1
|
Sun C, Fan Q, Xie R, Luo C, Hu B, Wang Q. Tetherless Optical Neuromodulation: Wavelength from Orange-red to Mid-infrared. Neurosci Bull 2024; 40:1173-1188. [PMID: 38372931 PMCID: PMC11306867 DOI: 10.1007/s12264-024-01179-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/11/2023] [Indexed: 02/20/2024] Open
Abstract
Optogenetics, a technique that employs light for neuromodulation, has revolutionized the study of neural mechanisms and the treatment of neurological disorders due to its high spatiotemporal resolution and cell-type specificity. However, visible light, particularly blue and green light, commonly used in conventional optogenetics, has limited penetration in biological tissue. This limitation necessitates the implantation of optical fibers for light delivery, especially in deep brain regions, leading to tissue damage and experimental constraints. To overcome these challenges, the use of orange-red and infrared light with greater tissue penetration has emerged as a promising approach for tetherless optical neuromodulation. In this review, we provide an overview of the development and applications of tetherless optical neuromodulation methods with long wavelengths. We first discuss the exploration of orange-red wavelength-responsive rhodopsins and their performance in tetherless optical neuromodulation. Then, we summarize two novel tetherless neuromodulation methods using near-infrared light: upconversion nanoparticle-mediated optogenetics and photothermal neuromodulation. In addition, we discuss recent advances in mid-infrared optical neuromodulation.
Collapse
Affiliation(s)
- Chao Sun
- Key Laboratory of Spectral Imaging Technology, Xi'an Institute of Optics and Precision Mechanics (XIOPM), Chinese Academy of Sciences, Xi'an, 710119, China
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China
| | - Qi Fan
- Key Laboratory of Spectral Imaging Technology, Xi'an Institute of Optics and Precision Mechanics (XIOPM), Chinese Academy of Sciences, Xi'an, 710119, China
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China
| | - Rougang Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Bingliang Hu
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China
| | - Quan Wang
- Key Laboratory of Spectral Imaging Technology, Xi'an Institute of Optics and Precision Mechanics (XIOPM), Chinese Academy of Sciences, Xi'an, 710119, China.
- Key Laboratory of Biomedical Spectroscopy of Xi'an, Key Laboratory of Spectral Imaging Technology, XIOPM, Chinese Academy of Sciences, Xi'an, 710119, China.
| |
Collapse
|
2
|
Katada Y, Yoshida K, Serizawa N, Lee D, Kobayashi K, Negishi K, Okano H, Kandori H, Tsubota K, Kurihara T. Highly sensitive visual restoration and protection via ectopic expression of chimeric rhodopsin in mice. iScience 2023; 26:107716. [PMID: 37720108 PMCID: PMC10504486 DOI: 10.1016/j.isci.2023.107716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/22/2023] [Accepted: 08/22/2023] [Indexed: 09/19/2023] Open
Abstract
Photoreception requires amplification by mammalian rhodopsin through G protein activation, which requires a visual cycle. To achieve this in retinal gene therapy, we incorporated human rhodopsin cytoplasmic loops into Gloeobacter rhodopsin, thereby generating Gloeobacter and human chimeric rhodopsin (GHCR). In a murine model of inherited retinal degeneration, we induced retinal GHCR expression by intravitreal injection of a recombinant adeno-associated virus vector. Retinal explant and visual thalamus electrophysiological recordings, behavioral tests, and histological analysis showed that GHCR restored dim-environment vision and prevented the progression of retinal degeneration. Thus, GHCR may be a potent clinical tool for the treatment of retinal disorders.
Collapse
Affiliation(s)
- Yusaku Katada
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazuho Yoshida
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Aichi 466-0061, Japan
| | - Naho Serizawa
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Nutritional Sciences, Toyo University, Kita-ku, Tokyo 115-8650, Japan
| | - Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideki Kandori
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, Aichi 466-0061, Japan
| | - Kazuo Tsubota
- Tsubota Laboratory, Inc., Shinjuku-ku, Tokyo 160-0016, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
3
|
Hatakeyama A, Sugano E, Sayama T, Watanabe Y, Suzuki T, Tabata K, Endo Y, Sakajiri T, Fukuda T, Ozaki T, Tomita H. Properties of a Single Amino Acid Residue in the Third Transmembrane Domain Determine the Kinetics of Ambient Light-Sensitive Channelrhodopsin. Int J Mol Sci 2023; 24:ijms24055054. [PMID: 36902480 PMCID: PMC10003734 DOI: 10.3390/ijms24055054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Channelrhodopsins have been utilized in gene therapy to restore vision in patients with retinitis pigmentosa and their channel kinetics are an important factor to consider in such applications. We investigated the channel kinetics of ComV1 variants with different amino acid residues at the 172nd position. Patch clamp methods were used to record the photocurrents induced by stimuli from diodes in HEK293 cells transfected with plasmid vectors. The channel kinetics (τon and τoff) were considerably altered by the replacement of the 172nd amino acid and was dependent on the amino acid characteristics. The size of amino acids at this position correlated with τon and decay, whereas the solubility correlated with τon and τoff. Molecular dynamic simulation indicated that the ion tunnel constructed by H172, E121, and R306 widened due to H172A variant, whereas the interaction between A172 and the surrounding amino acids weakened compared with H172. The bottleneck radius of the ion gate constructed with the 172nd amino acid affected the photocurrent and channel kinetics. The 172nd amino acid in ComV1 is a key residue for determining channel kinetics as its properties alter the radius of the ion gate. Our findings can be used to improve the channel kinetics of channelrhodopsins.
Collapse
|
4
|
Sakai D, Tomita H, Maeda A. Optogenetic Therapy for Visual Restoration. Int J Mol Sci 2022; 23:15041. [PMID: 36499371 PMCID: PMC9735806 DOI: 10.3390/ijms232315041] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022] Open
Abstract
Optogenetics is a recent breakthrough in neuroscience, and one of the most promising applications is the treatment of retinal degenerative diseases. Multiple clinical trials are currently ongoing, less than a decade after the first attempt at visual restoration using optogenetics. Optogenetic therapy has great value in providing hope for visual restoration in late-stage retinal degeneration, regardless of the genotype. This alternative gene therapy consists of multiple elements including the choice of target retinal cells, optogenetic tools, and gene delivery systems. Currently, there are various options for each element, all of which have been developed as a product of technological success. In particular, the performance of optogenetic tools in terms of light and wavelength sensitivity have been improved by engineering microbial opsins and applying human opsins. To provide better post-treatment vision, the optimal choice of optogenetic tools and effective gene delivery to retinal cells is necessary. In this review, we provide an overview of the advancements in optogenetic therapy for visual restoration, focusing on available options for optogenetic tools and gene delivery methods.
Collapse
Affiliation(s)
- Daiki Sakai
- Department of Ophthalmology, Kobe City Eye Hospital, Kobe 650-0047, Japan
- Department of Ophthalmology, Kobe City Medical Center General Hospital, Kobe 650-0047, Japan
- Department of Surgery, Division of Ophthalmology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Hiroshi Tomita
- Graduate Course in Biological Sciences, Division of Science and Engineering, Iwate University, Iwate 020-8550, Japan
| | - Akiko Maeda
- Department of Ophthalmology, Kobe City Eye Hospital, Kobe 650-0047, Japan
- Department of Ophthalmology, Kobe City Medical Center General Hospital, Kobe 650-0047, Japan
| |
Collapse
|
5
|
Lindner M, Gilhooley MJ, Hughes S, Hankins MW. Optogenetics for visual restoration: From proof of principle to translational challenges. Prog Retin Eye Res 2022; 91:101089. [PMID: 35691861 DOI: 10.1016/j.preteyeres.2022.101089] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 01/04/2023]
Abstract
Degenerative retinal disorders are a diverse family of diseases commonly leading to irreversible photoreceptor death, while leaving the inner retina relatively intact. Over recent years, innovative gene replacement therapies aiming to halt the progression of certain inherited retinal disorders have made their way into clinics. By rendering surviving retinal neurons light sensitive optogenetic gene therapy now offers a feasible treatment option that can restore lost vision, even in late disease stages and widely independent of the underlying cause of degeneration. Since proof-of-concept almost fifteen years ago, this field has rapidly evolved and a detailed first report on a treated patient has recently been published. In this article, we provide a review of optogenetic approaches for vision restoration. We discuss the currently available optogenetic tools and their relative advantages and disadvantages. Possible cellular targets will be discussed and we will address the question how retinal remodelling may affect the choice of the target and to what extent it may limit the outcomes of optogenetic vision restoration. Finally, we will analyse the evidence for and against optogenetic tool mediated toxicity and will discuss the challenges associated with clinical translation of this promising therapeutic concept.
Collapse
Affiliation(s)
- Moritz Lindner
- The Nuffield Laboratory of Ophthalmology, Jules Thorn SCNi, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3QU, United Kingdom; Institute of Physiology and Pathophysiology, Department of Neurophysiology, Philipps University, 35037, Marburg, Germany
| | - Michael J Gilhooley
- The Nuffield Laboratory of Ophthalmology, Jules Thorn SCNi, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3QU, United Kingdom; The Institute of Ophthalmology, University College London, EC1V 9EL, United Kingdom; Moorfields Eye Hospital, London, EC1V 2PD, United Kingdom
| | - Steven Hughes
- The Nuffield Laboratory of Ophthalmology, Jules Thorn SCNi, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3QU, United Kingdom
| | - Mark W Hankins
- The Nuffield Laboratory of Ophthalmology, Jules Thorn SCNi, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX1 3QU, United Kingdom.
| |
Collapse
|
6
|
Tredicine M, Camponeschi C, Pirolli D, Lucchini M, Valentini M, Geloso MC, Mirabella M, Fidaleo M, Righino B, Moliterni C, Giorda E, Rende M, De Rosa MC, Foti M, Constantin G, Ria F, Di Sante G. A TLR/CD44 axis regulates T cell trafficking in experimental and human multiple sclerosis. iScience 2022; 25:103763. [PMID: 35128357 PMCID: PMC8804271 DOI: 10.1016/j.isci.2022.103763] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/28/2021] [Accepted: 01/07/2022] [Indexed: 12/14/2022] Open
Abstract
In the pathogenesis of autoimmune disorders, the modulation of leukocytes' trafficking plays a central role, still poorly understood. Here, we focused on the effect of TLR2 ligands in trafficking of T helper cells through reshuffling of CD44 isoforms repertoire. Concurrently, strain background and TLR2 haplotype affected Wnt/β-catenin signaling pathway and expression of splicing factors. During EAE, mCD44 v9- v 10 was specifically enriched in the forebrain and showed an increased ability to bind stably to osteopontin. Similarly, we observed that hCD44 v7 was highly enriched in cells of cerebrospinal fluid from MS patients with active lesions. Moreover, TLRs engagement modulated the composition of CD44 variants also in human T helper cells, supporting the hypothesis that pathogens or commensals, through TLRs, in turn modulate the repertoire of CD44 isoforms, thereby controlling the distribution of lesions in the CNS. The interference with this mechanism(s) represents a potential tool for prevention and treatment of autoimmune relapses and exacerbations.
Collapse
Affiliation(s)
- Maria Tredicine
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Chiara Camponeschi
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Davide Pirolli
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC) -CNR, Largo Francesco Vito 1,00168 Rome, Italy
| | - Matteo Lucchini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli1-8,00168 Rome, Italy
- Centro di ricerca per la Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1,00168 Rome, Italy
| | - Mariagrazia Valentini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli1-8,00168 Rome, Italy
| | - Maria Concetta Geloso
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli1-8,00168 Rome, Italy
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1,00168 Rome, Italy
| | - Massimiliano Mirabella
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli1-8,00168 Rome, Italy
- Centro di ricerca per la Sclerosi Multipla (CERSM), Università Cattolica del Sacro Cuore, Largo Francesco Vito 1,00168 Rome, Italy
| | - Marco Fidaleo
- Department of Biology and Biotechnology Charles Darwin, University of Rome Sapienza,00185 Rome, Italy
| | - Benedetta Righino
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC) -CNR, Largo Francesco Vito 1,00168 Rome, Italy
| | - Camilla Moliterni
- Department of Biology and Biotechnology Charles Darwin, University of Rome Sapienza,00185 Rome, Italy
| | - Ezio Giorda
- Core Facilities di Ricerca, Ospedale Pediatrico Bambino Gesù Roma – IRCCS, V.le Ferdinando Baldelli,40,00146 Roma, Italy
| | - Mario Rende
- Department of Medicine and Surgery, Section of Human, Clinic and Forensic Anatomy, University of Perugia, Piazza L. Severi, 06132 Perugia, Italy
| | - Maria Cristina De Rosa
- Institute of Chemical Sciences and Technologies “Giulio Natta” (SCITEC) -CNR, Largo Francesco Vito 1,00168 Rome, Italy
| | - Maria Foti
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Gabriela Constantin
- Department of Medicine, Section of General Pathology, University of Verona, Strada le Grazie 8,37134 Verona, Italy
| | - Francesco Ria
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli1-8,00168 Rome, Italy
| | - Gabriele Di Sante
- Department of Translational Medicine and Surgery, Section of General Pathology, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Medicine and Surgery, Section of Human, Clinic and Forensic Anatomy, University of Perugia, Piazza L. Severi, 06132 Perugia, Italy
| |
Collapse
|
7
|
Development of an optogenetic gene sensitive to daylight and its implications in vision restoration. NPJ Regen Med 2021; 6:64. [PMID: 34650094 PMCID: PMC8516861 DOI: 10.1038/s41536-021-00177-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/29/2021] [Indexed: 11/14/2022] Open
Abstract
Optogenetic gene-mediated therapy for restoring vision is thought to be a useful treatment for blind patients. However, light sensitivity achieved using this gene therapy is inferior to that of daylight vision. To increase light sensitivity, we designed three mutants using a bioinformatics approach. Nucleotide sequences encoding two sites in the extracellular loops (ex1, ex3) of mVChR1 close to simulated ion-conducting pathways were replaced by homologous amino acid-encoding sequences of ChR1 or ChR2. The light sensitivity of ex3mV1 was higher than that of mVChR1 at 405–617 nm. Visual responses were restored in Royal College of Surgeons rats with genetically degenerating photoreceptor cells transfected with ex3mV1Co, wherein transmembrane of sixth (TM6) in ex3mV1 was additionally replaced with the corresponding domain of CoChR; these rats responded to light in the order of μW/mm2. Thus, ex3mV1Co might be useful for the restoration of advanced visual function.
Collapse
|
8
|
Van Gelder RN. Gene Therapy Approaches to Slow or Reverse Blindness From Inherited Retinal Degeneration: Growth Factors and Optogenetics. Int Ophthalmol Clin 2021; 61:209-228. [PMID: 34584058 PMCID: PMC8486303 DOI: 10.1097/iio.0000000000000386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To date, clinical gene therapy efforts for inherited retinal degeneration (IRD) have focused largely on gene replacement. The large number of genes and alleles causing IRD, however, makes this approach practical only for the most common causes. Additionally, gene replacement therapy cannot reverse existing retinal degeneration. Viral-mediated gene therapy can be used for two other approaches to slow or reverse IRD. First, by driving intraocular expression of growth factors or neuroprotective proteins, retinal degeneration can be slowed. Second, by expressing light-sensitive proteins (either microbial channelopsins or mammalian G-protein coupled opsins) in preserved inner retinal neurons, light sensitivity can be restored to the blind retina. Both approaches have advanced substantially in the past decade, and both are nearing clinical tests. This review surveys recent progress in these approaches.
Collapse
|
9
|
Zhou Z, Liao L. Optogenetic Neuromodulation of the Urinary Bladder. Neuromodulation 2021; 24:1229-1236. [PMID: 34375470 DOI: 10.1111/ner.13516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/27/2021] [Accepted: 07/13/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Nerve stimulation and neuromodulation have become acceptable interventions for bladder dysfunction. However, electrical stimulation indiscriminately affects all types of cells and can lead to treatment failure and off-target effects. In recent years, advancement of knowledge of optogenetics provides a powerful tool to enable precise, minimally invasive neuromodulation. MATERIALS AND METHODS In this review, we introduce basic knowledge about optogenetics; discuss the progression of engineered opsins, gene-targeting methods, and light-delivery approaches; we also summarize the application of optogenetics in neuromodulation of the bladder and discuss the possible clinical translation in the future. RESULTS AND CONCLUSION Optogenetics offers a powerful tool to investigate the neural circuit of bladder storage and voiding and provides a promising approach for manipulating neurons and muscles. It is possible to achieve coordinated modulation of the bladder and its sphincter through a "closed-loop" system. Optogenetics neuromodulation could also be applied in urinary bladder control in the clinic in the future.
Collapse
Affiliation(s)
- Zhonghan Zhou
- Cheeloo College of Medicine, Shandong University, Jinan, China; Department of Urology, China Rehabilitation Research Center, Beijing, China.,University of Health and Rehabilitation Sciences, Qingdao, China
| | - Limin Liao
- Cheeloo College of Medicine, Shandong University, Jinan, China; Department of Urology, China Rehabilitation Research Center, Beijing, China.,University of Health and Rehabilitation Sciences, Qingdao, China.,School of Rehabilitation, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Phototoxicities Caused by Continuous Light Exposure Were Not Induced in Retinal Ganglion Cells Transduced by an Optogenetic Gene. Int J Mol Sci 2021; 22:ijms22136732. [PMID: 34201658 PMCID: PMC8269149 DOI: 10.3390/ijms22136732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 11/16/2022] Open
Abstract
The death of photoreceptor cells is induced by continuous light exposure. However, it is unclear whether light damage was induced in retinal ganglion cells with photosensitivity by transduction of optogenetic genes. In this study, we evaluated the phototoxicities of continuous light exposure on retinal ganglion cells after transduction of the optogenetic gene mVChR1 using an adeno-associated virus vector. Rats were exposed to continuous light for a week, and visually evoked potentials (VEPs) were recorded. The intensities of continuous light (500, 1000, 3000, and 5000 lx) increased substantially after VEP recordings. After the final recording of VEPs, retinal ganglion cells (RGCs) were retrogradely labeled with a fluorescein tracer, FluoroGold, and the number of retinal ganglion cells was counted under a fluorescent microscope. There was no significant reduction in the amplitudes of VEPs and the number of RGCs after exposure to any light intensity. These results indicated that RGCs were photosensitive after the transduction of optogenetic genes and did not induce any phototoxicity by continuous light exposure.
Collapse
|
11
|
Optogenetics-Mediated Gene Therapy for Retinal Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:535-543. [PMID: 33398840 DOI: 10.1007/978-981-15-8763-4_37] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The visual system consists of various types of neurons and a single-nucleotide mutation can sometimes lead to blindness. The phototransduction pathway in the retina starts from the first-order neurons, the photoreceptor cells, and transmits the signals to second-order neurons. Finally, the output signal from third-order neurons, the ganglion cells, is carried to the brain. The photoreceptor cells are the only neurons in the retina that can respond to a light signal; they are hyperpolarised when they receive light, and the ganglion cells carry the signals to the brain following the depolarization. Recently, various types of channelrhodopsins have been found and developed. It is expected that the gene therapies using the cation channel as well as anion channelrhodopsin genes would be effective against diseases that cause severe destruction of visual function, such as the retinitis pigmentosa and age-related macular degeneration. In this review, we mainly describe mVChR1-mediated gene therapy for retinitis pigmentosa and the future application of optogenetic genes in retinal diseases.
Collapse
|
12
|
Lam PY, Thawani AR, Balderas E, White AJP, Chaudhuri D, Fuchter MJ, Peterson RT. TRPswitch-A Step-Function Chemo-optogenetic Ligand for the Vertebrate TRPA1 Channel. J Am Chem Soc 2020; 142:17457-17468. [PMID: 32966062 DOI: 10.1021/jacs.0c06811] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chemo-optogenetics has produced powerful tools for optical control of cell activity, but current tools suffer from a variety of limitations including low unitary conductance, the need to modify the target channel, or the inability to control both on and off switching. Using a zebrafish behavior-based screening strategy, we discovered "TRPswitch", a photoswitchable nonelectrophilic ligand scaffold for the transient receptor potential ankyrin 1 (TRPA1) channel. TRPA1 exhibits high unitary channel conductance, making it an ideal target for chemo-optogenetic tool development. Key molecular determinants for the activity of TRPswitch were elucidated and allowed for replacement of the TRPswitch azobenzene with a next-generation azoheteroarene. The TRPswitch compounds enable reversible, repeatable, and nearly quantitative light-induced activation and deactivation of the vertebrate TRPA1 channel with violet and green light, respectively. The utility of TRPswitch compounds was demonstrated in larval zebrafish hearts exogenously expressing zebrafish Trpa1b, where the heartbeat could be controlled using TRPswitch and light. Therefore, TRPA1/TRPswitch represents a novel step-function chemo-optogenetic system with a unique combination of high conductance, high efficiency, activity against an unmodified vertebrate channel, and capacity for bidirectional optical switching. This chemo-optogenetic system will be particularly applicable in systems where a large depolarization current is needed or sustained channel activation is desirable.
Collapse
Affiliation(s)
- Pui-Ying Lam
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, United States
| | - Aditya R Thawani
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, White City Campus, Wood Lane, London W12 OBZ, United Kingdom
| | - Enrique Balderas
- Department of Internal Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah 84112, United States
| | - Andrew J P White
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, White City Campus, Wood Lane, London W12 OBZ, United Kingdom
| | - Dipayan Chaudhuri
- Department of Internal Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah 84112, United States
| | - Matthew J Fuchter
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, White City Campus, Wood Lane, London W12 OBZ, United Kingdom
| | - Randall T Peterson
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
13
|
Shen Y, Campbell RE, Côté DC, Paquet ME. Challenges for Therapeutic Applications of Opsin-Based Optogenetic Tools in Humans. Front Neural Circuits 2020; 14:41. [PMID: 32760252 PMCID: PMC7373823 DOI: 10.3389/fncir.2020.00041] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/16/2020] [Indexed: 12/29/2022] Open
Abstract
As the technological hurdles are overcome and optogenetic techniques advance to have more control over neurons, therapies based on these approaches will begin to emerge in the clinic. Here, we consider the technical challenges surrounding the transition of this breakthrough technology from an investigative tool to a true therapeutic avenue. The emerging strategies and remaining tasks surrounding genetically encoded molecules which respond to light as well as the vehicles required to deliver them are discussed.The use of optogenetics in humans would represent a completely new paradigm in medicine and would be associated with unprecedented technical considerations. To be applied for stimulation of neurons in humans, an ideal optogenetic tool would need to be non-immunogenic, highly sensitive, and activatable with red light or near-infrared light (to maximize light penetration while minimizing photodamage). To enable sophisticated levels of neuronal control, the combined use of optogenetic actuators and indicators could enable closed-loop all-optical neuromodulation. Such systems would introduce additional challenges related to spectral orthogonality between actuator and indicator, the need for decision making computational algorithms and requirements for large gene cassettes. As in any gene therapy, the therapeutic efficiency of optogenetics will rely on vector delivery and expression in the appropriate cell type. Although viral vectors such as those based on AAVs are showing great potential in human trials, barriers to their general use remain, including immune responses, delivery/transport, and liver clearance. Limitations associated with the gene cassette size which can be packaged in currently approved vectors also need to be addressed.
Collapse
Affiliation(s)
- Yi Shen
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Robert E Campbell
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada.,Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Daniel C Côté
- Centre de Recherche CERVO, Université Laval, Quebec City, QC, Canada.,Département de Physique et Génie Physique, Université Laval, Quebec City, QC, Canada
| | - Marie-Eve Paquet
- Centre de Recherche CERVO, Université Laval, Quebec City, QC, Canada.,Département de Biochimie, Microbiologie et Bioinformatique, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
14
|
Richardson RT, Ibbotson MR, Thompson AC, Wise AK, Fallon JB. Optical stimulation of neural tissue. Healthc Technol Lett 2020; 7:58-65. [PMID: 32754339 PMCID: PMC7353819 DOI: 10.1049/htl.2019.0114] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/08/2020] [Accepted: 05/15/2020] [Indexed: 12/23/2022] Open
Abstract
Electrical stimulation has been used for decades in devices such as pacemakers, cochlear implants and more recently for deep brain and retinal stimulation and electroceutical treatment of disease. However, current spread from the electrodes limits the precision of neural activation, leading to a low quality therapeutic outcome or undesired side-effects. Alternative methods of neural stimulation such as optical stimulation offer the potential to deliver higher spatial resolution of neural activation. Direct optical stimulation is possible with infrared light, while visible light can be used to activate neurons if the neural tissue is genetically modified with a light sensitive ion channel. Experimentally, both methods have resulted in highly precise stimulation with little spread of activation at least in the cochlea, each with advantages and disadvantages. Infrared neural stimulation does not require modification of the neural tissue, but has very high power requirements. Optogenetics can achieve precision of activation with lower power, but only in conjunction with targeted insertion of a light sensitive ion channel into the nervous system via gene therapy. This review will examine the advantages and limitations of optical stimulation of neural tissue, using the cochlea as an exemplary model and recent developments for retinal and deep brain stimulation.
Collapse
Affiliation(s)
- Rachael Theresa Richardson
- Bionics Institute, Melbourne 3002, Australia.,University of Melbourne, Medical Bionics Department, Melbourne, 3002, Australia.,University of Melbourne, Department of Surgery (Otolaryngology), Melbourne, 3002, Australia
| | - Michael R Ibbotson
- National Vision Research Institute, Australian College of Optometry, and Department of Optometry and Vision Science, University of Melbourne, Melbourne, Australia
| | | | - Andrew K Wise
- Bionics Institute, Melbourne 3002, Australia.,University of Melbourne, Medical Bionics Department, Melbourne, 3002, Australia.,University of Melbourne, Department of Surgery (Otolaryngology), Melbourne, 3002, Australia
| | - James B Fallon
- Bionics Institute, Melbourne 3002, Australia.,University of Melbourne, Medical Bionics Department, Melbourne, 3002, Australia.,University of Melbourne, Department of Surgery (Otolaryngology), Melbourne, 3002, Australia
| |
Collapse
|
15
|
Gundelach LA, Hüser MA, Beutner D, Ruther P, Bruegmann T. Towards the clinical translation of optogenetic skeletal muscle stimulation. Pflugers Arch 2020; 472:527-545. [PMID: 32415463 PMCID: PMC7239821 DOI: 10.1007/s00424-020-02387-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/05/2020] [Accepted: 04/28/2020] [Indexed: 12/27/2022]
Abstract
Paralysis is a frequent phenomenon in many diseases, and to date, only functional electrical stimulation (FES) mediated via the innervating nerve can be employed to restore skeletal muscle function in patients. Despite recent progress, FES has several technical limitations and significant side effects. Optogenetic stimulation has been proposed as an alternative, as it may circumvent some of the disadvantages of FES enabling cell type–specific, spatially and temporally precise stimulation of cells expressing light-gated ion channels, commonly Channelrhodopsin2. Two distinct approaches for the restoration of skeletal muscle function with optogenetics have been demonstrated: indirect optogenetic stimulation through the innervating nerve similar to FES and direct optogenetic stimulation of the skeletal muscle. Although both approaches show great promise, both have their limitations and there are several general hurdles that need to be overcome for their translation into clinics. These include successful gene transfer, sustained optogenetic protein expression, and the creation of optically active implantable devices. Herein, a comprehensive summary of the underlying mechanisms of electrical and optogenetic approaches is provided. With this knowledge in mind, we substantiate a detailed discussion of the advantages and limitations of each method. Furthermore, the obstacles in the way of clinical translation of optogenetic stimulation are discussed, and suggestions on how they could be overcome are provided. Finally, four specific examples of pathologies demanding novel therapeutic measures are discussed with a focus on the likelihood of direct versus indirect optogenetic stimulation.
Collapse
Affiliation(s)
- Lili A Gundelach
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
| | - Marc A Hüser
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center, Göttingen, Germany
| | - Dirk Beutner
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center, Göttingen, Germany
| | - Patrick Ruther
- Microsystem Materials Laboratory, Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- BrainLinks-BrainTools Cluster of Excellence at the University of Freiburg, Freiburg, Germany
| | - Tobias Bruegmann
- Institute of Cardiovascular Physiology, University Medical Center, Göttingen, Germany.
- DZHK e.V. (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
| |
Collapse
|
16
|
Xu X, Mee T, Jia X. New era of optogenetics: from the central to peripheral nervous system. Crit Rev Biochem Mol Biol 2020; 55:1-16. [PMID: 32070147 DOI: 10.1080/10409238.2020.1726279] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Optogenetics has recently gained recognition as a biological technique to control the activity of cells using light stimulation. Many studies have applied optogenetics to cell lines in the central nervous system because it has the potential to elucidate neural circuits, treat neurological diseases and promote nerve regeneration. There have been fewer studies on the application of optogenetics in the peripheral nervous system. This review introduces the basic principles and approaches of optogenetics and summarizes the physiology and mechanism of opsins and how the technology enables bidirectional control of unique cell lines with superior spatial and temporal accuracy. Further, this review explores and discusses the therapeutic potential for the development of optogenetics and its capacity to revolutionize treatment for refractory epilepsy, depression, pain, and other nervous system disorders, with a focus on neural regeneration, especially in the peripheral nervous system. Additionally, this review synthesizes the latest preclinical research on optogenetic stimulation, including studies on non-human primates, summarizes the challenges, and highlights future perspectives. The potential of optogenetic stimulation to optimize therapy for peripheral nerve injuries (PNIs) is also highlighted. Optogenetic technology has already generated exciting, preliminary evidence, supporting its role in applications to several neurological diseases, including PNIs.
Collapse
Affiliation(s)
- Xiang Xu
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Thomas Mee
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Gruber A, Edri O, Gepstein L. Cardiac optogenetics: the next frontier. Europace 2019; 20:1910-1918. [PMID: 29315402 DOI: 10.1093/europace/eux371] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022] Open
Abstract
The emerging technology of optogenetics uses optical and genetic means to monitor and modulate the electrophysiological properties of excitable tissues. While transforming the field of neuroscience, the technology has recently gained popularity also in the cardiac arena. Here, we describe the basic principles of optogenetics, the available and evolving optogenetic tools, and the unique potential of this technology for basic and translational cardiac electrophysiology. Specifically, we discuss the ability to control (augment or suppress) the cardiac tissue's excitable properties using optogenetic actuators (microbial opsins), which are light-gated ion channels and pumps that can cause light-triggered membrane depolarization or hyperpolarization. We then focus on the potential clinical implications of this technology for the treatment of cardiac arrhythmias by describing recent efforts for developing optogenetic-based cardiac pacing, resynchronization, and defibrillation experimental strategies. Finally, the significant obstacles and challenges that need to be overcome before any future clinical translation can be expected are discussed.
Collapse
Affiliation(s)
- Amit Gruber
- The Sohnis Family Reaserch Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine and Research Institute, Technion- Israel Institute of Technology, Haifa, Israel
| | - Oded Edri
- The Sohnis Family Reaserch Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine and Research Institute, Technion- Israel Institute of Technology, Haifa, Israel
| | - Lior Gepstein
- The Sohnis Family Reaserch Laboratory for Cardiac Electrophysiology and Regenerative Medicine, Rappaport Faculty of Medicine and Research Institute, Technion- Israel Institute of Technology, Haifa, Israel.,Cardiology Department of Rambam Health Care Campus, HaAliya HaShniya St 8, Haifa, Israel
| |
Collapse
|
18
|
Richter C, Bruegmann T. No light without the dark: Perspectives and hindrances for translation of cardiac optogenetics. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 154:39-50. [PMID: 31515056 DOI: 10.1016/j.pbiomolbio.2019.08.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/18/2019] [Accepted: 08/27/2019] [Indexed: 12/30/2022]
Abstract
Over the last decade, optogenetic stimulation of the heart and its translational potential for rhythm control attracted more and more interest. Optogenetics allows to stimulate cardiomyocytes expressing the light-gated cation channel Channelrhodopsin 2 (ChR2) with light and thus high spatio-temporal precision. Therefore this new approach can overcome the technical limitations of electrical stimulation. In regard of translational approaches, the prospect of pain-free stimulation, if ChR2 expression is restricted to cardiomyocytes, is especially intriguing and could be highly beneficial for cardioversion and defibrillation. However, there is no light without shadow and cardiac optogenetics has to surmount critical hurdles, namely "how" to inscribe light-sensitivity by expressing ChR2 in a native heart and how to avoid side effects such as possible immune responses against the gene transfer. Furthermore, implantable light devices have to be developed which ensure sufficient illumination in a highly contractile environment. Therefore this article reviews recent advantages in the field of cardiac optogenetics with a special focus on the hindrances for the potential translation of this new approach into clinics and provides an outlook how these have to be carefully investigated and could be solved step by step.
Collapse
Affiliation(s)
- Claudia Richter
- RG Biomedical Physics, Max Planck Institute for Dynamics & Self-Organization, Am Fassberg 17, 37077, Goettingen, Germany; Department of Cardiology and Pneumology, University Medical Center, Robert-Koch-Str. 42a, 37075, Goettingen, Germany; DZHK e.V. (German Center for Cardiovascular Research), Partner Site Goettingen, 37075, Goettingen, Germany.
| | - Tobias Bruegmann
- DZHK e.V. (German Center for Cardiovascular Research), Partner Site Goettingen, 37075, Goettingen, Germany; Institute for Cardiovascular Physiology, University Medical Center Goettingen, Humboldtallee 23, 37073, Goettingen, Germany.
| |
Collapse
|
19
|
A light in the dark: state of the art and perspectives in optogenetics and optopharmacology for restoring vision. Future Med Chem 2019; 11:463-487. [DOI: 10.4155/fmc-2018-0315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the last decade, innovative therapeutic strategies against inherited retinal degenerations (IRDs) have emerged. In particular, chemical- and opto-genetics approaches or a combination of them have been identified for modulating neuronal/optical activity in order to restore vision in blinding diseases. The ‘chemical-genetics approach’ (optopharmacology) uses small molecules (exogenous photoswitches) for restoring light sensitivity by activating ion channels. The ‘opto-genetics approach’ employs light-activated photosensitive proteins (exogenous opsins), introduced by viral vectors in injured tissues, to restore light response. These approaches offer control of neuronal activities with spatial precision and limited invasiveness, although with some drawbacks. Currently, a combined therapeutic strategy (optogenetic pharmacology) is emerging. This review describes the state of the art and provides an overview of the future perspectives in vision restoration.
Collapse
|
20
|
Garita-Hernandez M, Guibbal L, Toualbi L, Routet F, Chaffiol A, Winckler C, Harinquet M, Robert C, Fouquet S, Bellow S, Sahel JA, Goureau O, Duebel J, Dalkara D. Optogenetic Light Sensors in Human Retinal Organoids. Front Neurosci 2018; 12:789. [PMID: 30450028 PMCID: PMC6224345 DOI: 10.3389/fnins.2018.00789] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/12/2018] [Indexed: 01/26/2023] Open
Abstract
Optogenetic technologies paved the way to dissect complex neural circuits and monitor neural activity using light in animals. In retinal disease, optogenetics has been used as a therapeutic modality to reanimate the retina after the loss of photoreceptor outer segments. However, it is not clear today which ones of the great diversity of microbial opsins are best suited for therapeutic applications in human retinas as cell lines, primary cell cultures and animal models do not predict expression patterns of microbial opsins in human retinal cells. Therefore, we sought to generate retinal organoids derived from human induced pluripotent stem cells (hiPSCs) as a screening tool to explore the membrane trafficking efficacy of some recently described microbial opsins. We tested both depolarizing and hyperpolarizing microbial opsins including CatCh, ChrimsonR, ReaChR, eNpHR 3.0, and Jaws. The membrane localization of eNpHR 3.0, ReaChR, and Jaws was the highest, likely due to their additional endoplasmic reticulum (ER) release and membrane trafficking signals. In the case of opsins that were not engineered to improve trafficking efficiency in mammalian cells such as CatCh and ChrimsonR, membrane localization was less efficient. Protein accumulation in organelles such as ER and Golgi was observed at high doses with CatCh and ER retention lead to an unfolded protein response. Also, cytoplasmic localization was observed at high doses of ChrimsonR. Our results collectively suggest that retinal organoids derived from hiPSCs can be used to predict the subcellular fate of optogenetic proteins in a human retinal context. Such organoids are also versatile tools to validate other gene therapy products and drug molecules.
Collapse
Affiliation(s)
| | - Laure Guibbal
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Lyes Toualbi
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Fiona Routet
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Antoine Chaffiol
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Celine Winckler
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Marylin Harinquet
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Camille Robert
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Stephane Fouquet
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - José-Alain Sahel
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
- CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DGOS CIC 1423, Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Olivier Goureau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Jens Duebel
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Deniz Dalkara
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| |
Collapse
|
21
|
Maimon BE, Diaz M, Revol ECM, Schneider AM, Leaker B, Varela CE, Srinivasan S, Weber MB, Herr HM. Optogenetic Peripheral Nerve Immunogenicity. Sci Rep 2018; 8:14076. [PMID: 30232391 PMCID: PMC6145901 DOI: 10.1038/s41598-018-32075-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/28/2018] [Indexed: 12/20/2022] Open
Abstract
Optogenetic technologies have been the subject of great excitement within the scientific community for their ability to demystify complex neurophysiological pathways in the central (CNS) and peripheral nervous systems (PNS). The excitement surrounding optogenetics has also extended to the clinic with a trial for ChR2 in the treatment of retinitis pigmentosa currently underway and additional trials anticipated for the near future. In this work, we identify the cause of loss-of-expression in response to transdermal illumination of an optogenetically active peroneal nerve following an anterior compartment (AC) injection of AAV6-hSyn-ChR2(H134R) with and without a fluorescent reporter. Using Sprague Dawley Rag2-/- rats and appropriate controls, we discover optogenetic loss-of-expression is chiefly elicited by ChR2-mediated immunogenicity in the spinal cord, resulting in both CNS motor neuron death and ipsilateral muscle atrophy in both low and high Adeno-Associated Virus (AAV) dosages. We further employ pharmacological immunosuppression using a slow-release tacrolimus pellet to demonstrate sustained transdermal optogenetic expression up to 12 weeks. These results suggest that all dosages of AAV-mediated optogenetic expression within the PNS may be unsafe. Clinical optogenetics for both PNS and CNS applications should take extreme caution when employing opsins to treat disease and may require concurrent immunosuppression. Future work in optogenetics should focus on designing opsins with lesser immunogenicity.
Collapse
Affiliation(s)
- Benjamin E Maimon
- MIT Media Lab, Center for Extreme Bionics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences and Technology (HST), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Maurizio Diaz
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emilie C M Revol
- Department of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alexis M Schneider
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ben Leaker
- Harvard-MIT Division of Health Sciences and Technology (HST), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Claudia E Varela
- Harvard-MIT Division of Health Sciences and Technology (HST), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shriya Srinivasan
- MIT Media Lab, Center for Extreme Bionics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences and Technology (HST), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matthew B Weber
- MIT Media Lab, Center for Extreme Bionics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences and Technology (HST), Harvard Medical School, Boston, MA, USA
| | - Hugh M Herr
- MIT Media Lab, Center for Extreme Bionics, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
22
|
Watanabe Y, Sugano E, Tabata K, Ozaki T, Saito T, Tamai M, Tomita H. Kinetic profiles of photocurrents in cells expressing two types of channelrhodopsin genes. Biochem Biophys Res Commun 2018; 496:814-819. [PMID: 29395082 DOI: 10.1016/j.bbrc.2018.01.149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 01/24/2018] [Indexed: 12/21/2022]
Abstract
Channelrhodopsin-2 (ChR2), a light-activated cation-selective ion channel, has been widely used as a tool in optogenetic research. ChR2 is specifically sensitive to wavelengths less than 550 nm. One of the methods to expand the sensitivity of a channelrhodopsin to a wider range of wavelengths is to express another channelrhodopsin in the cells by the transduction of an additional gene. Here, we report the characteristic features of cells expressing two types of channelrhodopsins, each having different wavelength sensitivities. In HEK293 cells stably expressing ChR2, photocurrents were elicited at stimuli of 400-550 nm, and the wavelength sensitivity range was expanded by the additional transduction of the modified Volvox channelrhodopsin-1 (mVChR1) gene, which has broad wavelength sensitivities, ranging from 400 to 600 nm. However, the photocurrent at 550 nm was lower than that of the mVChR1-expressing cell; moreover, the turning-on and turning-off constants were delayed, and the deactivation rates were decreased. Meanwhile, the response to lower light intensity was improved by the additional gene. Thus, the transduction of an additional gene is a useful method to improve the light and wavelength sensitivities, as well as photocurrent kinetic profiles, of channelrhodopsins.
Collapse
Affiliation(s)
- Yoshito Watanabe
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan.
| | - Eriko Sugano
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan; Soft-Path Engineering Research Center (SPERC), Iwate University Division of Science and Engineering, Morioka, 020-8551, Japan.
| | - Kitako Tabata
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan.
| | - Taku Ozaki
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan; Soft-Path Engineering Research Center (SPERC), Iwate University Division of Science and Engineering, Morioka, 020-8551, Japan.
| | - Takehiko Saito
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan.
| | - Makoto Tamai
- Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aoba, Sendai, Miyagi, 980-8574, Japan.
| | - Hiroshi Tomita
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate, 020-8551, Japan; Soft-Path Engineering Research Center (SPERC), Iwate University Division of Science and Engineering, Morioka, 020-8551, Japan; Clinical Research, Innovation and Education Center, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai, Miyagi, 980-8574, Japan.
| |
Collapse
|
23
|
Cardiac Optogenetics: 2018. JACC Clin Electrophysiol 2018; 4:155-167. [PMID: 29749932 DOI: 10.1016/j.jacep.2017.12.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/04/2017] [Accepted: 12/14/2017] [Indexed: 01/28/2023]
Abstract
Cardiac optogenetics is an emergent research area involving the delivery of light-sensitive proteins (opsins) to excitable heart tissue to enable optical modulation of cardiac electrical function. Optogenetic stimulation has many noteworthy advantages over conventional electrical methods, including selective electrophysiological modulation in specifically targeted cell subpopulations, high-resolution spatiotemporal control via patterned illumination, and use of different opsins to elicit inward or outward transmembrane current. This review summarizes developments achieved since the inception of cardiac optogenetics research, which has spanned nearly a decade. The authors first provide an overview of recent methodological advances in opsin engineering, light sensitization of cardiac tissue, strategies for illuminating the heart, and frameworks for simulating optogenetics in realistic computational models of patient hearts. They then review recent cardiac optogenetics applications, including: 1) all-optical, high-throughput, contactless assays for quantification of electrophysiological properties; 2) optogenetic perturbation of cardiac tissue to unveil mechanistic insights on the initiation, perpetuation, and termination of arrhythmia; and 3) disruptive translational innovations such as light-based pacemaking and defibrillation.
Collapse
|
24
|
Chaffiol A, Caplette R, Jaillard C, Brazhnikova E, Desrosiers M, Dubus E, Duhamel L, Macé E, Marre O, Benoit P, Hantraye P, Bemelmans AP, Bamberg E, Duebel J, Sahel JA, Picaud S, Dalkara D. A New Promoter Allows Optogenetic Vision Restoration with Enhanced Sensitivity in Macaque Retina. Mol Ther 2017; 25:2546-2560. [PMID: 28807567 PMCID: PMC5675708 DOI: 10.1016/j.ymthe.2017.07.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/15/2017] [Accepted: 07/16/2017] [Indexed: 01/28/2023] Open
Abstract
The majority of inherited retinal degenerations converge on the phenotype of photoreceptor cell death. Second- and third-order neurons are spared in these diseases, making it possible to restore retinal light responses using optogenetics. Viral expression of channelrhodopsin in the third-order neurons under ubiquitous promoters was previously shown to restore visual function, albeit at light intensities above illumination safety thresholds. Here, we report (to our knowledge, for the first time) activation of macaque retinas, up to 6 months post-injection, using channelrhodopsin-Ca2+-permeable channelrhodopsin (CatCh) at safe light intensities. High-level CatCh expression was achieved due to a new promoter based on the regulatory region of the gamma-synuclein gene (SNCG) allowing strong expression in ganglion cells across species. Our promoter, in combination with clinically proven adeno-associated virus 2 (AAV2), provides CatCh expression in peri-foveolar ganglion cells responding robustly to light under the illumination safety thresholds for the human eye. On the contrary, the threshold of activation and the proportion of unresponsive cells were much higher when a ubiquitous promoter (cytomegalovirus [CMV]) was used to express CatCh. The results of our study suggest that the inclusion of optimized promoters is key in the path to clinical translation of optogenetics.
Collapse
Affiliation(s)
- Antoine Chaffiol
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France
| | - Romain Caplette
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France
| | - Céline Jaillard
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France
| | - Elena Brazhnikova
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France
| | - Mélissa Desrosiers
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France
| | - Elisabeth Dubus
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France
| | - Laëtitia Duhamel
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France
| | - Emilie Macé
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France
| | - Olivier Marre
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France
| | - Patrick Benoit
- Sanofi Ophthalmology Unit, 17 rue Moreau, 75012 Paris, France
| | - Philippe Hantraye
- Département des Sciences du Vivant (DSV), MIRCen, Institut d'Imagerie Biomédicale (I2BM), Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Fontenay-aux-Roses 92260, France; Neurodegenerative Diseases Laboratory, CNRS UMR9199, Université Paris-Sud, Université Paris-Saclay, Fontenay-aux-Roses 92260, France
| | - Alexis-Pierre Bemelmans
- Département des Sciences du Vivant (DSV), MIRCen, Institut d'Imagerie Biomédicale (I2BM), Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Fontenay-aux-Roses 92260, France; Neurodegenerative Diseases Laboratory, CNRS UMR9199, Université Paris-Sud, Université Paris-Saclay, Fontenay-aux-Roses 92260, France
| | - Ernst Bamberg
- Department of Biophysical Chemistry, Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Jens Duebel
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France.
| | - José-Alain Sahel
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France; CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC, 28 rue de Charenton, 75012 Paris, France; Fondation Ophtalmologique Adolphe de Rothschild, 75019 Paris, France.
| | - Serge Picaud
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France.
| | - Deniz Dalkara
- INSERM U968, Institut de la Vision, 75012 Paris, France; UMRS968, Institut de la Vision, Sorbonne Universités, Pierre et Marie Curie University (UPMC) University Paris 06, 75012 Paris, France; Centre National de la Recherche Scientifique (CNRS) UMR7210, Institut de la Vision, 75012 Paris, France.
| |
Collapse
|
25
|
Komatsu M, Sugano E, Tomita H, Fujii N. A Chronically Implantable Bidirectional Neural Interface for Non-human Primates. Front Neurosci 2017; 11:514. [PMID: 28966573 PMCID: PMC5605616 DOI: 10.3389/fnins.2017.00514] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 08/31/2017] [Indexed: 11/17/2022] Open
Abstract
Optogenetics has potential applications in the study of epilepsy and neuroprostheses, and for studies on neural circuit dynamics. However, to achieve translation to clinical usage, optogenetic interfaces that are capable of chronic stimulation and monitoring with minimal brain trauma are required. We aimed to develop a chronically implantable device for photostimulation of the brain of non-human primates. We used a micro-light-emitting diode (LED) array with a flexible polyimide film. The array was combined with a whole-cortex electrocorticographic (ECoG) electrode array for simultaneous photostimulation and recording. Channelrhodopsin-2 (ChR2) was virally transduced into the cerebral cortex of common marmosets, and then the device was epidurally implanted into their brains. We recorded the neural activity during photostimulation of the awake monkeys for 4 months. The neural responses gradually increased after the virus injection for ~8 weeks and remained constant for another 8 weeks. The micro-LED and ECoG arrays allowed semi-invasive simultaneous stimulation and recording during long-term implantation in the brains of non-human primates. The development of this device represents substantial progress in the field of optogenetic applications.
Collapse
Affiliation(s)
- Misako Komatsu
- Ichinohe Group, Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Brain Science InstituteSaitama, Japan
| | - Eriko Sugano
- Department of Chemistry and Biological Sciences, Iwate UniversityIwate, Japan
| | - Hiroshi Tomita
- Department of Chemistry and Biological Sciences, Iwate UniversityIwate, Japan
| | - Naotaka Fujii
- Laboratory for Adaptive Intelligence, RIKEN Brain Science InstituteSaitama, Japan
| |
Collapse
|
26
|
Long-term expression of melanopsin and channelrhodopsin causes no gross alterations in the dystrophic dog retina. Gene Ther 2017; 24:735-741. [PMID: 28880021 DOI: 10.1038/gt.2017.63] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/05/2017] [Accepted: 07/10/2017] [Indexed: 01/25/2023]
Abstract
Several preclinical studies have investigated the potential of algal channelrhodopsin and human melanopsin as optogenetic tools for vision restoration. In the present study, we assessed the potentially deleterious effects of long-term expression of these optogenes on the diseased retina in a large animal model of retinal degeneration, the RPE65-deficient Briard dog model of Leber congenital amaurosis. Intravitreal injection of adeno-associated virus vectors expressing channelrhodopsin and melanopsin had no effect on retinal thickness over a 16-month period post injection. Our data support the safety of the optogenetic approach for the treatment of blindness.
Collapse
|
27
|
Sato M, Sugano E, Tabata K, Sannohe K, Watanabe Y, Ozaki T, Tamai M, Tomita H. Visual Responses of Photoreceptor-Degenerated Rats Expressing Two Different Types of Channelrhodopsin Genes. Sci Rep 2017; 7:41210. [PMID: 28112267 PMCID: PMC5255552 DOI: 10.1038/srep41210] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/16/2016] [Indexed: 12/02/2022] Open
Abstract
Optogenetic technologies are expected to be applicable for clinical use in restoring vision. However, the degree of recovered visual function is highly dependent on the function of the chosen optogenetic gene. To investigate the effect on visual function of dual expression of genes with different wavelength sensitivities, we transduced a modified Volvox-derived channelrhodopsin gene (mVChR1) via an adeno-associated virus vector into transgenic rats harbouring the ChR2 gene in retinal ganglion cells. These transgenic rats were given an intraperitoneal injection of N-methyl-N-nitrosourea to induce the degeneration of native photoreceptor cells prior to transduction of mVChR1. Optical coherence tomography images indicated the degeneration of the native photoreceptor cells after the N-methyl-N-nitrosourea injection. Complete loss of function of the native photoreceptor cells was confirmed using electroretinograms. In the ChR2 transgenic rats, visually evoked potentials were clearly detectable in spite of native photoreceptor function abolishment; however the responses were limited to within blue wavelengths. In contrast, the limited wavelength sensitivities were improved by the additional transduction of mVChR1, which exhibited sensitivities to green and red. Thus, the transductions of dual genes encoding channelrhodopsins that exhibit different wavelength sensitivities represents a promising candidate method to expand and to enhance rescued wavelength sensitivities in blind subjects.
Collapse
Affiliation(s)
- Masatoshi Sato
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Eriko Sugano
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan.,Soft-Path Engineering Research Center (SPERC), Iwate University Division of Science and Engineering, Morioka 020-8551, Japan
| | - Kitako Tabata
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Kei Sannohe
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Yoshito Watanabe
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan
| | - Taku Ozaki
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan.,Soft-Path Engineering Research Center (SPERC), Iwate University Division of Science and Engineering, Morioka 020-8551, Japan
| | - Makoto Tamai
- Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aoba, Sendai, Miyagi 980-8574, Japan
| | - Hiroshi Tomita
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan.,Soft-Path Engineering Research Center (SPERC), Iwate University Division of Science and Engineering, Morioka 020-8551, Japan.,Clinical Research, Innovation and Education Center, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai, Miyagi 980-8574, Japan
| |
Collapse
|
28
|
Maybeck V, Schnitker J, Li W, Heuschkel M, Offenhäusser A. An evaluation of extracellular MEA versus optogenetic stimulation of cortical neurons. Biomed Phys Eng Express 2016. [DOI: 10.1088/2057-1976/2/5/055017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
29
|
Tabata K, Sugano E, Murakami F, Yamashita T, Ozaki T, Tomita H. Improved transduction efficiencies of adeno-associated virus vectors by synthetic cell-permeable peptides. Biochem Biophys Res Commun 2016; 478:1732-8. [PMID: 27614311 DOI: 10.1016/j.bbrc.2016.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 09/02/2016] [Indexed: 12/01/2022]
Abstract
Various serotypes of adeno-associated virus (AAV) vectors have been used for gene therapy and as research tools. Among these serotypes, the AAV type 2 vector has been used successfully in human gene therapies. However, the transduction efficiency of AAV2 depends on the cell type, and this poses a problem in the efficacy of gene therapy. To improve the transduction efficiency of AAV2, we designed a small peptide consisting of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor peptide and the HIV-Tat sequence Tat-Y1068. Pre- or co-treatment of CYNOM-K1 cells from cynomolgus monkey embryo skin with Tat-Y1068 increased the transduction efficiencies in a dose-dependent manner and caused p38 phosphorylation. The transduction efficiency of AAV2 into the rat fibroblast cell line RAT-1 highly expressing EGFR was less than the transduction efficiency of AAV2 into CYNOM-K1 cells. Tat-Y1068 increased the transduction efficiency in RAT-1 cells in the same manner as in CYNOM-K1 cells. In conclusion, cell-permeable peptides possessing the EGFR tyrosine kinase inhibitor function might serve as a useful ingredient of AAV2 vector solution for increasing the transduction efficiency of gene therapies.
Collapse
Affiliation(s)
- Kitako Tabata
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan.
| | - Eriko Sugano
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan; Soft-Path Engineering Research Center (SPERC), Faculty of Engineering, Iwate University, Morioka, 020-8551, Japan.
| | - Fumika Murakami
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan.
| | - Tetsuro Yamashita
- Soft-Path Engineering Research Center (SPERC), Faculty of Engineering, Iwate University, Morioka, 020-8551, Japan; Department of Biological Chemistry, Iwate University Faculty of Agriculture, Morioka, Japan.
| | - Taku Ozaki
- Soft-Path Engineering Research Center (SPERC), Faculty of Engineering, Iwate University, Morioka, 020-8551, Japan.
| | - Hiroshi Tomita
- Laboratory of Visual Neuroscience, Graduate Course in Biological Sciences, Iwate University Division of Science and Engineering, 4-3-5 Ueda, Morioka, Iwate 020-8551, Japan; Soft-Path Engineering Research Center (SPERC), Faculty of Engineering, Iwate University, Morioka, 020-8551, Japan; Clinical Research, Innovation and Education Center, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai, Miyagi 980-8574, Japan.
| |
Collapse
|
30
|
Klapper SD, Swiersy A, Bamberg E, Busskamp V. Biophysical Properties of Optogenetic Tools and Their Application for Vision Restoration Approaches. Front Syst Neurosci 2016; 10:74. [PMID: 27642278 PMCID: PMC5009148 DOI: 10.3389/fnsys.2016.00074] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/17/2016] [Indexed: 11/13/2022] Open
Abstract
Optogenetics is the use of genetically encoded light-activated proteins to manipulate cells in a minimally invasive way using light. The most prominent example is channelrhodopsin-2 (ChR2), which allows the activation of electrically excitable cells via light-dependent depolarization. The combination of ChR2 with hyperpolarizing-light-driven ion pumps such as the Cl(-) pump halorhodopsin (NpHR) enables multimodal remote control of neuronal cells in culture, tissue, and living animals. Very soon, it became obvious that this method offers a chance of gene therapy for many diseases affecting vision. Here, we will give a brief introduction to retinal function and retinal diseases; optogenetic vision restoration strategies will be highlighted. We will discuss the functional and structural properties of rhodopsin-based optogenetic tools and analyze the potential for the application of vision restoration.
Collapse
Affiliation(s)
- Simon D Klapper
- Center for Regenerative Therapies Dresden, Technische Universität Dresden Dresden, Germany
| | - Anka Swiersy
- Center for Regenerative Therapies Dresden, Technische Universität Dresden Dresden, Germany
| | - Ernst Bamberg
- Max Planck Institute of Biophysics Frankfurt, Germany
| | - Volker Busskamp
- Center for Regenerative Therapies Dresden, Technische Universität Dresden Dresden, Germany
| |
Collapse
|
31
|
Montgomery KL, Iyer SM, Christensen AJ, Deisseroth K, Delp SL. Beyond the brain: Optogenetic control in the spinal cord and peripheral nervous system. Sci Transl Med 2016; 8:337rv5. [DOI: 10.1126/scitranslmed.aad7577] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 04/18/2016] [Indexed: 12/12/2022]
|
32
|
Abstract
Severe loss of photoreceptor cells in inherited or acquired retinal degenerative diseases can result in partial loss of sight or complete blindness. The optogenetic strategy for restoration of vision utilizes optogenetic tools to convert surviving inner retinal neurons into photosensitive cells; thus, light sensitivity is imparted to the retina after the death of photoreceptor cells. Proof-of-concept studies, especially those using microbial rhodopsins, have demonstrated restoration of light responses in surviving retinal neurons and visually guided behaviors in animal models. Significant progress has also been made in improving microbial rhodopsin-based optogenetic tools, developing virus-mediated gene delivery, and targeting specific retinal neurons and subcellular compartments of retinal ganglion cells. In this article, we review the current status of the field and outline further directions and challenges to the advancement of this strategy toward clinical application and improvement in the outcomes of restored vision.
Collapse
Affiliation(s)
- Zhuo-Hua Pan
- Department of Ophthalmology, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan 48201; , , .,Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201;
| | - Qi Lu
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan 48201;
| | - Anding Bi
- Department of Ophthalmology, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan 48201; , ,
| | | | - Gary W Abrams
- Department of Ophthalmology, Kresge Eye Institute, Wayne State University School of Medicine, Detroit, Michigan 48201; , ,
| |
Collapse
|
33
|
Local and systemic responses following intravitreous injection of AAV2-encoded modified Volvox channelrhodopsin-1 in a genetically blind rat model. Gene Ther 2015; 23:158-66. [PMID: 26440056 DOI: 10.1038/gt.2015.99] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/11/2015] [Accepted: 09/04/2015] [Indexed: 01/22/2023]
Abstract
We previously designed a modified channelrhodopsin-1 (mVChR1) protein chimera with a broader action than that of Chlamydomonas channelrhodopsin-2 and reported that its transduction into retinal ganglion cells can restore visual function in genetically blind, dystrophic Royal College of Surgeons (RCS) rats, with photostimuli ranging from 486 to 640 nm. In the current study, we sought to investigate the safety and influence of mVChR1 transgene expression. Adeno-associated virus type 2 encoding mVChR1 was administered by intravitreous injection into dystrophic RCS rats. Reverse-transcription PCR was used to monitor virus and transgene dissemination and the results demonstrated that their expression was restricted specifically within the eye tissues, and not in non-target organs. Moreover, examination of the blood, plasma and serum revealed that no excess immunoreactivity was present, as determined using standard clinical hematological parameters. Serum antibodies targeting the recombinant adeno-associated virus (rAAV) capsid increased after the injection; however, no increase in mVChR1 antibody was detected during the observation period. In addition, retinal histological examination showed no signs of inflammation in rAAV-injected rats. In conclusion, our results demonstrate that mVChR1 can be exogenously expressed without harmful immunological reactions in vivo. These findings will aid in studies of AAV gene transfer to restore vision in late-stage retinitis pigmentosa.
Collapse
|
34
|
BARRETT JOHNMARTIN, BERLINGUER-PALMINI ROLANDO, DEGENAAR PATRICK. Optogenetic approaches to retinal prosthesis. Vis Neurosci 2014; 31:345-54. [PMID: 25100257 PMCID: PMC4161214 DOI: 10.1017/s0952523814000212] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 05/07/2014] [Indexed: 01/14/2023]
Abstract
The concept of visual restoration via retinal prosthesis arguably started in 1992 with the discovery that some of the retinal cells were still intact in those with the retinitis pigmentosa disease. Two decades later, the first commercially available devices have the capability to allow users to identify basic shapes. Such devices are still very far from returning vision beyond the legal blindness. Thus, there is considerable continued development of electrode materials, and structures and electronic control mechanisms to increase both resolution and contrast. In parallel, the field of optogenetics--the genetic photosensitization of neural tissue holds particular promise for new approaches. Given that the eye is transparent, photosensitizing remaining neural layers of the eye and illuminating from the outside could prove to be less invasive, cheaper, and more effective than present approaches. As we move toward human trials in the coming years, this review explores the core technological and biological challenges related to the gene therapy and the high radiance optical stimulation requirement.
Collapse
Affiliation(s)
- JOHN MARTIN BARRETT
- Institute of Neuroscience,
Newcastle University, Newcastle upon
Tyne, United Kingdom
| | | | - PATRICK DEGENAAR
- School of EEE,
Newcastle University, Newcastle upon
Tyne, United Kingdom
| |
Collapse
|
35
|
Tomita H, Sugano E, Murayama N, Ozaki T, Nishiyama F, Tabata K, Takahashi M, Saito T, Tamai M. Restoration of the majority of the visual spectrum by using modified Volvox channelrhodopsin-1. Mol Ther 2014; 22:1434-1440. [PMID: 24821344 PMCID: PMC4435592 DOI: 10.1038/mt.2014.81] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 04/29/2014] [Indexed: 11/12/2022] Open
Abstract
We previously showed that blind rats whose vision was restored by gene transfer of Chlamydomonas channelrhodopsin-2 (ChR2) could only detect wavelengths less than 540 nm because of the action spectrum of the transgene product. Volvox-derived channelrhodopsin-1, VChR1, has a broader spectrum than ChR2. However, the VChR1 protein was mainly localized in the cytoplasm and showed weak ion channel properties when the VChR1 gene was transfected into HEK293 cells. We generated modified Volvox channelrhodopsin-1 (mVChR1), which is a chimera of Volvox channelrhodopsin-1 and Chlamydomonas channelrhodopsin-1 and demonstrated increased plasma membrane integration and dramatic improvement in its channel properties. Under whole-cell patch clamp, mVChR1-expressing cells showed a photo-induced current upon stimulation at 468–640 nm. The evoked currents in mVChR1-expressing cells were ~30 times larger than those in VChR1-expressing cells. Genetically, blind rats expressing mVChR1 via an adeno-associated virus vector regained their visual responses to light with wavelengths between 468 and 640 nm and their recovered visual responses were maintained for a year. Thus, mVChR1 is a candidate gene for gene therapy for restoring vision, and gene delivery of mVChR1 may provide blind patients access to the majority of the visible light spectrum.
Collapse
Affiliation(s)
- Hiroshi Tomita
- Laboratory of Visual Neuroscience, Department of Chemistry and Bioengineering, Iwate University Graduate School of Engineering, Morioka, Iwate, Japan; Clinical Research, Innovation and Education Center, Tohoku University Hospital, Sendai, Miyagi, Japan.
| | - Eriko Sugano
- Laboratory of Visual Neuroscience, Department of Chemistry and Bioengineering, Iwate University Graduate School of Engineering, Morioka, Iwate, Japan
| | - Namie Murayama
- Laboratory of Visual Neuroscience, Department of Chemistry and Bioengineering, Iwate University Graduate School of Engineering, Morioka, Iwate, Japan
| | - Taku Ozaki
- Department of Ophthalmology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Fumiaki Nishiyama
- Laboratory of Visual Neuroscience, Department of Chemistry and Bioengineering, Iwate University Graduate School of Engineering, Morioka, Iwate, Japan
| | - Kitako Tabata
- Laboratory of Visual Neuroscience, Department of Chemistry and Bioengineering, Iwate University Graduate School of Engineering, Morioka, Iwate, Japan
| | - Maki Takahashi
- Laboratory of Visual Neuroscience, Department of Chemistry and Bioengineering, Iwate University Graduate School of Engineering, Morioka, Iwate, Japan
| | - Takehiko Saito
- Laboratory of Visual Neuroscience, Department of Chemistry and Bioengineering, Iwate University Graduate School of Engineering, Morioka, Iwate, Japan
| | - Makoto Tamai
- Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
36
|
Francis PJ, Mansfield B, Rose S. Proceedings of the First International Optogenetic Therapies for Vision Symposium. Transl Vis Sci Technol 2013; 2:4. [PMID: 24349882 DOI: 10.1167/tvst.2.7.4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 10/17/2013] [Indexed: 11/24/2022] Open
Abstract
Optogenetics is a research field that uses gene therapy to deliver a gene encoding a light-activated protein to cells providing light-regulated control of targeted cell pathways. The technology is a popular tool in many fields of neuroscience, used to transiently switch cells on and off, for example, to map neural circuits. In inherited retinal degenerative diseases, where loss of vision results from the loss of photoreceptors, optogenetics can be applied to either augment the function of surviving photoreceptors or confer light sensitivity to naturally nonlight sensitive retinal cells, such as a bipolar cells. This can be achieved either by the light sensitive protein integrating with native internal signaling pathways, or by using a dual function membrane protein that integrates light signaling with an ion channel or pump activity. Exposing treated cells to light of the correct wavelength activates the protein, resulting in cellular depolarization or hyperpolarization that triggers neurological signaling to the visual cortex. While there is a lot of interest in optogenetics as a pan-disease clinical treatment for end-stage application in the inherited degenerative diseases of the retina, research to date has been limited to nonhuman clinical studies. To address the clinical translational needs of this technology, the Foundation Fighting Blindness and Massachusetts Eye and Ear Infirmary cohosted an International Optogenetic Therapies for Vision Workshop, which was held at Massachusetts Eye and Ear Infirmary, Boston, Massachusetts on June 1, 2012.
Collapse
|
37
|
Isago H, Sugano E, Murayama N, Tamai M, Tomita H. Establishment of monocular-limited photoreceptor degeneration models in rabbits. BMC Ophthalmol 2013; 13:19. [PMID: 23683117 PMCID: PMC3679785 DOI: 10.1186/1471-2415-13-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 05/15/2013] [Indexed: 11/24/2022] Open
Abstract
Background Numerous rodent models of photoreceptor degeneration have been developed for the study of visual function. However, no viable model has been established in a species that is more closely related to Homo sapiens. Here, we present a rabbit model of monocular photoreceptor degeneration. Methods We tested 2 chemicals, verteporfin and sodium nitroprusside (SNP), for developing a 1-eye limited photoreceptor degeneration model in pigmented rabbits. After the intravenous injection of verteporfin, the retina was exposed to light from a halogen lamp for 0, 10, 30, or 60 min. Alternately, 100 μL of various concentrations of sodium nitroprusside (0.1 mM, 0.5 mM, and 1 mM) were intravitreously injected into the rabbit eye. Retinal degeneration was evaluated by fundus photography, electroretinogram (ERG), and histological examinations. Results Fundus photographs of animals in the verteporfin- or SNP-treated groups showed evidence of retinal degeneration. The severity of this degradation depended on the duration of light exposure and the concentration of SNP administered. The degeneration was clearly limited to the light-exposed areas in the verteporfin-treated groups. Extensive retinal atrophy was observed in the SNP-treated groups. The a- and b-wave amplitudes were dramatically decreased on the ERGs from SNP-treated groups. Histological examination revealed that either verteporfin or SNP induced severe photoreceptor degeneration. High-dose SNP treatment (1 mM) was also associated with inner retinal layer degeneration. Conclusions Both SNP and verteporfin clearly caused photoreceptor degeneration without any effect on the contralateral eye. These compounds therefore represent valuable tools for the empirical investigation of visual function recovery. The findings will inform guidelines for clinical applications such as retinal prostheses, cell-based therapy, and gene therapy.
Collapse
Affiliation(s)
- Hitomi Isago
- Laboratory of Visual Neuroscience, Department of Chemistry and Bioengineering, Iwate University Graduate School of Engineering, 4-3-5 Ueda Morioka, Iwate 020-8551, Japan
| | | | | | | | | |
Collapse
|
38
|
G N, Tan A, Farhatnia Y, Rajadas J, Hamblin MR, Khaw PT, Seifalian AM. Channelrhodopsins: visual regeneration and neural activation by a light switch. N Biotechnol 2013; 30:461-74. [PMID: 23664865 DOI: 10.1016/j.nbt.2013.04.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/28/2013] [Accepted: 04/16/2013] [Indexed: 01/09/2023]
Abstract
The advent of optogenetics provides a new direction for the field of neuroscience and biotechnology, serving both as a refined investigative tool and as potential cure for many medical conditions via genetic manipulation. Although still in its infancy, recent advances in optogenetics has made it possible to remotely manipulate in vivo cellular functions using light. Coined Nature Methods' 'Method of the Year' in 2010, the optogenetic toolbox has the potential to control cell, tissue and even animal behaviour. This optogenetic toolbox consists of light-sensitive proteins that are able to modulate membrane potential in response to light. Channelrhodopsins (ChR) are light-gated microbial ion channels, which were first described in green algae. ChR2 (a subset of ChR) is a seven transmembrane α helix protein, which evokes membrane depolarization and mediates an action potential upon photostimulation with blue (470 nm) light. By contrast to other seven-transmembrane proteins that require second messengers to open ion channels, ChR2 form ion channels themselves, allowing ultrafast depolarization (within 50 milliseconds of illumination). It has been shown that integration of ChR2 into various tissues of mice can activate neural circuits, control heart muscle contractions, and even restore breathing after spinal cord injury. More compellingly, a plethora of evidence has indicated that artificial expression of ChR2 in retinal ganglion cells can reinstate visual perception in mice with retinal degeneration.
Collapse
Affiliation(s)
- Natasha G
- Centre for Nanotechnology & Regenerative Medicine, UCL Division of Surgery & Interventional Science, University College London, London, UK
| | | | | | | | | | | | | |
Collapse
|
39
|
Optogenetically induced seizure and the longitudinal hippocampal network dynamics. PLoS One 2013; 8:e60928. [PMID: 23593349 PMCID: PMC3622611 DOI: 10.1371/journal.pone.0060928] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Accepted: 03/04/2013] [Indexed: 11/19/2022] Open
Abstract
Epileptic seizure is a paroxysmal and self-limited phenomenon characterized by abnormal hypersynchrony of a large population of neurons. However, our current understanding of seizure dynamics is still limited. Here we propose a novel in vivo model of seizure-like afterdischarges using optogenetics, and report on investigation of directional network dynamics during seizure along the septo-temporal (ST) axis of hippocampus. Repetitive pulse photostimulation was applied to the rodent hippocampus, in which channelrhodopsin-2 (ChR2) was expressed, under simultaneous recording of local field potentials (LFPs). Seizure-like afterdischarges were successfully induced after the stimulation in both W-TChR2V4 transgenic (ChR2V-TG) rats and in wild type rats transfected with adeno-associated virus (AAV) vectors carrying ChR2. Pulse frequency at 10 and 20 Hz, and a 0.05 duty ratio were optimal for afterdischarge induction. Immunohistochemical c-Fos staining after a single induced afterdischarge confirmed neuronal activation of the entire hippocampus. LFPs were recorded during seizure-like afterdischarges with a multi-contact array electrode inserted along the ST axis of hippocampus. Granger causality analysis of the LFPs showed a bidirectional but asymmetric increase in signal flow along the ST direction. State space presentation of the causality and coherence revealed three discrete states of the seizure-like afterdischarge phenomenon: 1) resting state; 2) afterdischarge initiation with moderate coherence and dominant septal-to-temporal causality; and 3) afterdischarge termination with increased coherence and dominant temporal-to-septal causality. A novel in vivo model of seizure-like afterdischarge was developed using optogenetics, which was advantageous in its reproducibility and artifact-free electrophysiological observations. Our results provide additional evidence for the potential role of hippocampal septo-temporal interactions in seizure dynamics in vivo. Bidirectional networks work hierarchically along the ST hippocampus in the genesis and termination of epileptic seizures.
Collapse
|
40
|
Channelrhodopsins-Their potential in gene therapy for neurological disorders. Neurosci Res 2012; 75:6-12. [PMID: 23026479 DOI: 10.1016/j.neures.2012.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 08/18/2012] [Accepted: 09/10/2012] [Indexed: 11/23/2022]
Abstract
Recently, channelrhodopsins (ChRs) have begun to be used to manipulate the neuronal activity, since they can be targeted to specific neurons or neural circuits using genetic methods. To advance the potential applications in the investigation and treatment of neurological disorders, the following types of basic research should receive extensive financial support. The spectral and kinetic properties of ChRs should be optimized according to the application by generating variants of ChRs or exploring new rhodopsins from other species. These ChRs should be targeted to the specific types of neurons involved in the neurological disorders through a gene expression system using cell- or tissue-specific promoters/enhancers as well as gene delivery systems with modified virus vectors. The methods have to be developed to apply the genes of interest with safety and long-term effectiveness. Sophisticated opto-electrical devices should be developed. Appropriate primate animal model systems should be established to minimize the structural differences between small animals such as rodents and human beings. In this paper, we will review the current progress in the basic research concerned with the potential clinical application of ChRs and discuss the future directions of research on ChRs so that they could be applied for human welfare.
Collapse
|
41
|
Yao JP, Hou WS, Yin ZQ. Optogenetics: a novel optical manipulation tool for medical investigation. Int J Ophthalmol 2012; 5:517-22. [PMID: 22937517 DOI: 10.3980/j.issn.2222-3959.2012.04.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 07/25/2012] [Indexed: 11/02/2022] Open
Abstract
Optogenetics is a new and rapidly evolving gene and neuroengineering technology that allows optical control of specific populations of neurons without affecting other neurons in the brain at high temporal and spatial resolution. By heterologous expression of the light-sensitive membrane proteins, cell type-specific depolarization or hyperpolarization can be optically induced on a millisecond time scale. Optogenetics has the higher selectivity and specificity compared to traditional electrophysiological techniques and pharmaceutical methods. It has been a novel promising tool for medical research. Because of easy handling, high temporal and spatial precision, optogenetics has been applied to many aspects of nervous system research, such as tactual neural circuit, visual neural circuit, auditory neural circuit and olfactory neural circuit, as well as research of some neurological diseases. The review highlights the recent advances of optogenetics in medical study.
Collapse
Affiliation(s)
- Jun-Ping Yao
- College of Biology Engineering, Chongqing University, Chongqing 400044, China
| | | | | |
Collapse
|
42
|
Yao J, Hou W, Wang H, Liu H, Weng C, Yin Z. Optical control after transfection of channelrhodopsin-2 recombinant adenovirus in visual cortical cells. Neural Regen Res 2012; 7:1228-33. [PMID: 25709620 PMCID: PMC4336956 DOI: 10.3969/j.issn.1673-5374.2012.16.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 05/03/2012] [Indexed: 11/18/2022] Open
Abstract
Channelrhodopsin-2 ectopically expressed in the retina can recover the response to blue light in genetically blind mice and rats, but is unable to restore visual function due to optic nerve or optic tract lesions. Long Evans rats at postnatal day 1 were used for primary culture of visual cortical cells, and 24 hours later, cells were transfected with recombinant adenovirus carrying channelrhodopsin-2 and green fluorescent protein genes. After 2-4 days of transfection, green fluorescence was visible in the cultured cells. Cells were stimulated with blue light (470 nm), and light-induced action potentials were recorded in patch-clamp experiments. Our findings indicate that channelrhodopsin-2-recombinant adenovirus transfection of primary cultured visual cortical cells can control the production of action potentials via blue light stimulation.
Collapse
Affiliation(s)
- Junping Yao
- College of Bioengineering, Chongqing University, Chongqing 400044, China ; Chongqing Key Lab of Visual Damage and Regeneration & Restoration, Chongqing 400038, China
| | - Wensheng Hou
- College of Bioengineering, Chongqing University, Chongqing 400044, China ; Chongqing Key Lab of Visual Damage and Regeneration & Restoration, Chongqing 400038, China
| | - Hao Wang
- Department of Ophthalmology, Southwest Hospital, Third Military Medical University of Chinese PLA, Chongqing 400038, China
| | - Hui Liu
- Department of Ophthalmology, Southwest Hospital, Third Military Medical University of Chinese PLA, Chongqing 400038, China
| | - Chuanhuang Weng
- Department of Ophthalmology, Southwest Hospital, Third Military Medical University of Chinese PLA, Chongqing 400038, China
| | - Zhengqin Yin
- Department of Ophthalmology, Southwest Hospital, Third Military Medical University of Chinese PLA, Chongqing 400038, China
| |
Collapse
|
43
|
Vugler AA, Semo M, Coffey PJ. Seeing again through ancient eyes: microbial opsins and the promise of restoring vision. EXPERT REVIEW OF OPHTHALMOLOGY 2011. [DOI: 10.1586/eop.11.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
44
|
Isago H, Sugano E, Wang Z, Murayama N, Koyanagi E, Tamai M, Tomita H. Age-dependent differences in recovered visual responses in Royal College of Surgeons rats transduced with the Channelrhodopsin-2 gene. J Mol Neurosci 2011; 46:393-400. [PMID: 21792608 DOI: 10.1007/s12031-011-9599-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 07/12/2011] [Indexed: 11/26/2022]
Abstract
The objective of this study is to investigate age-related differences in recovered visual function in Royal College of Surgeons (RCS) rats transduced with the Channelrhodopsin-2 (ChR2) gene. An adeno-associated virus vector that contained ChR2 was injected intravitreously into young or aged RCS rats. After 4 months, visual evoked potentials were recorded. To estimate the transduction efficiencies, ChR2V-expressing cells and retrograde labeled retinal ganglion cells (RGCs) were counted. After photoreceptor degradation, immunohistochemistry was used to detect glial fibrillary acidic protein (GFAP) in the retinas. The amplitudes and latencies from young RCS rats were higher and shorter, respectively, than those from aged RCS rats. ChR2V was expressed in the RGCs of both groups of rats; there was no significant difference in the transduction efficiency of either group. However, the number of RGCs in aged RCS rats was significantly less than that in young RCS rats. In addition, strong GFAP immunoreactivity was observed after photoreceptor degeneration, whereas it was weaker in ChR2V-expressing RGCs. ChR2 transduction produced photosensitive RGCs in both young and aged rats. However, the degree of recovery depended on the age at the time of transduction.
Collapse
Affiliation(s)
- Hitomi Isago
- Institute for International Advanced Interdisciplinary Research, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Busskamp V, Roska B. Optogenetic approaches to restoring visual function in retinitis pigmentosa. Curr Opin Neurobiol 2011; 21:942-6. [PMID: 21708457 DOI: 10.1016/j.conb.2011.06.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 06/03/2011] [Indexed: 12/15/2022]
Abstract
Retinitis pigmentosa is a hereditary eye disease that affects photoreceptors and leads to blindness. The discovery of a microbial light-gated channel and the subsequent development of similar 'optogenetic' sensors have opened the door to creating artificial photoreceptors in the remaining retinal circuits of retinitis pigmentosa retinas via gene therapy. Here we review recent studies in animal models of retinitis pigmentosa that have combined knowledge of retinal cell types, circuits and computations with the ability to equip cell types with optogenetic sensors in order to restore visual activity. We also discuss the translational potential of this therapy.
Collapse
Affiliation(s)
- Volker Busskamp
- Neural Circuit Laboratories, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | | |
Collapse
|