy Zuber, Mustapha Oulad-Abdelghani, Deniz Dalkara, Gabrielle Zeder-Lutz, Annie-Paule Sibler, and Jérôme Courtête
- Abstract
Cervical cancer is caused by high-risk types of human papillomaviruses (HPV) that encode the E6 and E7 oncogenes. Silencing of E6 gene expression in HPV-positive cell lines by transfection of small interfering RNA (siRNA) with cationic lipids restores the dormant p53 tumor suppressor pathway. Because cationic lipids can also be used for intracytoplasmic delivery of proteins, we tested whether the delivery of monoclonal antibodies that bind to HPV16 E6 and neutralize its biological activity in vitro could restore p53 function in tumor cells. Here, we show that the 4C6 antibody is efficiently delivered into the cell cytoplasm using a lipidic reagent used for siRNA transfection. The delivery of 4C6 resulted in the nuclear accumulation of p53 protein in CaSki and SiHa cells but not in HeLa cells. Furthermore, the antibody-mediated p53 response was dramatically increased when a peptide corresponding to the 4C6 epitope and bearing a COOH-terminal cysteine residue was added to the transduction mixture. We found that a fraction of the added peptides were dimers that allowed the formation of antibody polymers adsorbed onto the lipidic matrix. With this system, the proliferation of CaSki and SiHa cells was strongly diminished, but no apoptosis was detectable. Remarkably, cell growth was almost totally suppressed by the addition of E6-specific siRNA to the transduction complex. The results indicate that the activity of E6 oncoprotein can be down-regulated in vivo by lipid-mediated antibody delivery and that antibodies and siRNA act synergistically when codelivered. This novel targeting strategy is simple to implement and may find therapeutic applications. [Mol Cancer Ther 2007;16(5):1728–35]
- Published
- 2023
5. Uncursing winner’s curse: on-line monitoring of directed evolution convergence
- Author
-
Takahiro Nemoto, Tommaso Ocari, Arthur Planul, Muge Tekinsoy, Emilia A. Zin, Deniz Dalkara, and Ulisse Ferrari
- Abstract
Directed evolution (DE) is a versatile protein-engineering strategy, successfully applied to a range of proteins, including enzymes, antibodies, and viral vectors. However, DE can be time-consuming and costly, as it typically requires many rounds of selection to identify desired mutants. Next-generation sequencing allows monitoring of millions of variants during DE and can be leveraged to reduce the number of selection rounds. Unfortunately the noisy nature of the sequencing data impedes the estimation of the performance of individual variants. Here, we propose ACIDES that combines statistical inference and in-silico simulations to improve performance estimation in DE by providing accurate statistical scores. We tested ACIDES first on a novel random-peptide-insertion experiment and then on several public datasets from DE of viral vectors and phage-display. ACIDES allows experimentalists to reliably estimate variant performanceon the flyand can aid protein engineering pipelines in a range of applications, including gene therapy.
- Published
- 2023
6. Cellular Immune Responses Induced by Subretinal AAV Gene Transfer can be Restrained by the Subretinal Associated Immune Inhibition Mechanism
- Author
-
Julie Vendomèle, Gaëlle Anne Chauveau, Deniz Dalkara, Anne Galy, and Sylvain Fisson
- Abstract
For more than a decade, AAV-mediated gene transfer has been tested successfully in clinical trials to treat inherited retinal diseases. Despite the eye’s immune-privileged status and the use of corticoids as an adjunct treatment, some patients display inflammatory events which led us to question the immune consequences of a subretinal AAV administration. We first characterized anti-transgene immune responses induced in the periphery by injecting increasing doses of AAV8 encoding reporter proteins fused with the HY male antigen into the subretinal space of female C57BL/6 and rd10 mice. Transgene expression was monitored over time with bioluminescence imaging and T-cell immune responses in the spleen were analyzed by IFNγ ELISpot and cytokine multiplex assays. Our data show that an AAV8 injection causes proinflammatory T-cell immune response against the transgene product, correlated with the transgene expression level at 2.109vg and above. Additionally, co-injection of immunodominant peptides from the transgene product, along with AAV8, modulates the immune response at all AAV doses tested. Taken together, our data suggest that injection of AAV8 in the subretinal space induces proinflammatory peripheral T-cell responses to the transgene product that can be modulated by the subretinal associated immune inhibition (SRAII) mechanism.
- Published
- 2023
7. Inducible non-human primate models of retinal degeneration for testing end stage therapies and understanding disease mechanisms
- Author
-
Divya Ail, Diane Nava, In Pyo Hwang, Elena Brazhnikova, Céline Nouvel-Jaillard, Alexandre Dentel, Corentin Joffrois, Lionel Rousseau, Julie Dégardin, Stephane Bertin, José-Alain Sahel, Olivier Goureau, Serge Picaud, and Deniz Dalkara
- Abstract
The anatomical differences between the retinas of humans and most animal models pose a challenge for testing novel therapies. Non-human primate (NHP) retina is anatomically closest to the human retina with the presence of a high acuity region called the fovea. However, there is a lack of relevant NHP models for retinal degeneration that can be used for preclinical studies of vision restoration. To address this unmet need we aimed to generate inducible NHP models of photoreceptor degeneration. We generated three cynomolgus macaque models using distinct strategies. We used two genetically targeted strategies using optogenetics and Crispr-Cas9 to ablate specifically rods to mimic rod-cone dystrophy. Additionally, we created an acute model by physical separation of the photoreceptors and retinal pigment epithelium using a polymer patch. Retinal degeneration was evaluated in all three models by in-life exams such as fundus imaging, optical coherence tomography, adaptive optics and electroretinography. In the genetic models we observed punctuate areas of degeneration in the injected area marked by disorganization of outer segments, loss of rod photoreceptors and thinning of the outer nuclear layer. In the acute model, the degeneration was faster and involved both rods and cones. Among the three distinct NHP models, the Crispr-Cas9 based approach was the most advantageous model in view of recapitulating disease specific features and its ease of implementation. The acute model however resulted in the fastest degeneration making it the most relevant model for testing end-stage vision restoration therapies such as stem cell transplantation.
- Published
- 2022
8. Assessing Photoreceptor Status in Retinal Dystrophies: From High-Resolution Imaging to Functional Vision
- Author
-
Michel Paques, Chloé Pagot, Christina Zeitz, José-Alain Sahel, Deniz Dalkara, Isabelle Audo, Serge Picaud, Saddek Mohand-Said, Line Azoulay, Kate Grieve, Katia Marazova, Thierry Leveillard, Karine Becker, Colas N. Authié, Emmanuel Gutman, Anne-Elisabeth Chaumet-Riffaud, Marazova, Katia, University of Pittsburgh School of Medicine, Pennsylvania Commonwealth System of Higher Education (PCSHE), Sorbonne Université (SU), Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts (CHNO), Institut de la Vision, and Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
medicine.medical_specialty ,Visual acuity ,genetic structures ,[SDV]Life Sciences [q-bio] ,media_common.quotation_subject ,Visual impairment ,Adaptation (eye) ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,03 medical and health sciences ,0302 clinical medicine ,Foveal ,Ophthalmology ,Retinal Dystrophies ,Humans ,AOS Thesis ,Medicine ,Contrast (vision) ,External limiting membrane ,Retrospective Studies ,030304 developmental biology ,media_common ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,0303 health sciences ,business.industry ,eye diseases ,Visual field ,[SDV] Life Sciences [q-bio] ,Cross-Sectional Studies ,medicine.anatomical_structure ,Retinal Cone Photoreceptor Cells ,030221 ophthalmology & optometry ,sense organs ,medicine.symptom ,business ,Tomography, Optical Coherence - Abstract
International audience; Purpose: To describe the value of integrating phenotype/genotype data, disease staging, and evaluation of functional vision in patient-centered management of retinal dystrophies.Methods: (1) Cross-sectional structure-function and retrospective longitudinal studies to assess the correlations between standard fundus autofluorescence (FAF), optical coherence tomography, visual acuity (VA), and perimetry (visual field [VF]) examinations to evaluate photoreceptor functional loss in a cohort of patients with rod-cone dystrophy (RCD); (2) flood-illumination adaptive optics (FIAO) imaging focusing on photoreceptor misalignment and orientation of outer segments; and (3) evaluation of the impact of visual impairment in daily life activities, based on functional (visual and mobility) vision assessment in a naturalistic environment in visually impaired subjects with RCD and subjects treated with LuxturnaⓇ for RPE65-related Leber congenital amaurosis before and after therapy.Results: The results of the cross-sectional transversal study showed that (1) VA and macular sensitivity were weakly correlated with the structural variables; and (2) functional impairment (VF) was correlated with reduction of anatomical markers of photoreceptor structure and increased width of autofluorescent ring. The dimensions of the ring of increased FAF evolved faster. Other criteria that differed among groups were the lengths of the ellipsoid zone, the external limiting membrane, and the foveal thickness. FIAO revealed a variety of phenotypes: paradoxical visibility of foveal cones; heterogeneous brightness of cones; dim, inner segment-like, and RPE-like mosaic. Directional illumination by varying orientation of incident light (Stiles-Crawford effect) and the amount of side illumination (gaze-dependent imaging) affected photoreceptor visibility. Mobility assessment under different lighting conditions showed correlation with VF, VA, contrast sensitivity (CS), and dark adaptation, with different predictive values depending on mobility study paradigms and illumination level. At high illumination level (235 lux), VF was a predictor for all mobility performance models. Under low illumination (1 and 2 lux), VF was the most significant predictor of mobility performance variables, while CS best explained the number of collisions and segments. In subjects treated with LuxturnaⓇ, a very favorable impact on travel speed and reduction in the number of collisions, especially at low luminance, was observable 6 months following injection, in both children and adults.Conclusions: Our results suggest the benefit of development and implementation of quantitative and reproducible tools to evaluate the status of photoreceptors and the impact of both visual impairment and novel therapies in real-life conditions. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.
- Published
- 2021
9. Substantial restoration of night vision in adult mice with congenital stationary night blindness
- Author
-
Miguel Miranda de Sousa Dias, José-Alain Sahel, Serge Picaud, Christelle Michiels, Melissa Desrosiers, Isabelle Audo, Marco Nassisi, Camille Robert, Deniz Dalkara, Christina Zeitz, Marion Neuillé, Nassima Bouzidi, Corentin Joffrois, Juliette Varin, Gregory Gauvain, Institut de la Vision, Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Hôpital de la Fondation Ophtalmologique Adolphe de Rothschild [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Académie des Sciences [Paris], Institut de France, University of Pittsburgh School of Medicine, Pennsylvania Commonwealth System of Higher Education (PCSHE), Centre d'investigation clinique Quinze-Vingts [CHNO] (CIC1423 - CIC QUINZE-VINGTS), Institut Hospitalo-Universitaire FOReSIGHT, Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts (CHNO)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts (CHNO)-Sorbonne Université (SU), University College of London [London] (UCL), HAL-SU, Gestionnaire, Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts (CHNO), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), and Académie des Sciences
- Subjects
bipolar cells ,medicine.medical_specialty ,Retinal Disorder ,Outer plexiform layer ,QH426-470 ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Night vision ,Genetics ,medicine ,Scotopic vision ,[SDV.MHEP.OS]Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,CSNB ,Molecular Biology ,TRPM1 ,030304 developmental biology ,congenital stationary night blindness ,Congenital stationary night blindness ,0303 health sciences ,QH573-671 ,business.industry ,photoreceptors ,AAV ,gene therapy ,3. Good health ,Ganglion ,Endocrinology ,medicine.anatomical_structure ,[SDV.MHEP.OS] Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,030221 ophthalmology & optometry ,Molecular Medicine ,Original Article ,LRIT3 ,sense organs ,Cytology ,business ,Erg - Abstract
Complete congenital stationary night blindness (cCSNB) due to mutations in TRPM1, GRM6, GPR179, NYX, or leucine-rich repeat immunoglobulin-like transmembrane domain 3 (LRIT3) is an incurable inherited retinal disorder characterized by an ON-bipolar cell (ON-BC) defect. Since the disease is non-degenerative and stable, treatment could theoretically be administrated at any time in life, making it a promising target for gene therapy. Until now, adeno-associated virus (AAV)-mediated therapies lead to significant functional improvements only in newborn cCSNB mice. Here we aimed to restore protein localization and function in adult Lrit3−/− mice. LRIT3 localizes in the outer plexiform layer and is crucial for TRPM1 localization at the dendritic tips of ON-BCs and the electroretinogram (ERG)-b-wave. AAV2-7m8-Lrit3 intravitreal injections were performed targeting either ON-BCs, photoreceptors (PRs), or both. Protein localization of LRIT3 and TRPM1 at the rod-to-rod BC synapse, functional rescue of scotopic responses, and ON-responses detection at the ganglion cell level were achieved in a few mice when ON-BCs alone or both PRs and ON-BCs, were targeted. More importantly, a significant number of treated adult Lrit3−/− mice revealed an ERG b-wave recovery under scotopic conditions, improved optomotor responses, and on-time ON-responses at the ganglion cell level when PRs were targeted. Functional rescue was maintained for at least 4 months after treatment., Graphical abstract, Zeitz and colleagues report in this study strong restoration of scotopic function in adult Lrit3−/− mice affected with complete congenital stationary night blindness as shown by a gene therapy approach targeting bipolar and/or photoreceptor cells. Findings were validated by immunolocalization studies, electroretinogram, multi-electrode array measurements, and optomotor response evaluation.
- Published
- 2021
10. Numb regulates Tau levels and prevents neurodegeneration in tauopathy mouse models
- Author
-
Marine Lacomme, Sarah C. Hales, Thomas W. Brown, Katarina Stevanovic, Christine Jolicoeur, Jenny Cai, Therence Bois, Melissa Desrosiers, Deniz Dalkara, and Michel Cayouette
- Subjects
Retinal Ganglion Cells ,Mice ,Disease Models, Animal ,Multidisciplinary ,Tauopathies ,Animals ,Membrane Proteins ,tau Proteins ,Nerve Tissue Proteins ,Axons - Abstract
Accumulation of the microtubule-associated protein Tau is linked to neuronal cell death in tauopathies, but how intraneuronal Tau levels are regulated in health and disease remains unclear. Here, we show that conditional inactivation of the trafficking adaptor protein Numb in retinal ganglion cells (RGCs) increases Tau levels and leads to axonal blebbing, which is followed by neuronal cell loss in aged mice. In the TauP301S mouse model of tauopathy, conditional inactivation of Numb in RGCs and spinal motoneurons accelerates neurodegeneration, and loss of Numb in motoneurons also leads to precocious hindlimb paralysis. Conversely, overexpression of the long isoform of Numb (Numb-72) decreases intracellular Tau levels and reduces axonal blebbing in TauP301S RGCs, leading to improved electrical activity in cultured neurons and improves performance in a visually guided behavior test in vivo. These results uncover Numb as a key regulator of intracellular Tau levels and identify Numb-72 as a potential therapeutic factor for tauopathies.
- Published
- 2022
11. Immune Responses to Gene Editing by Viral and Non-Viral Delivery Vectors Used in Retinal Gene Therapy
- Author
-
Duohao Ren, Sylvain Fisson, Deniz Dalkara, Divya Ail, Institut de la Vision, Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Approches génétiques intégrées et nouvelles thérapies pour les maladies rares (INTEGRARE), Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay-Généthon, and Ail, Divya
- Subjects
[SDV.IMM] Life Sciences [q-bio]/Immunology ,Adeno-associated virus ,innate immune response ,Pharmaceutical Science ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,CRISPR-Cas9 ,immune responses ,adaptive immune response ,ocular gene therapy - Abstract
International audience; Inherited retinal diseases (IRDs) are a leading cause of blindness in industrialized countries, and gene therapy is quickly becoming a viable option to treat this group of diseases. Gene replacement using a viral vector has been successfully applied and advanced to commercial use for a rare group of diseases. This, and the advances in gene editing, are paving the way for the emergence of a new generation of therapies that use CRISPR-Cas9 to edit mutated genes in situ. These CRISPR-based agents can be delivered to the retina as transgenes in a viral vector, unpackaged transgenes or as proteins or messenger RNA using non-viral vectors. Although the eye is considered to be an immune-privileged organ, studies in animals, as well as evidence from clinics, have concluded that ocular gene therapies elicit an immune response that can under certain circumstances result in inflammation. In this review, we evaluate studies that have reported on pre-existing immunity, and discuss both innate and adaptive immune responses with a specific focus on immune responses to gene editing, both with non-viral and viral delivery in the ocular space. Lastly, we discuss approaches to prevent and manage the immune responses to ensure safe and efficient gene editing in the retina.
- Published
- 2022
12. Optogenetic Targeting of AII Amacrine Cells restores Retinal Computations performed by the Inner Retina
- Author
-
Hanen Khabou, Elaine Orendorff, Francesco Trapani, Marco Rucli, Melissa Desrosiers, Pierre Yger, Deniz Dalkara, and Olivier Marre
- Abstract
Most inherited retinal dystrophies display progressive photoreceptor cell degeneration leading to severe visual impairment. Optogenetic reactivation of inner retinal neurons is a promising avenue to restore vision in retinas having lost their photoreceptors. Expression of optogenetic proteins in surviving ganglion cells, the retinal output, allows them to take on the lost photoreceptive function. Nonetheless, this creates an exclusively ON retina by expression of depolarizing optogenetic proteins in all classes of ganglion cells, whereas a normal retina extracts several features from the visual scene, with different ganglion cells detecting light increase (ON) and light decrease (OFF). Refinement of this therapeutic strategy should thus aim at restoring these computations. In an attempt to do so, we used a promoter that targets gene expression to a specific interneuron of the retina called the AII amacrine cell. The AII amacrine cell simultaneously activates the ON pathway and inhibits the OFF pathway. We show that the optogenetic stimulation of AII amacrine cells allows restoration of both ON and OFF responses in the retina, but also mediates other types of retinal processing such as sustained and transient responses. Targeting amacrine cells with optogenetics is thus a promising avenue to restore better retinal function and visual perception in patients suffering from retinal degeneration.
- Published
- 2022
13. Partial recovery of visual function in a blind patient after optogenetic therapy
- Author
-
Laure Blouin, Jean-Baptiste de Saint Aubert, Deniz Dalkara, Joseph N. Martel, Alexandre Delaux, Simona Degli Esposti, Serge Picaud, Emmanuel Gutman, Francesco Galluppi, Chloé Pagot, Caroline de Montleau, Magali Taiel, Botond Roska, José-Alain Sahel, Elise Boulanger-Scemama, Angelo Arleo, Jens Duebel, Isabelle Audo, Institut de la Vision, Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Streetlab [Paris], Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), GenSight Biologics, Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA, National Institute for Health Research Moorfields Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, INSERM-Centre d'Investigation Clinique 1423, Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, F-75012 Paris, France, University of Basel (Unibas), ARLEO, Angelo, Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM), and Department of Ophthalmology, University of Basel, Basel, Switzerland
- Subjects
Male ,Retinal Ganglion Cells ,0301 basic medicine ,Visual perception ,genetic structures ,Eye disease ,Genetic Vectors ,Review Article ,Optogenetics ,Blindness ,Retinal ganglion ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Retinitis pigmentosa ,Humans ,Medicine ,Photoreceptor Cells ,[SDV.NEU] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,Vision, Ocular ,ComputingMilieux_MISCELLANEOUS ,Visual Cortex ,Retina ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,business.industry ,Genetic Therapy ,General Medicine ,Dependovirus ,Middle Aged ,medicine.disease ,Brain Waves ,eye diseases ,3. Good health ,Light intensity ,030104 developmental biology ,Visual cortex ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Visual Perception ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,sense organs ,Eye Protective Devices ,business ,Neuroscience ,Retinitis Pigmentosa ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
Optogenetics may enable mutation-independent, circuit-specific restoration of neuronal function in neurological diseases. Retinitis pigmentosa is a neurodegenerative eye disease where loss of photoreceptors can lead to complete blindness. In a blind patient, we combined intraocular injection of an adeno-associated viral vector encoding ChrimsonR with light stimulation via engineered goggles. The goggles detect local changes in light intensity and project corresponding light pulses onto the retina in real time to activate optogenetically transduced retinal ganglion cells. The patient perceived, located, counted and touched different objects using the vector-treated eye alone while wearing the goggles. During visual perception, multichannel electroencephalographic recordings revealed object-related activity above the visual cortex. The patient could not visually detect any objects before injection with or without the goggles or after injection without the goggles. This is the first reported case of partial functional recovery in a neurodegenerative disease after optogenetic therapy.
- Published
- 2021
14. Developing New Vectors for Retinal Gene Therapy
- Author
-
Emilia A. Zin, Bilge E. Ozturk, Deniz Dalkara, and Leah C. Byrne
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2023
15. All-optical inter-layers functional connectivity investigation in the mouse retina
- Author
-
Giulia Lia Beatrice Spampinato, Emiliano Ronzitti, Valeria Zampini, Ulisse Ferrari, Francesco Trapani, Hanen Khabou, Anurag Agraval, Deniz Dalkara, Serge Picaud, Eirini Papagiakoumou, Olivier Marre, and Valentina Emiliani
- Subjects
Genetics ,Radiology, Nuclear Medicine and imaging ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Biochemistry ,Computer Science Applications ,Biotechnology - Abstract
We developed a multi-unit microscope for all-optical inter-layers circuits interrogation. The system performs two-photon (2P) functional imaging and 2P multiplexed holographic optogenetics at axially distinct planes. We demonstrated the capability of the system to map, in the mouse retina, the functional connectivity between rod bipolar cells (RBCs) and ganglion cells (GCs) by activating single or defined groups of RBCs while recording the evoked response in the GC layer with cell-type specificity and single-cell resolution. We then used a logistic model to probe the functional connectivity between cell types by deriving the "cellular receptive field" describing how RBCs impact each GC type. With the capability to simultaneously image and control neuronal activity at axially distinct planes, the system enables a precise interrogation of multi-layered circuits. Understanding this information transfer is a promising avenue to dissect complex neural circuits and understand the neural basis of computations.
- Published
- 2022
16. Opsins for vision restoration
- Author
-
Jens Duebel, José-Alain Sahel, Cardillia-Joe Simon, Deniz Dalkara, and Stefan Herlitze
- Subjects
0301 basic medicine ,Retinal degeneration ,Opsin ,genetic structures ,Biophysics ,Stimulus (physiology) ,Optogenetics ,Biology ,Blindness ,Biochemistry ,Retina ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Retinitis pigmentosa ,medicine ,Animals ,Humans ,Molecular Biology ,Vision, Ocular ,Opsins ,Retinal Degeneration ,Retinal ,Cell Biology ,medicine.disease ,eye diseases ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Ectopic expression ,sense organs ,Neuroscience - Abstract
Optogenetics is a biological technique that combines the advantageous spatial-temporal resolution of optics and genetic cell targeting to control cellular activity with unprecedented precision. It has found vast applications both in neurosciences and therapy, particularly in view of its application to restore vision in blind patients. Optogenetics requires the ectopic expression of a so-called opsin to render neurons sensitive to light. There are two types of opsins for modulating membrane potential of neurons: (i) microbial opsins from unicellular organisms that respond to a light stimulus by mediating a flow of ions across the membrane (ii) animal opsins that are naturally present in mammalian retinas that initiate G protein coupled signaling in response to light. The former category has been extensively employed for vision restoration in the past decade with two ongoing clinical trials employing microbial opsins to restore light sensation in retinitis pigmentosa patients. The latter subtype of animal opsins is emerging more recently as strong candidates to restore vision with the promise of greater light sensitivity and tolerability. In this review we will discuss each approach in view of its utility for vision restoration in retinal blindness.
- Published
- 2020
17. Reactivating the phototransduction cascade by universally applicable gene therapy preserves retinal function in Rod-Cone dystrophy
- Author
-
Cardillia-Joe Simon, Hanen Khabou, Marion Finzi, Antoine Chaffiol, Kate Grieve, Nat Norberg, Anais Grimaud, Dennis Eickelbeck, Marco Rucli, Mélissa Desrosiers, Serge Sancho, Vera Bonilha, Saddek Mohand-Said, Jens Duebel, Michel paques, Serge Picaud, José-Alain Sahel, Isabelle Audo, Stefan Herlitze, and Deniz Dalkara
- Subjects
genetic structures ,sense organs ,eye diseases - Abstract
Rod-cone dystrophy (RCD), also known as retinitis pigmentosa, is a group of inherited disorders characterized by rod degeneration followed by degeneration of peripheral cones, leaving patients with tunnel vision in mid stages and blindness in the latest stages. Previous studies expressed microbial chloride pumps, in degenerating cone photoreceptors to restore these cells’ activity and prolong vision. Microbial opsins lack intracellular signal amplification, requiring high light intensities. To develop a more sensitive strategy; we examined the phototransduction cascade in degenerating cones, in two RCD mouse models. We found that opsin and arrestin expression is maintained in the soma during outer segment degeneration. We thus hypothesized that cone reactivation based on cone opsin signaling may be feasible by expressing a target channel activated by G proteins recruited by cone opsin. Adeno-associated viral (AAV) mediated expression of G protein coupled inwardly rectifying K (GIRK) channel provided improvements in visual function in two RCD mouse models with mutations in two distinct genes. Importantly, we confirmed cone opsin and cone arrestin expression in cones of late-stage RCD patients, validating the rationale of GIRK-mediated gene therapy in humans. We propose GIRK channel expression in cones as a new approach to maintain high acuity, high sensitivity, color vision in RCD independently of the underlying mutation.
- Published
- 2022
18. Gene therapy for inherited retinal disease: long-term durability of effect
- Author
-
Bart P. Leroy, M. Dominik Fischer, John G. Flannery, Robert E. MacLaren, Deniz Dalkara, Hendrik P.N. Scholl, Daniel C. Chung, Claudio Spera, Daniel Viriato, and Judit Banhazi
- Subjects
Cellular and Molecular Neuroscience ,Ophthalmology ,General Medicine ,Sensory Systems - Abstract
The recent approval of voretigene neparvovec (Luxturna®) for patients with biallelic RPE65 mutation-associated inherited retinal dystrophy with viable retinal cells represents an important step in the development of ocular gene therapies. Herein, we review studies investigating the episomal persistence of different recombinant adeno-associated virus (rAAV) vector genomes and the pre-clinical and clinical evidence of long-term effects of different RPE65 gene replacement therapies. A targeted review of articles published between 1974 and January 2021 in Medline®, Embase®, and other databases, was conducted, followed by a descriptive longitudinal analysis of the clinical trial outcomes of voretigene neparvovec. Following an initial screening, 14 publications examining the episomal persistence of different rAAV genomes and 71 publications evaluating gene therapies in animal models were included. Viral genomes were found to persist for at least 22 months (longest study follow-up) as transcriptionally active episomes. Treatment effects lasting almost a decade were reported in canine disease models, with more pronounced effects the earlier the intervention. The clinical trial outcomes of voretigene neparvovec are consistent with pre-clinical findings and reveal sustained results for up to 7.5 years for the full-field light sensitivity threshold test and 5 years for the multi-luminance mobility test in the Phase I and Phase III trials, respectively. In conclusion, the therapeutic effect of voretigene neparvovec lasts for at least a decade in animal models and 7.5 years in human subjects. Since retinal cells can retain functionality over their lifetime after transduction, these effects may be expected to last even longer in patients with a sufficient number of outer retinal cells at the time of intervention.
- Published
- 2021
19. Chronic nicotine increases midbrain dopamine neuron activity and biases individual strategies towards reduced exploration in mice
- Author
-
Alexandre Mourot, Etienne K. Duranté, Jérémie Naudé, Raphaël Brito, Tarek Ahmed Yahia, Fabio Marti, Malou Dongelmans, Nicolas Torquet, Philippe Faure, Claire Nguyen, B. Hannesse, Deniz Dalkara, Sarah Mondoloni, Damien Lemoine, Elise Bousseyrol, Steve Didienne, Maxime Come, Romain Durand-de Cuttoli, Lauren M. Reynolds, Neuroscience Paris Seine (NPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie Paris Seine (IBPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Paris Seine (IBPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Icahn School of Medicine at Mount Sinai [New York] (MSSM), Laboratoire Plasticité du Cerveau Brain Plasticity (UMR 8249) (PdC), Ecole Superieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS), Institut de la Vision, This work was supported by the Centre National de la Recherche Scientifique CNRS UMR 8246, INSERM U1130, the Foundation for Medical Research (FRM, Equipe FRM DEQ2013326488 to P.F.), FRM FDT201904008060 (to S.M.), the French National Cancer Institute Grant TABAC-16022 et TABAC-19-020 (to P.F.), French state funds managed by the ANR (ANR-16 Nicostress, ANR-17 SNP-Nic, ANR-20 Nicado to P.F., ANR-19 Vampire to F.M.) and The LabEx Bio-Psy (to P.F.). M.L.D., R.D.C., and S.M. were the recipients of a fourthyear PhD fellowship from FRM (FDT20160435171, FDT20170437427, and FDT201904008060), and faure, philippe
- Subjects
Male ,Nicotine ,Science ,Dopamine ,General Physics and Astronomy ,Self Administration ,Learning algorithms ,Optogenetics ,Biology ,Neural circuits ,Article ,General Biochemistry, Genetics and Molecular Biology ,Stereotaxic Techniques ,Midbrain ,Mice ,Reward ,Mesencephalon ,Archetypal analysis ,medicine ,Animals ,Tonic (music) ,[SDV.NEU] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,Multidisciplinary ,Behavior, Animal ,Dopaminergic Neurons ,General Chemistry ,medicine.anatomical_structure ,nervous system ,Chronic nicotine ,Models, Animal ,Exploratory Behavior ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,Neuron ,Neuroscience ,Prejudice ,medicine.drug - Abstract
Long-term exposure to nicotine alters brain circuits and induces profound changes in decision-making strategies, affecting behaviors both related and unrelated to drug seeking and consumption. Using an intracranial self-stimulation reward-based foraging task, we investigated in mice the impact of chronic nicotine on midbrain dopamine neuron activity and its consequence on the trade-off between exploitation and exploration. Model-based and archetypal analysis revealed substantial inter-individual variability in decision-making strategies, with mice passively exposed to nicotine shifting toward a more exploitative profile compared to non-exposed animals. We then mimicked the effect of chronic nicotine on the tonic activity of dopamine neurons using optogenetics, and found that photo-stimulated mice adopted a behavioral phenotype similar to that of mice exposed to chronic nicotine. Our results reveal a key role of tonic midbrain dopamine in the exploration/exploitation trade-off and highlight a potential mechanism by which nicotine affects the exploration/exploitation balance and decision-making., Chronic nicotine exposure impacts various components of decision-making processes, such as exploratory behaviors. Here, the authors identify the cellular mechanism and show that chronic nicotine exposure increases the tonic activity of VTA dopaminergic neurons and reduces exploration in mice.
- Published
- 2021
20. Sonogenetic stimulation of the brain at a spatiotemporal resolution suitable for vision restoration
- Author
-
Manuel Simonutti, Gregory Gauvain, I. Alcala, G. Labernede, Deniz Dalkara, Olivier Marre, Ja Sahel, D. Nelidova, M. Provansal, Emma Burban, S. Cadoni, Julie Degardin, Mickael Tanter, Fabrice Arcizet, Diep Nguyen, J. Lubetzki, Botond Roska, Charlie Demene, Serge Picaud, and R. Goulet
- Subjects
business.industry ,Computer science ,Ultrasound ,Stimulation ,Neuromodulation (medicine) ,Associative learning ,Visual cortex ,medicine.anatomical_structure ,In vivo ,medicine ,Premovement neuronal activity ,Mechanosensitive channels ,business ,Neuroscience - Abstract
Remote, precisely controlled activation of the brain is a fundamental challenge in the development of brain-machine interfaces providing feasible rehabilitation strategies for neurological disorders. Low-frequency ultrasound stimulation can be used to modulate neuronal activity deep in the brain1–7, but this approach lacks spatial resolution and cellular selectivity and loads the brain with high levels of acoustic energy. The combination of the expression of ultrasound-sensitive proteins with ultrasound stimulation (‘sonogenetic stimulation’) can provide cellular selectivity and higher sensitivity, but such strategies have been subject to severe limitations in terms of spatiotemporal resolution in vivo8–10, precluding their use for real-life applications. We used the expression of large-conductance mechanosensitive ion channels (MscL) with high-frequency ultrasonic stimulation for a duration of milliseconds to activate neurons selectively at a relatively high spatiotemporal resolution in the rat retina ex vivo and the primary visual cortex of rodents in vivo. This spatiotemporal resolution was achieved at low energy levels associated with negligible tissue heating and far below those leading to complications in ultrasound neuromodulation6,11. We showed, in an associative learning test, that sonogenetic stimulation of the visual cortex generated light perception. Our findings demonstrate that sonogenetic stimulation is compatible with millisecond pattern presentation for visual restoration at the cortical level. They represent a step towards the precise transfer of information over large distances to the cortical and subcortical regions of the brain via an approach less invasive than that associated with current brain-machine interfaces and with a wide range of applications in neurological disorders.
- Published
- 2021
21. Numb reduces Tau levels and prevents neurodegeneration in mouse models of tauopathy in an isoform-specific manner
- Author
-
Lacomme M, Hales Sc, Michel Cayouette, Melissa Desrosiers, Cai J, Jolicoeur C, Deniz Dalkara, Bois T, and Stevanovic K
- Subjects
Gene isoform ,animal structures ,biology ,Chemistry ,fungi ,Tau protein ,Neurodegeneration ,Signal transducing adaptor protein ,medicine.disease ,Retinal ganglion ,Cell biology ,embryonic structures ,medicine ,NUMB ,biology.protein ,Tauopathy ,hormones, hormone substitutes, and hormone antagonists ,Intracellular - Abstract
Accumulation of the microtubule-associated protein Tau is linked to neuronal cell death in tauopathies, but how exactly intraneuronal Tau levels are regulated in health and disease remains unclear. Here we identify the trafficking adaptor protein Numb as an essential regulator of Tau homeostasis. Conditional inactivation of Numb in retinal ganglion cells (RGCs) increases monomeric and oligomeric Tau levels, leading to axonal blebbing followed by neuronal cell loss in aged mice. Moreover, in a mouse model of tauopathy, inactivation of Numb in RGCs and spinal motoneurons accelerates neurodegeneration, and leads to precocious hindlimb paralysis. Conversely, overexpression of the long isoform of Numb (Numb-72), but not other isoforms, decreases intracellular Tau levels by promoting the extracellular release of monomeric Tau, and AAV-mediated delivery of Numb-72 in RGCs in vivo prevents neurodegeneration in two different mouse models of tauopathy. Taken together, these results uncover Numb as a modulator of intracellular Tau levels and identify Numb-72 as a novel therapeutic factor for tauopathies.
- Published
- 2021
22. Systemic and local immune response to intraocular AAV vector administration in non-human primates
- Author
-
Duohao Ren, Céline Nouvel-Jaillard, Elena Brazhnikova, Stéphane Bertin, Divya Ail, Deniz Dalkara, and Sylvain Fisson
- Subjects
biology ,business.industry ,viruses ,Immunogenicity ,Genetic enhancement ,Transgene ,Inflammation ,Virus ,Immune system ,Immunology ,medicine ,biology.protein ,Vector (molecular biology) ,medicine.symptom ,Antibody ,business - Abstract
The positive clinical outcomes in adeno-associated virus (AAV)-mediated retinal gene therapy have often been attributed to the low immunogenicity of AAVs along with the immune-privilege of the eye. However, several recent preclinical studies and clinical trials have shown potential for inflammatory responses to AAV mediated gene therapy. Our current understanding of the factors contributing to intraocular inflammation such as the existence of serum antibodies against AAVs prior to injection and their contribution to increases in antibody levels post-injection is incomplete. The parameters that regulate the generation of new antibodies in response to the AAV capsid or transgene post-injection after intraocular administration are also insufficiently described. In this study we carried out a retrospective analysis of the pre-existing serum antibodies in correlation with changes in antibody levels after intraocular injections of AAV in non-human primates (NHPs). We analyzed NHP serums for the presence of both Binding Antibodies (BABs), as well as a subset of these called Neutralizing Antibodies (NABs) that impede AAV transduction upon binding. We observed significantly higher pre-existing serum BABs against AAV8 compared to other serotypes. We observed a dose-dependent increase in both BABs and NABs in the serums collected post-injection, irrespective of the serotype or the mode of injection. Lastly, we were able to demonstrate a co-relation between the serum BAB levels with clinical grading of inflammation and levels of transgene expression.
- Published
- 2021
23. Outer retinal transduction by AAV2-7m8 following intravitreal injection in a sheep model of CNGA3 achromatopsia
- Author
-
A. Rosov, Deniz Dalkara, H. Honig, Ron Ofri, Esther Yamin, Eyal Banin, Edward Averbukh, Maya Ross, Melissa Desrosiers, E. Gootwine, Alexey Obolensky, Raaya Ezra-Elia, and H. Dvir
- Subjects
Achromatopsia ,genetic structures ,Transgene ,Genetic Vectors ,Cyclic Nucleotide-Gated Cation Channels ,Endogeny ,Color Vision Defects ,Biology ,Retina ,Transduction (genetics) ,chemistry.chemical_compound ,Transduction, Genetic ,Genetics ,medicine ,Animals ,Humans ,Vector (molecular biology) ,Molecular Biology ,Messenger RNA ,Sheep ,Retinal ,Genetic Therapy ,Dependovirus ,medicine.disease ,Cell biology ,chemistry ,Intravitreal Injections ,Molecular Medicine ,Photopic vision - Abstract
Sheep carrying a mutated CNGA3 gene exhibit diminished cone function and provide a naturally occurring large animal model of achromatopsia. Subretinal injection of a vector carrying the CNGA3 transgene resulted in long-term recovery of cone function and photopic vision in these sheep. Research is underway to develop efficacious vectors that would enable safer transgene delivery, while avoiding potential drawbacks of subretinal injections. The current study evaluated two modified vectors, adeno-associated virus 2-7m8 (AAV2-7m8) and AAV9-7m8. Intravitreal injection of AAV2-7m8 carrying enhanced green fluorescent protein under a cone-specific promoter resulted in moderate photoreceptor transduction in wild-type sheep, whereas peripheral subretinal delivery of AAV9-7m8 resulted in the radial spread of the vector beyond the point of deposition. Intravitreal injection of AAV2-7m8 carrying human CNGA3 in mutant sheep resulted in mild photoreceptor transduction, but did not lead to the clinical rescue of photopic vision, while day-blind sheep treated with a subretinal injection exhibited functional recovery of photopic vision. Transgene messenger RNA levels in retinas of intravitreally treated eyes amounted to 4-23% of the endogenous CNGA3 levels, indicating that expression levels >23% are needed to achieve clinical rescue. Overall, our results indicate intravitreal injections of AAV2.7m8 transduce ovine photoreceptors, but not with sufficient efficacy to achieve clinical rescue in CNGA3 mutant sheep.
- Published
- 2021
24. Restoration of mGluR6 Localization Following AAV-Mediated Delivery in a Mouse Model of Congenital Stationary Night Blindness
- Author
-
Juliette, Varin, Nassima, Bouzidi, Miguel Miranda De Sousa, Dias, Thomas, Pugliese, Christelle, Michiels, Camille, Robert, Melissa, Desrosiers, José-Alain, Sahel, Isabelle, Audo, Deniz, Dalkara, and Christina, Zeitz
- Subjects
bipolar cells ,Retinal Bipolar Cells ,mGluR6 ,Genetic Vectors ,Gene Transfer Techniques ,Eye Diseases, Hereditary ,Genetic Diseases, X-Linked ,Dependovirus ,Receptors, Metabotropic Glutamate ,Transfection ,gene therapy ,Retina ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Night Blindness ,Intravitreal Injections ,Electroretinography ,Myopia ,Animals ,sense organs ,Visual Neuroscience ,Promoter Regions, Genetic ,CSNB - Abstract
Purpose Complete congenital stationary night blindness (cCSNB) is an incurable inherited retinal disorder characterized by an ON-bipolar cell (ON-BC) defect. GRM6 mutations are the third most prevalent cause of cCSNB. The Grm6−/− mouse model mimics the human phenotype, showing no b-wave in the electroretinogram (ERG) and a loss of mGluR6 and other proteins of the same cascade at the outer plexiform layer (OPL). Our aim was to restore protein localization and function in Grm6−/− adult mice targeting specifically ON-BCs or the whole retina. Methods Adeno-associated virus-encoding Grm6 under two different promoters (GRM6-Grm6 and CAG-Grm6) were injected intravitreally in P15 Grm6−/− mice. ERG recordings at 2 and 4 months were performed in Grm6+/+, untreated and treated Grm6−/− mice. Similarly, immunolocalization studies were performed on retinal slices before or after treatment using antibodies against mGluR6, TRPM1, GPR179, RGS7, RGS11, Gβ5, and dystrophin. Results Following treatment, mGluR6 was localized to the dendritic tips of ON-BCs when expressed with either promoter. The relocalization efficiency in mGluR6-transduced retinas at the OPL was 2.5% versus 11% when the GRM6-Grm6 and CAG-Grm6 were used, respectively. Albeit no functional rescue was seen in ERGs, relocalization of TRPM1, GPR179, and Gβ5 was also noted using both constructs. The restoration of the localization of RGS7, RGS11, and dystrophin was more obvious in retinas treated with GRM6-Grm6 than in retinas treated with CAG-Grm6. Conclusions Our findings show the potential of treating cCSNB with GRM6 mutations; however, it appears that the transduction rate must be improved to restore visual function.
- Published
- 2021
25. Early and late stage gene therapy interventions for inherited retinal degenerations
- Author
-
José-Alain Sahel, Deniz Dalkara, Catherine Botto, Müge Defne Tekinsoy, Juliette Pulman, Marco Rucli, Institut de la Vision, Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM), University of Pittsburgh School of Medicine, Pennsylvania Commonwealth System of Higher Education (PCSHE), Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts (CHNO), Hôpital de la Fondation Ophtalmologique Adolphe de Rothschild [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), and HAL-SU, Gestionnaire
- Subjects
0301 basic medicine ,Retinal degeneration ,Genetic enhancement ,Disease ,Bioinformatics ,Retina ,03 medical and health sciences ,chemistry.chemical_compound ,Therapeutic approach ,Macular Degeneration ,0302 clinical medicine ,Gene therapy ,Retinitis pigmentosa ,medicine ,Humans ,CRISPR/Cas9 ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,business.industry ,Retinal Degeneration ,Retinal ,AAV ,Genetic Therapy ,Macular degeneration ,medicine.disease ,Sensory Systems ,Neuroprotection ,3. Good health ,Optogenetics ,Ophthalmology ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030221 ophthalmology & optometry ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,Retinitis Pigmentosa - Abstract
International audience; Inherited and age-related retinal degeneration is the hallmark of a large group of heterogeneous diseases and is the main cause of untreatable blindness today. Genetic factors play a major pathogenic role in retinal degenerations for both monogenic diseases (such as retinitis pigmentosa) and complex diseases with established genetic risk factors (such as age-related macular degeneration). Progress in genotyping techniques and back of the eye imaging are completing our understanding of these diseases and their manifestations in patient populations suffering from retinal degenerations. It is clear that whatever the genetic cause, the majority of vision loss in retinal diseases results from the loss of photoreceptor function. The timing and circumstances surrounding the loss of photoreceptor function determine the adequate therapeutic approach to use for each patient. Among such approaches, gene therapy is rapidly becoming a therapeutic reality applicable in the clinic. This massive move from laboratory work towards clinical application has been propelled by the advances in our understanding of disease genetics and mechanisms, gene delivery vectors, gene editing systems, and compensatory strategies for loss of photoreceptor function. Here, we provide an overview of existing modalities of retinal gene therapy and their relevance based on the needs of patient populations suffering from inherited retinal degenerations.
- Published
- 2021
26. AAV-Mediated Gene Delivery to Foveal Cones
- Author
-
Stéphane, Bertin, Elena, Brazhnikova, Céline, Jaillard, José-Alain, Sahel, and Deniz, Dalkara
- Subjects
Male ,Genetic Vectors ,Retinal Cone Photoreceptor Cells ,Animals ,Female ,Genetic Therapy ,Dependovirus ,Retina ,Tomography, Optical Coherence - Abstract
Adeno-associated virus (AAV) has emerged as the vector of choice for delivering genes to the mammalian retina. From the first gene therapy to receive FDA approval for the inherited retinal disease (Luxturna™) to more recent clinical trials using microbial opsins to regain light sensitivity, therapeutic transgenes rely on AAV vectors for safe and efficient gene delivery to retinal cells. Such vectors are administered to the retina via subretinal (SR) injection or intravitreal (IVT) injection routes depending on the targeted retinal cell type. An attractive target for gene therapy is the fovea, bearing the highest concentration of cone cells responsible for our high acuity daylight vision. However, previous clinical trials and large animal studies reported that SR administration of vector under the cone-exclusive fovea disrupts its fine structure and might impair visual acuity. Due to its technical difficulty and potential risks, alternatives to vector injection under this delicate region have been investigated by using novel AAV capsid variants identified via rational design or directed evolution. We recently established new vector-promoter combinations to overcome the limitations associated with AAV-mediated cone transduction in the fovea. Our methods provide efficient foveal cone transduction without detaching this delicate region and rely on the use of engineered AAVs and optimal promoters compatible with optogenetic vision restoration. Here we describe in detail our AAV vectors, methods for intravitreal and subretinal injections as well as pre- and postoperative procedures as performed in cynomolgus macaques.
- Published
- 2020
27. Optogenetic therapy: High spatiotemporal resolution and pattern recognition compatible with vision restoration in non-human primates
- Author
-
José-Alain Sahel, Fabrice Arcizet, Philippe Hantraye, Serge Picaud, Gregory Gauvain, Claire-Maëlle Fovet, Jens Duebel, Pierre Pouget, Mina A. Khoei, Deniz Dalkara, Joël Chavas, Melissa Desrosiers, Elena Brazhnikova, Ryad Benosman, Didier Pruneau, Olivier Marre, Antoine Chaffiol, Joanna Demilly, Himanshu Akolkar, Anne Douar, Valérie Forster, Romain Caplette, Céline Jaillard, and Stéphane Bertin
- Subjects
Retina ,Opsin ,Visual acuity ,genetic structures ,Stimulus (physiology) ,Optogenetics ,Biology ,medicine.disease ,Retinal ganglion ,eye diseases ,medicine.anatomical_structure ,Perifovea ,Retinitis pigmentosa ,medicine ,sense organs ,medicine.symptom ,Neuroscience - Abstract
Restoring vision using optogenetics is an ideal medical application because the eye offers a direct window to access and stimulate the pathological area: the retina. Optogenetic therapy could be applied to diseases with photoreceptor degeneration such as retinitis pigmentosa. Here, we select the specific optogenetic construct that is now used in the clinical trial and assess the opsin functional efficacy on non-human primate’s retinal ganglion cells (RGCs).We chose the microbial opsin ChrimsonR and showed that the vector AAV2.7m8 produced greater transfection in RGCs compared to AAV2, and that ChrimsonR attached to tdTomato (ChR-tdT) is more efficiently expressed than ChrimsonR. The 600 nm light activates the RGCs transfected with the vector AAV2.7m8-ChR-tdT from an irradiance of 1015 photons.cm-2.s-1. Vector doses of 5.1010 and 5.1011 vg/eye transfect up to 7000 RGCs/mm2 in the perifovea, with no significant immune reaction. Furthermore, using a multielectrode array we recorded RGCs responses starting from 1ms stimulus duration. Using the recorded activity we were able to decode stimulus information and estimate a theoretical visual acuity of 20/249, above legal blindness. Altogether, our results pave the way for the ongoing clinical trial with the AAV2.7m8-ChrimsonR-tdT vector for vision restoration in patients affected by retinitis pigmentosa.One Sentence SummaryWe select here the vector and genetic construct best suited to provide vision restoration in patients suffering from retinopathies, we demonstrate temporal resolution compatible with high dynamic visual scenes and a visual acuity above legal blindness.
- Published
- 2020
28. Optogenetic therapy: high spatiotemporal resolution and pattern discrimination compatible with vision restoration in non-human primates
- Author
-
Céline Jaillard, Gregory Gauvain, Serge Picaud, Olivier Marre, Antoine Chaffiol, Fabrice Arcizet, Stéphane Bertin, Pierre Pouget, Jens Duebel, Didier Pruneau, Anne Douar, Joël Chavas, Romain Caplette, Joanna Demilly, Elena Brazhnikova, Philippe Hantraye, Melissa Desrosiers, Valérie Forster, Claire-Maëlle Fovet, Mina A. Khoei, José-Alain Sahel, Himanshu Akolkar, Deniz Dalkara, Ryad Benosman, Institut de la Vision, Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM), University of Pittsburgh (PITT), Pennsylvania Commonwealth System of Higher Education (PCSHE), Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts (CHNO), Service MIRCEN (MIRCEN), Université Paris-Saclay-Institut de Biologie François JACOB (JACOB), Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Laboratoire d'Imagerie Biomédicale (LIB), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Commissariat à l'énergie atomique et aux énergies alternatives (CEA), Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Gestionnaire, Hal Sorbonne Université, Centre d'investigation clinique Quinze-Vingts [CHNO] (CIC1423 - CIC QUINZE-VINGTS), Institut Hospitalo-Universitaire FOReSIGHT, Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts (CHNO)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts (CHNO)-Sorbonne Université (SU), Institut d'Imagerie BioMédicale (I2BM), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay, Institut du Cerveau = Paris Brain Institute (ICM), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), GenSight Biologics, Institut Hospitalo-Universitaire FOReSIGHT (IHU FOReSIGHT), Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie François JACOB (JACOB), and ANR-18-IAHU-0001,FOReSIGHT,Enabling Vision Restoration(2018)
- Subjects
0301 basic medicine ,Male ,Primates ,Opsin ,Visual acuity ,genetic structures ,QH301-705.5 ,[SDV]Life Sciences [q-bio] ,viruses ,Medicine (miscellaneous) ,Optogenetics ,Stimulus (physiology) ,Retinal ganglion ,General Biochemistry, Genetics and Molecular Biology ,Article ,Retina ,03 medical and health sciences ,0302 clinical medicine ,Retinitis pigmentosa ,medicine ,Animals ,Humans ,Biology (General) ,Vision, Ocular ,business.industry ,Therapies, Investigational ,Retinal Degeneration ,Macular degeneration ,Translational research ,medicine.disease ,eye diseases ,[SDV] Life Sciences [q-bio] ,Macaca fascicularis ,030104 developmental biology ,medicine.anatomical_structure ,Equipment and Supplies ,Pattern Recognition, Visual ,Female ,sense organs ,medicine.symptom ,General Agricultural and Biological Sciences ,business ,Neuroscience ,030217 neurology & neurosurgery ,Photic Stimulation - Abstract
Vision restoration is an ideal medical application for optogenetics, because the eye provides direct optical access to the retina for stimulation. Optogenetic therapy could be used for diseases involving photoreceptor degeneration, such as retinitis pigmentosa or age-related macular degeneration. We describe here the selection, in non-human primates, of a specific optogenetic construct currently tested in a clinical trial. We used the microbial opsin ChrimsonR, and showed that the AAV2.7m8 vector had a higher transfection efficiency than AAV2 in retinal ganglion cells (RGCs) and that ChrimsonR fused to tdTomato (ChR-tdT) was expressed more efficiently than ChrimsonR. Light at 600 nm activated RGCs transfected with AAV2.7m8 ChR-tdT, from an irradiance of 1015 photons.cm−2.s−1. Vector doses of 5 × 1010 and 5 × 1011 vg/eye transfected up to 7000 RGCs/mm2 in the perifovea, with no significant immune reaction. We recorded RGC responses from a stimulus duration of 1 ms upwards. When using the recorded activity to decode stimulus information, we obtained an estimated visual acuity of 20/249, above the level of legal blindness (20/400). These results lay the groundwork for the ongoing clinical trial with the AAV2.7m8 - ChR-tdT vector for vision restoration in patients with retinitis pigmentosa., Gauvain et al demonstrate that optogenetic therapy using the AAV2.7m8- ChR-tdT construct can partially restore vision in non-human primates to levels above those considered legally-blind. This study enables the identification of the most suitable construct for ongoing clinical trials attempting vision restoration in patients with retinitis pigmentosa.
- Published
- 2020
29. Rescue of Defective Electroretinographic Responses in Dp71-Null Mice With AAV-Mediated Reexpression of Dp71
- Author
-
Dora Fix Ventura, Deniz Dalkara, Anneka Joachimsthaler, Jan Kremers, Lucile Vignaud, Hanen Khabou, Ophélie Vacca, Alvaro Rendon, Mirella Telles Salgueiro Barboni, Cyrille Vaillend, Andre Liber, Michel Roux, Xavier Guillonneau, Universidade de São Paulo (USP), Institut des Neurosciences Paris-Saclay (NeuroPSI), Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Institut de la Vision, Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Universidade de São Paulo = University of São Paulo (USP), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), and PERIGNON, Alain
- Subjects
0301 basic medicine ,Male ,retina ,genetic structures ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,electroretinogram ,chemistry.chemical_compound ,0302 clinical medicine ,Müller glial cells ,Mice, Knockout ,medicine.diagnostic_test ,[SDV.NEU.SC]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Cognitive Sciences ,General Medicine ,Dependovirus ,gene therapy ,Aquaporin 4 ,medicine.anatomical_structure ,[SDV.MHEP.OS] Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Female ,medicine.symptom ,Dystrophin ,Erg ,Photopic vision ,Genotype ,Ependymoglial Cells ,Genetic Vectors ,Photopsia ,Dark Adaptation ,Biology ,dystrophin ,Andrology ,03 medical and health sciences ,Retinal Diseases ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,medicine ,Electroretinography ,Animals ,[SDV.MHEP.OS]Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,Retina ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Retinal ,Genetic Therapy ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,biology.protein ,sense organs ,Visual Neuroscience ,[SDV.NEU.SC] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Cognitive Sciences ,030217 neurology & neurosurgery - Abstract
International audience; Purpose: To study the potential effect of a gene therapy, designed to rescue the expression of dystrophin Dp71 in the retinas of Dp71-null mice, on retinal physiology.Methods: We recorded electroretinograms (ERGs) in Dp71-null and wild-type littermate mice. In dark-adapted eyes, responses to flashes of several strengths were measured. In addition, flash responses on a 25-candela/square meters background were measured. On- and Off-mediated responses to sawtooth stimuli and responses to photopic sine-wave modulation (3-30 Hz) were also recorded. After establishing the ERG phenotype, the ShH10-GFP adeno-associated virus (AAV), which has been previously shown to target specifically Müller glial cells (MGCs), was delivered intravitreously with or without (sham therapy) the Dp71 coding sequence under control of a CBA promoter. ERG recordings were repeated three months after treatment. Real-time quantitative PCR and Western blotting analyses were performed in order to quantify Dp71 expression in the retinas.Results: Dp71-null mice displayed reduced b-waves in dark- and light-adapted flash ERGs and smaller response amplitudes to photopic rapid-on sawtooth modulation and to sine-wave stimuli. Three months after intravitreal injections of the ShH10-GFP-2A-Dp71 AAV vector, ERG responses were completely recovered in treated eyes of Dp71-null mice. The functional rescue was associated with an overexpression of Dp71 in treated retinas.Conclusions: The present results show successful functional recovery accompanying the reexpression of Dp71. In addition, this experimental model sheds light on MGCs influencing ERG components, since previous reports showed that aquaporin 4 and Kir4.1 channels were mislocated in MGCs of Dp71-null mice, while their distribution could be normalized following intravitreal delivery of the same ShH10-GFP-2A-Dp71 vector.
- Published
- 2020
30. AAV-Mediated Gene Delivery to Foveal Cones
- Author
-
Céline Jaillard, Deniz Dalkara, José-Alain Sahel, Elena Brazhnikova, and Stéphane Bertin
- Subjects
0301 basic medicine ,Retina ,Opsin ,Visual acuity ,genetic structures ,business.industry ,Genetic enhancement ,Retinal ,Gene delivery ,3. Good health ,03 medical and health sciences ,Transduction (genetics) ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,Foveal ,030221 ophthalmology & optometry ,Medicine ,sense organs ,medicine.symptom ,business ,Neuroscience - Abstract
Adeno-associated virus (AAV) has emerged as the vector of choice for delivering genes to the mammalian retina. From the first gene therapy to receive FDA approval for the inherited retinal disease (Luxturna™) to more recent clinical trials using microbial opsins to regain light sensitivity, therapeutic transgenes rely on AAV vectors for safe and efficient gene delivery to retinal cells. Such vectors are administered to the retina via subretinal (SR) injection or intravitreal (IVT) injection routes depending on the targeted retinal cell type. An attractive target for gene therapy is the fovea, bearing the highest concentration of cone cells responsible for our high acuity daylight vision. However, previous clinical trials and large animal studies reported that SR administration of vector under the cone-exclusive fovea disrupts its fine structure and might impair visual acuity. Due to its technical difficulty and potential risks, alternatives to vector injection under this delicate region have been investigated by using novel AAV capsid variants identified via rational design or directed evolution. We recently established new vector-promoter combinations to overcome the limitations associated with AAV-mediated cone transduction in the fovea. Our methods provide efficient foveal cone transduction without detaching this delicate region and rely on the use of engineered AAVs and optimal promoters compatible with optogenetic vision restoration. Here we describe in detail our AAV vectors, methods for intravitreal and subretinal injections as well as pre- and postoperative procedures as performed in cynomolgus macaques.
- Published
- 2020
31. Towards optogenetic vision restoration with high resolution
- Author
-
Ulisse Ferrari, Stéphane Deny, Abhishek Sengupta, Romain Caplette, Francesco Trapani, José-Alain Sahel, Deniz Dalkara, Serge Picaud, Jens Duebel, Olivier Marre
- Published
- 2020
- Full Text
- View/download PDF
32. Sphingosine 1-Phosphate Receptor 1 Modulates CNTF-Induced Axonal Growth and Neuroprotection in the Mouse Visual System
- Author
-
Deniz Dalkara, Sandrine Joly, Vincent Pernet, Université Laval [Québec] (ULaval), Institut de la Vision, Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and HAL UPMC, Gestionnaire
- Subjects
Retinal Ganglion Cells ,STAT3 Transcription Factor ,0301 basic medicine ,Article Subject ,Cell Survival ,Down-Regulation ,Ciliary neurotrophic factor ,Neuroprotection ,lcsh:RC321-571 ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,Ciliary Neurotrophic Factor ,Phosphorylation ,STAT3 ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,S1PR1 ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,Sphingosine ,biology ,Axons ,Nerve Regeneration ,Receptors, Lysosphingolipid ,Neuroprotective Agents ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,chemistry ,Retinal ganglion cell ,biology.protein ,Optic nerve ,sense organs ,Neurology (clinical) ,Signal transduction ,Neuroscience ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,030217 neurology & neurosurgery ,Signal Transduction ,Research Article - Abstract
The lack of axonal regeneration and neuronal cell death causes permanent neurological deficits in the injured CNS. Using the classical CNS injury model of optic nerve crush in mice,ciliary neurotrophic factor(CNTF) was found to stimulate retinal ganglion cell (RGC) survival and axonal growth, but in an incomplete fashion. The elucidation of molecular mechanisms impairing CNTF-induced axonal regeneration is paramount to promote visual recovery. In the present study, we sought to evaluate the contribution ofsphingosine 1-phosphate receptor 1(S1PR1) to the neuroprotective and regenerative effects of CNTF. The transduction of retinal cells with adeno-associated viruses (AAV) allowed to activate CNTF/signal transducer and activator of transcription 3(Stat3) signaling and to modulate S1PR1 expression in RGCs. Our results showed that CNTF/Stat3 prevented injury-induced S1PR1 downregulation. Silencing S1PR1 in RGCs significantly enhanced CNTF-induced axonal growth in the injured optic nerve. In contrast, RGC survival was markedly decreased when S1PR1 was repressed with viral vectors. The level of phosphorylated Stat3 (P-Stat3), an intracellular mediator of CNTF, did not fluctuate after S1PR1 inhibition and CNTF stimulation. Collectively, these results suggest that S1PR1 acts as a major regulator of retinal neuron survival and restricts the RGC growth response induced by CNTF.
- Published
- 2017
33. In vivo directed evolution of AAV in the primate retina
- Author
-
Cécile Fortuny, Timothy P Day, Leah C. Byrne, David V. Schaffer, Meike Visel, William H. Merigan, John G. Flannery, and Deniz Dalkara
- Subjects
0303 health sciences ,Retina ,viruses ,Computational biology ,Gene delivery ,Biology ,Directed evolution ,Deep sequencing ,Viral vector ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,In vivo ,030220 oncology & carcinogenesis ,medicine ,Gene ,Tropism ,030304 developmental biology - Abstract
Efficient AAV-mediated gene delivery remains a significant obstacle to effective retinal gene therapies. Here, we apply directed evolution – guided by deep sequencing and followed by direct in vivo secondary selection of high-performing vectors with a GFP-barcoded library – to create AAV viral capsids with new capabilities to deliver genes to the outer retina in primates. A replication incompetent library, produced via providing rep in trans, was created to mitigate risk of AAV propagation. Six rounds of in vivo selection with this library in primates – involving intravitreal library administration, recovery of genomes from outer retina, and extensive next generation sequencing of each round – resulted in vectors with redirected tropism to the outer retina and increased gene delivery efficiency to retinal cells. These new viral vectors expand the toolbox of vectors available for primate retina, and may enable less invasive delivery of therapeutic genes to patients, potentially offering retina-wide infection at a similar dosage to vectors currently in clinical use.
- Published
- 2019
34. In vivo-directed evolution of adeno-associated virus in the primate retina
- Author
-
Meike Visel, William H. Merigan, Jennifer Strazzeri, Timothy P Day, Cécile Fortuny, David V. Schaffer, John G. Flannery, Deniz Dalkara, and Leah C. Byrne
- Subjects
0301 basic medicine ,Genetic enhancement ,viruses ,Genetic Vectors ,Computational biology ,Biology ,Gene delivery ,medicine.disease_cause ,Deep sequencing ,Retina ,Viral vector ,03 medical and health sciences ,0302 clinical medicine ,Transduction, Genetic ,medicine ,Animals ,Humans ,Gene ,Adeno-associated virus ,Tropism ,General Medicine ,Haplorhini ,Dependovirus ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,030220 oncology & carcinogenesis ,Directed Molecular Evolution ,Research Article - Abstract
Efficient adeno-associated virus-mediated (AAV-mediated) gene delivery remains a significant obstacle to effective retinal gene therapies. Here, we apply directed evolution - guided by deep sequencing and followed by direct in vivo secondary selection of high-performing vectors with a GFP-barcoded library - to create AAV viral capsids with the capability to deliver genes to the outer retina in primates. A replication-incompetent library, produced via providing rep in trans, was created to mitigate risk of AAV propagation. Six rounds of in vivo selection with this library in primates - involving intravitreal library administration, recovery of genomes from outer retina, and extensive next-generation sequencing of each round - resulted in vectors with redirected tropism to the outer retina and increased gene delivery efficiency to retinal cells. These viral vectors expand the toolbox of vectors available for primate retina, and they may enable less invasive delivery of therapeutic genes to patients, potentially offering retina-wide infection at a similar dosage to vectors currently in clinical use.
- Published
- 2019
35. Non-viral delivery of chemically modified mRNA to the retina: Subretinal versus intravitreal administration
- Author
-
Stefaan C. De Smedt, Deniz Dalkara, Sinem Yilmaz Ozcan, Lieve Moons, Christian Vanhove, Heleen Dewitte, Katrien Remaut, Joke Devoldere, Lies De Groef, and Karen Peynshaert
- Subjects
Green Fluorescent Proteins ,Pharmaceutical Science ,Gene Expression ,02 engineering and technology ,Gene delivery ,Retina ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,In vivo ,medicine ,Animals ,Humans ,RNA, Messenger ,Transgenes ,Luciferases ,030304 developmental biology ,0303 health sciences ,Reporter gene ,Drug Carriers ,Chemistry ,Gene Transfer Techniques ,Retinal ,Inner limiting membrane ,Epithelial Cells ,Intravitreal administration ,DNA ,021001 nanoscience & nanotechnology ,Lipids ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cattle ,Injections, Intraocular ,0210 nano-technology ,Neuroglia ,Ex vivo ,Plasmids - Abstract
mRNA therapeutics have recently experienced a new wave of interest, mainly due to the discovery that chemical modifications to mRNA's molecular structure could drastically reduce its inherent immunogenicity and perceived instability. On this basis, we aimed to explore the potential of chemically stabilized mRNA for ocular applications. More specifically, we investigated the behavior of mRNA-loaded lipid-based carriers in human retinal cells (in vitro), in bovine retinal explants (ex vivo) and in mouse retinas (in vivo). We demonstrate a clear superiority of mRNA over pDNA to induce protein expression in different retinal cell types, which was further enhanced by chemical modification of the mRNA, providing up to ~1800-fold higher reporter gene expression compared to pDNA. Moreover, transgene expression could be detected for at least 20 days after a single administration of chemically modified mRNA in vitro. We furthermore determined the localization and extent of mRNA expression depending on the administration route. After subretinal (SR) administration, mRNA expression was observed in vivo and ex vivo. By contrast, intravitreal (IVT) administration resulted in limited expression in vivo. Using ex vivo bovine explants with an intact vitreoretinal (VR) interface we could attribute this to the inner limiting membrane (ILM), which presents a large barrier for non-viral delivery of mRNA, trapping mRNA complexes at the vitreal side. When the vitreous was removed, which compromises the ILM, mRNA expression was apparent and seemed to colocalize with Muller cells or photoreceptors after respectively IVT or SR administration. Taken together, this study represents a first step towards mRNA-mediated therapy for retinal diseases.
- Published
- 2019
36. All-optical interrogation of a direction selective retinal circuit by holographic wave front shaping
- Author
-
Deniz Dalkara, Ulisse Ferrari, Emiliano Ronzitti, Emiliani, Serge Picaud, Eirini Papagiakoumou, Zampini, Trapani F, Hanen Khabou, Giulia Lb Spampinato, and Olivier Marre
- Subjects
Wavefront ,Physics ,Retina ,business.industry ,Holography ,Sensory system ,Retinal ,Direction selective ,law.invention ,chemistry.chemical_compound ,medicine.anatomical_structure ,Optics ,chemistry ,law ,medicine ,sense organs ,business ,Ganglion cell layer ,Electronic circuit - Abstract
Direction selective (DS) ganglion cells (GC) in the retina maintain their tuning across a broad range of light levels. Yet very different circuits can shape their responses from bright to dim light, and their respective contributions are difficult to tease apart. In particular, the contribution of the rod bipolar cell (RBC) primary pathway, a key player in dim light, is unclear. To understand its contribution to DSGC response, we designed an all-optical approach allowing precise manipulation of single retinal neurons. Our system activates single cells in the bipolar cell (BC) layer by two-photon (2P) temporally focused holographic illumination, while recording the activity in the ganglion cell layer by 2P Ca2 imaging. By doing so, we demonstrate that RBCs provide an asymmetric input to DSGCs, suggesting they contribute to their direction selectivity. Our results suggest that every circuit providing an input to direction selective cells can generate direction selectivity by itself. This hints at a general principle to achieve robust selectivity in sensory areas.
- Published
- 2019
37. Restoration of visual function by transplantation of optogenetically engineered photoreceptors
- Author
-
Marcela Garita-Hernandez, Maruša Lampič, Antoine Chaffiol, Laure Guibbal, Fiona Routet, Tiago Santos-Ferreira, Sylvia Gasparini, Oliver Borsch, Giuliana Gagliardi, Sacha Reichman, Serge Picaud, José-Alain Sahel, Olivier Goureau, Marius Ader, Deniz Dalkara, Jens Duebel
- Published
- 2019
- Full Text
- View/download PDF
38. Gene and Cell Therapy for Inherited Retinal Dystrophies
- Author
-
Deniz Dalkara, Olivier Goureau, and Marcela Garita-Hernandez
- Subjects
0301 basic medicine ,Genetics ,Retinal pigment epithelium ,Genetic enhancement ,Cellular differentiation ,Biology ,Embryonic stem cell ,Viral vector ,Cell therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Induced pluripotent stem cell ,Cell potency ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Gene therapy and cell therapy are intersecting fields of biomedical research with the common aim of repairing the direct causes or consequences of genetic diseases by using genes and cells, respectively. Gene therapy is broadly defined as a set of strategies that modify the expression of an individual's genes for therapeutic benefit. It requires the administration of a specific deoxyribonucleic acid (DNA) (or ribonucleic acid (RNA)) into the patient's cells. Cell therapy on the other hand requires administration of a population of intact live cells to a patient whose cells have been lost due to injury or disease. These emerging strategies can also be applied in acquired diseases in order to re-establish equilibrium and halt disease progression. In some cases, gene and cell therapy may be combined in order to obtain a therapeutic effect. In addition to becoming emerging therapeutics in the clinic, these two fields of research continually generate tools, concepts and techniques for elucidating biological questions across multiple disciplines. Key Concepts The retina lines the back of the eye and comprises six different types of neuronal cells and their underlying retinal pigment epithelium (RPE). Photoreceptors are the light-sensitive first-order neurons of the retina which capture the light stimulus and transform it into an electric signal. Human iPSC have the same features of hESC in terms of their differentiation capacity. Gene therapy is the use of genetic material as a drug to treat a chronic condition. Keywords: cell therapy; gene therapy; human-induced pluripotent stem cell; human embryonic stem cell; stem-cell derived RPE; stem cell-derived retinal progenitor; photoreceptor; viral vectors; genetics; retina
- Published
- 2016
39. Insight into the mechanisms of enhanced retinal transduction by the engineered AAV2 capsid variant -7m8
- Author
-
Deniz Dalkara, Alexis-Pierre Bemelmans, Hanen Khabou, José-Alain Sahel, Stéphane Fouquet, Céline Winckler, Gwenaëlle Auregan, and Melissa Desrosiers
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Retina ,viruses ,Genetic enhancement ,Bioengineering ,Peptide ,Biology ,Gene delivery ,Applied Microbiology and Biotechnology ,Molecular biology ,Cell biology ,Amino acid ,03 medical and health sciences ,Transduction (genetics) ,030104 developmental biology ,medicine.anatomical_structure ,Capsid ,chemistry ,Gene expression ,medicine ,Biotechnology - Abstract
Recently, we described a modified AAV2 vector—AAV2-7m8—having a capsid-displayed peptide insertion of 10 amino acids with enhanced retinal transduction properties. The insertion of the peptide referred to as 7m8 is responsible for high-level gene delivery into deep layers of the retina when virus is delivered into the eye's vitreous. Here, we further characterize AAV2-7m8 mediated gene delivery to neural tissue and investigate the mechanisms by which the inserted peptide provides better transduction away from the injection site. First, in order to understand if the peptide exerts its effect on its own or in conjunction with the neighboring amino acids, we inserted the 7m8 peptide at equivalent positions on three other AAV capsids, AAV5, AAV8, and AAV9, and evaluated its effect on their infectivity. Intravitreal delivery of these peptide insertion vectors revealed that only AAV9 benefited from 7m8 insertion in the context of the retina. We then investigated AAV2-7m8 and AAV9-7m8 properties in the brain, to better evaluate the spread and efficacy of viral transduction in view of the peptide insertion. While 7m8 insertion led to higher intensity gene expression, the spread of gene expression remained unchanged compared to the parental serotypes. Our results indicate that the 7m8 peptide insertion acts by increasing efficacy of cellular entry, with little effect on the spread of viral particles in neural tissue. The effects of peptide insertion are capsid and tissue dependent, highlighting the importance of the microenvironment in gene delivery using AAV.
- Published
- 2016
40. Linking YAP to Müller Glia Quiescence Exit in the Degenerative Retina
- Author
-
Deniz Dalkara, Muriel Perron, Juliette Bitard, Jerome E. Roger, Annaïg Hamon, Diana García-García, Albert Chesneau, Divya Ail, Morgane Locker, Institut des Neurosciences Paris-Saclay (NeuroPSI), Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS), Institut de la Vision, Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d'Etudes et de Recherche Thérapeutique en Ophtalmologie (CERTO), Association RETINA France, Partenaires INRAE-Partenaires INRAE, Université Paris-Sud - Paris 11 (UP11)-Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), PERRON, Muriel, and Gestionnaire, Hal Sorbonne Université
- Subjects
0301 basic medicine ,Transcription, Genetic ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,[SDV]Life Sciences [q-bio] ,Hippo/YAP pathway ,Cell Cycle Proteins ,[SDV.BC.BC]Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,Xenopus Proteins ,Photoreceptor cell ,Xenopus laevis ,0302 clinical medicine ,lcsh:QH301-705.5 ,Retinal regeneration ,Mice, Knockout ,Cell Cycle ,Retinal Degeneration ,Cell biology ,Up-Regulation ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,[SDV.MHEP.OS] Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,Reprogramming ,Muller glia ,Neuroglia ,Photoreceptor Cells, Vertebrate ,Signal Transduction ,Ependymoglial Cells ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Retina ,03 medical and health sciences ,reactive gliosis ,Downregulation and upregulation ,[SDV.BC.BC] Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,medicine ,Animals ,Humans ,EGFR pathway ,[SDV.MHEP.OS]Life Sciences [q-bio]/Human health and pathology/Sensory Organs ,retinal regeneration ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Müller cells ,Hippo signaling pathway ,Epidermal Growth Factor ,Regeneration (biology) ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,YAP-Signaling Proteins ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Biology (General) ,Trans-Activators ,sense organs ,030217 neurology & neurosurgery - Abstract
Summary: Contrasting with fish or amphibian, retinal regeneration from Müller glia is largely limited in mammals. In our quest toward the identification of molecular cues that may boost their stemness potential, we investigated the involvement of the Hippo pathway effector YAP (Yes-associated protein), which is upregulated in Müller cells following retinal injury. Conditional Yap deletion in mouse Müller cells prevents cell-cycle gene upregulation that normally accompanies reactive gliosis upon photoreceptor cell death. We further show that, in Xenopus, a species endowed with efficient regenerative capacity, YAP is required for their injury-dependent proliferative response. In the mouse retina, where Müller cells do not spontaneously proliferate, YAP overactivation is sufficient to induce their reprogramming into highly proliferative cells. Overall, we unravel a pivotal role for YAP in tuning Müller cell proliferative response to injury and highlight a YAP-EGFR (epidermal growth factor receptor) axis by which Müller cells exit their quiescence state, a critical step toward regeneration. : While fish and amphibian Müller cells behave as retinal stem cells upon injury, their regenerative potential is limited in mammals. Hamon et al. show that YAP is required for their cell-cycle re-entry in Xenopus and is sufficient in mouse to awake them from quiescence and trigger their proliferative response. Keywords: Müller cells, reactive gliosis, retinal regeneration, Hippo/YAP pathway, EGFR pathway
- Published
- 2018
41. Neutralizing Antibodies Against Adeno-Associated Virus (AAV): Measurement and Influence on Retinal Gene Delivery
- Author
-
Deniz Dalkara and Melissa Desrosiers
- Subjects
0301 basic medicine ,viruses ,Genetic enhancement ,Genetic Vectors ,Gene delivery ,medicine.disease_cause ,Antibodies, Viral ,Retina ,Viral vector ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Transduction, Genetic ,medicine ,Humans ,Vector (molecular biology) ,Adeno-associated virus ,biology ,business.industry ,Gene Transfer Techniques ,Retinal ,Genetic Therapy ,Dependovirus ,Virology ,Antibodies, Neutralizing ,3. Good health ,Titer ,030104 developmental biology ,HEK293 Cells ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Antibody ,business - Abstract
Adeno-associated viral vectors have become widely used in the clinic for retinal gene therapy. Thanks to AAVs impeccable safety profile and positive functional outcomes in its clinical application, interest in retinal gene therapy has increased exponentially over the past decade. Although early clinical trials have shown there is little influence of neutralizing antibodies on the performance of AAV when vector is administered into the subretinal space, recent findings suggest neutralizing antibodies may play a role when AAV is delivered via the intravitreal route. These findings highlight the importance of microenvironment on gene delivery and stress the need for a versatile assay to screen subjects for the presence of AAV-neutralizing antibodies. Measuring NAb titers against AAV prior and after gene therapy will help us better understand the impact of preexisting immunity on gene transfer, especially when the vector is administered intravitreally.
- Published
- 2017
42. La conception de vecteurs adaptés à la thérapie génique oculaire
- Author
-
Deniz Dalkara and Hanen Khabou
- Subjects
General Medicine ,General Biochemistry, Genetics and Molecular Biology - Abstract
L’utilisation d’un gene comme medicament est l’un des concepts les plus passionnants de la medecine moderne. Les vecteurs viraux ont ete largement utilises a cette fin et ont montre une efficacite therapeutique dans une variete de modeles animaux de degenerescence retinienne. L’extension de ce succes a une application clinique a ete initialement lente, mais une expression a long terme de genes therapeutiques a recemment ete obtenue chez des patients atteints de deficits immunitaires, d’hemophilie B ou de troubles hereditaires de la retine. Ces resultats ont suscite des espoirs pour le traitement de nombreuses autres maladies, et ont ouvert la voie au developpement de nouveaux outils de transfert de genes. Comme nous le verrons ici, les perspectives et les defis de la therapie genique sont dans une large mesure dependants du tissu cible, de la maladie concernee et, surtout, de l’efficacite du transfert genique. L’acheminement du gene vers les cellules cibles depend de vecteurs, qui doivent assurer une expression durable du gene sans engendrer ni toxicite ni reaction immunitaire de la part de l’hote. La conception de tels vecteurs, proposee pour la premiere fois dans les annees 1970, s’est averee etre plus compliquee que prevu, limitant pendant de nombreuses annees le succes de la therapie genique. Les vecteurs doivent donc etre adaptes a chaque scenario ; nous discuterons ici la conception de tels vecteurs pour un transfert de genes vers la retine.
- Published
- 2015
43. Retinoschisin gene therapy in photoreceptors, Müller glia or all retinal cells in the Rs1h−/− mouse
- Author
-
Bilge Esin Ozturk, Cécile Fortuny, Leah C. Byrne, Deniz Dalkara, Trevor Lee, John G. Flannery, Meike Visel, and David V. Schaffer
- Subjects
Aging ,genetic structures ,Genetic enhancement ,Retinoschisis ,Inbred C57BL ,Eye ,Medical and Health Sciences ,Mice ,chemistry.chemical_compound ,Genetics ,medicine.diagnostic_test ,photoreceptors ,Gene Therapy ,Biological Sciences ,Cell biology ,Mutant Strains ,medicine.anatomical_structure ,Molecular Medicine ,Muller glia ,X-linked retinoschisis ,Photoreceptor Cells, Vertebrate ,Biotechnology ,Cell type ,Müller glia ,Green Fluorescent Proteins ,Genetic Vectors ,Biology ,Article ,Retina ,AAV vectors ,Gene therapy ,Organ Culture Techniques ,Rare Diseases ,Electroretinography ,medicine ,Animals ,Humans ,Photoreceptor Cells ,Eye Proteins ,Eye Disease and Disorders of Vision ,Molecular Biology ,5.2 Cellular and gene therapies ,Animal ,Vertebrate ,Neurosciences ,Retinal ,Genetic Therapy ,medicine.disease ,Mice, Mutant Strains ,eye diseases ,Mice, Inbred C57BL ,Disease Models, Animal ,chemistry ,Disease Models ,sense organs ,cell targeting ,RETINOSCHISIN ,Cell Adhesion Molecules - Abstract
X-linked retinoschisis, a disease characterized by splitting of the retina, is caused by mutations in the retinoschisin gene, which encodes a putative secreted cell adhesion protein. Currently, there is no effective treatment for retinoschisis, though viral vector-mediated gene replacement therapies offer promise. We used intravitreal delivery of three different AAV vectors to target delivery of the RS1 gene to Müller glia, photoreceptors or multiple cell types throughout the retina. Müller glia radially span the entire retina, are accessible from the vitreous, and remain intact throughout progression of the disease. However, photoreceptors, not glia, normally secrete retinoschisin. We compared the efficacy of rescue mediated by retinoschisin secretion from these specific subtypes of retinal cells in the Rs1h-/- mouse model of retinoschisis. Our results indicate that all three vectors deliver the RS1 gene, and that several cell types can secrete retinoschisin, leading to transport of the protein across the retina. The greatest long-term rescue was observed when photoreceptors produce retinoschisin. Similar rescue was observed with photoreceptor-specific or generalized expression, although photoreceptor secretion may contribute to rescue in the latter case. These results collectively point to the importance of cell targeting and appropriate vector choice in the success of retinal gene therapies.
- Published
- 2014
44. Vertebrate Cone Opsins Enable Sustained and Highly Sensitive Rapid Control of G i/o Signaling in Anxiety Circuitry
- Author
-
Johanna M. Rubelowski, Deniz Dalkara, Takashi Maejima, Evan S. Deneris, Lutz Wallhorn, Olivia Andrea Masseck, Stefan Herlitze, and Katharina Spoida
- Subjects
Opsin ,Light ,genetic structures ,Neuroscience(all) ,Anxiety ,GTP-Binding Protein alpha Subunits, Gi-Go ,Biology ,Mice ,Dorsal raphe nucleus ,In vivo ,Animals ,Humans ,Receptor ,G protein-coupled receptor ,Neurons ,Behavior, Animal ,General Neuroscience ,HEK 293 cells ,Rod Opsins ,Cone Opsins ,Receptor, Serotonin, 5-HT1A ,Retinal Cone Photoreceptor Cells ,sense organs ,Serotonin ,Signal transduction ,Neuroscience ,Signal Transduction - Abstract
SummaryG protein-coupled receptors (GPCRs) coupling to Gi/o signaling pathways are involved in the control of important physiological functions, which are difficult to investigate because of the limitation of tools to control the signaling pathway with precise kinetics and specificity. We established two vertebrate cone opsins, short- and long-wavelength opsin, for long-lasting and repetitive activation of Gi/o signaling pathways in vitro and in vivo. We demonstrate for both opsins the repetitive fast, membrane-delimited, ultra light-sensitive, and wavelength-dependent activation of the Gi/o pathway in HEK cells. We also show repetitive control of Gi/o pathway activation in 5-HT1A receptor domains in the dorsal raphe nucleus (DRN) in brain slices and in vivo, which is sufficient to modulate anxiety behavior in mice. Thus, vertebrate cone opsins represent a class of tools for understanding the role of Gi/o-coupled GPCRs in health and disease.Video Abstract
- Published
- 2014
45. Gene therapy for inherited retinal degenerations
- Author
-
Deniz Dalkara and José-Alain Sahel
- Subjects
Clinical Trials as Topic ,Retina ,General Immunology and Microbiology ,Genetic enhancement ,Retinal Degeneration ,Genetic Therapy ,General Medicine ,Helper dependent virus ,Biology ,Blindness ,General Biochemistry, Genetics and Molecular Biology ,Viral vector ,Clinical trial ,medicine.anatomical_structure ,RPE65 ,Immune privilege ,Immunology ,medicine ,Humans ,RNA Interference ,Vector (molecular biology) ,General Agricultural and Biological Sciences ,Neuroscience - Abstract
Gene therapy is quickly becoming a reality applicable in the clinic for inherited retinal diseases. Progress over the past decade has moved proof-of-concept gene therapies from bench to bedside. The remarkable success in safety and efficacy, in the phase I/II clinical trials for the form of the severe childhood-onset blindness, Leber's Congenital Amaurosis (LCA) type II (due to mutations in the RPE65 gene) generated significant interest and opened up possibilities for a new era of retinal gene therapies. Success in these clinical trials was due to combining the favorable features of both the retina as a target organ and adeno-associated virus (AAV) as a vector. The retina offers several advantages for gene therapy approaches. It is an anatomically defined structure that is readily accessible for therapy and has some degree of immune privilege, making it suitable for application of viral vectors. AAV, on the other hand, is a non-pathogenic helper dependent virus that has little immunogenicity. This viral vector transduces quiescent cells efficiently and thanks to its small size diffuses well in the interneural matrix, making it suitable for applications in neural tissue. Building on this initial clinical success with LCA II, we have now many opportunities to extend this proof-of-concept to other retinal diseases. This article will discuss what are some of the most imminent targets for such therapies and what are the challenges that we face in moving these therapies to the clinic.
- Published
- 2014
46. Emerging therapies for inherited retinal degeneration
- Author
-
Botond Roska, Mandeep S. Singh, Serge Picaud, Deniz Dalkara, Rupert W. Strauss, José-Alain Sahel, and Hendrik P. N. Scholl
- Subjects
0301 basic medicine ,Retinal degeneration ,Pathology ,medicine.medical_specialty ,genetic structures ,Genetic enhancement ,medicine.medical_treatment ,Disease ,Biology ,Retina ,Photoreceptor cell ,03 medical and health sciences ,chemistry.chemical_compound ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Photoreceptor Cells ,Molecular Biology ,Vision, Ocular ,Clinical Trials as Topic ,Stem Cells ,Retinal Degeneration ,Retinal ,Genetic Therapy ,General Medicine ,Stem-cell therapy ,medicine.disease ,Visual Prosthesis ,Optogenetics ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Stem cell ,Neuroscience ,Stem Cell Transplantation - Abstract
Inherited retinal degenerative diseases, a genetically and phenotypically heterogeneous group of disorders, affect the function of photoreceptor cells and are among the leading causes of blindness. Recent advances in molecular genetics and cell biology are elucidating the pathophysiological mechanisms underlying these disorders and are helping to identify new therapeutic approaches, such as gene therapy, stem cell therapy, and optogenetics. Several of these approaches have entered the clinical phase of development. Artificial replacement of dying photoreceptor cells using retinal prostheses has received regulatory approval. Precise retinal imaging and testing of visual function are facilitating more efficient clinical trial design. In individual patients, disease stage will determine whether the therapeutic strategy should comprise photoreceptor cell rescue to delay or arrest vision loss or retinal replacement for vision restoration.
- Published
- 2016
47. Visual restoration by an infrared photovoltaic implant and by optogenetic therapy: validation in non-human primates
- Author
-
Deniz Dalkara, Kevin Gehere, Stéphane Bertin, Serge Picaud, Céline Nouvel-Jaillard, Gregory Gauvain, Elena Brazhnikova, Mina A. Khoei, Ryad Benosman, Jens Duebel, Philippe Hantraye, Claire-Maëlle Fovet, Pierre Pouget, Paul-Henri Prevot, José-Alain Sahel, Himanshu Akolkar, and Yannick LeMer
- Subjects
Ophthalmology ,Computer science ,Photovoltaic system ,Implant ,Optogenetics ,Sensory Systems ,Biomedical engineering - Published
- 2019
48. In vivo sub-millisecond two-photon optogenetics with temporally focused patterned light
- Author
-
Deniz Dalkara, Valentina Emiliani, Tanya L. Daigle, Emiliano Ronzitti, Brian Lee, I-Wen Chen, Hongkui Zeng, Eirini Papagiakoumou, Chen, I-Wen, Appel à projets générique - Éclairer la fonction des neurones hubs de l'hippocampe adulte chez la souris éveillée par microscopie à modulation de front d'onde - - HOLOHUB2014 - ANR-14-CE13-0016 - Appel à projets générique - VALID, Holographie tri-dimensionelle pour l'activation parallèle des neurones - - 3DHoloPAc2015 - ANR-15-CE19-0001 - AAPG2015 - VALID, Holistic evaluation of light and multiwave applications to high resolution imaging in ophthalmic translational research revisiting the helmholtzian synergies - HELMHOLTZ - - EC:FP7:ERC2014-08-01 - 2020-07-31 - 610110 - VALID, Institut de la Vision, Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Laboratoire de Neurophotonique (UMR 8250), Université Paris Descartes - Paris 5 (UPD5)-Centre National de la Recherche Scientifique (CNRS), Allen Institute for Brain Science [Seattle, WA, USA], This work was supported by the European Union’s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie Grant Agreement no. 747598 (to I.-W.C.). E.P. and V.E. acknowledge support from the Agence Nationale de la Recherche ANR (Grants ANR-14-CE13-0016, Holohub and ANR-15-CE19-0001-01, 3DHoloPAc). V.E. acknowledges support from the Human Frontiers Science Program (Grant RGP0015/2016), the National Institutes of Health (Grant NIH U01NS090501-03), and Gettylab. E.R. received funding from the European Research Council Synergy Grant scheme (Helmholtz, ERC Grant Agreement 610110)., ANR-14-CE13-0016,HOLOHUB,Éclairer la fonction des neurones hubs de l'hippocampe adulte chez la souris éveillée par microscopie à modulation de front d'onde(2014), ANR-15-CE19-0001,3DHoloPAc,Holographie tri-dimensionelle pour l'activation parallèle des neurones(2015), European Project: 610110,EC:FP7:ERC,ERC-2013-SyG,HELMHOLTZ(2014), and Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,computer-generated holography ,millisecond photoactivation ,Optogenetics ,in vivo two-photon optogenetics ,Cell morphology ,03 medical and health sciences ,0302 clinical medicine ,Calcium imaging ,Two-photon excitation microscopy ,medicine ,Premovement neuronal activity ,[SDV.NEU] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,Research Articles ,Physics ,General Neuroscience ,Computer-generated holography ,030104 developmental biology ,Visual cortex ,medicine.anatomical_structure ,Temporal resolution ,temporal focusing ,mouse visual cortex ,[SDV.NEU]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC] ,Neuroscience ,030217 neurology & neurosurgery - Abstract
To better examine circuit mechanisms underlying perception and behavior, researchers need tools to enable temporally precise control of action-potential generation of individual cells from neuronal ensembles. Here we demonstrate that such precision can be achieved with two-photon (2P) temporally focused computer-generated holography to control neuronal excitability at the supragranular layers of anesthetized and awake visual cortex in both male and female mice. Using 2P-guided whole-cell or cell-attached recordings in positive neurons expressing any of the three opsins ReaChR, CoChR, or ChrimsonR, we investigated the dependence of spiking activity on the opsin's channel kinetics. We found that in all cases the use of brief illumination (≤10 ms) induces spikes of millisecond temporal resolution and submillisecond precision, which were preserved upon repetitive illuminations up to tens of hertz. To reach high temporal precision, we used a large illumination spot covering the entire cell body and an amplified laser at high peak power and low excitation intensity (on average ≤0.2 mW/μm(2)), thus minimizing the risk for nonlinear photodamage effects. Finally, by combining 2P holographic excitation with electrophysiological recordings and calcium imaging using GCaMP6s, we investigated the factors, including illumination shape and intensity, opsin distribution in the target cell, and cell morphology, which affect the spatial selectivity of single-cell and multicell holographic activation. Parallel optical control of neuronal activity with cellular resolution and millisecond temporal precision should make it easier to investigate neuronal connections and find further links between connectivity, microcircuit dynamics, and brain functions. SIGNIFICANCE STATEMENT Recent developments in the field of optogenetics has enabled researchers to probe the neuronal microcircuit with light by optically actuating genetically encoded light-sensitive opsins expressed in the target cells. Here, we applied holographic light shaping and temporal focusing to simultaneously deliver axially confined holographic patterns to opsin-positive cells in the living mouse cortex. Parallel illumination efficiently induced action potentials with high temporal resolution and precision for three opsins of different kinetics. We extended the parallel optogenetic activation at low intensity to multiple neurons and concurrently monitored their calcium dynamics. These results demonstrate fast and temporally precise in vivo control of a neuronal subpopulation, opening new opportunities for revealing circuit mechanisms underlying brain functions.
- Published
- 2019
49. AAV-mediated gene delivery in Dp71-null mouse model with compromised barriers
- Author
-
Ophélie Vacca, Deniz Dalkara, Alvaro Rendon, Marie Darche, John G. Flannery, David V. Schaffer, and José-Alain Sahel
- Subjects
Genetic enhancement ,Cell ,Blood–retinal barrier ,Inner limiting membrane ,Retinal ,Biology ,Gene delivery ,Cell biology ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Transduction (genetics) ,medicine.anatomical_structure ,Neurology ,chemistry ,Immunology ,medicine ,sense organs ,Receptor - 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 Muller 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 Muller 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 Muller cells specifically. ShH10 coding GFP was introduced by intravitreal injection and Muller cell transduction was studied in Dp71-null mice in comparison to wild-type animals. We show that Muller 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.
- Published
- 2013
50. Smart CRISPR-Cas9 liver delivery repairs a genetic defect
- Author
-
Deniz Dalkara
- Subjects
chemistry.chemical_classification ,chemistry.chemical_compound ,Ammonia ,Enzyme ,chemistry ,Biochemistry ,Urea cycle ,Carbamoyl phosphate ,Citrulline ,Ornithine transcarbamylase ,CRISPR ,General Medicine ,Ornithine - Abstract
Babies born deficient in the enzyme ornithine transcarbamylase (OTC) quickly experience lethal metabolic crises. In this common urea-cycle disorder, carbamoyl phosphate and ornithine cannot be converted into citrulline, a key intermediate in the urea cycle through which mammals excrete toxic ammonia
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.