1
|
Andrabi M, Upton BA, Lang RA, Vemaraju S. An Expanding Role for Nonvisual Opsins in Extraocular Light Sensing Physiology. Annu Rev Vis Sci 2023; 9:245-267. [PMID: 37196422 DOI: 10.1146/annurev-vision-100820-094018] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
We live on a planet that is bathed in daily and seasonal sunlight cycles. In this context, terrestrial life forms have evolved mechanisms that directly harness light energy (plants) or decode light information for adaptive advantage. In animals, the main light sensors are a family of G protein-coupled receptors called opsins. Opsin function is best described for the visual sense. However, most animals also use opsins for extraocular light sensing for seasonal behavior and camouflage. While it has long been believed that mammals do not have an extraocular light sensing capacity, recent evidence suggests otherwise. Notably, encephalopsin (OPN3) and neuropsin (OPN5) are both known to mediate extraocular light sensing in mice. Examples of this mediation include photoentrainment of circadian clocks in skin (by OPN5) and acute light-dependent regulation of metabolic pathways (by OPN3 and OPN5). This review summarizes current findings in the expanding field of extraocular photoreception and their relevance for human physiology.
Collapse
Affiliation(s)
- Mutahar Andrabi
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; ,
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Brian A Upton
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; ,
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Molecular and Developmental Biology Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- Medical Scientist Training Program, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Richard A Lang
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; ,
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Shruti Vemaraju
- The Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; ,
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
2
|
Katada Y, Kunimi H, Serizawa N, Lee D, Kobayashi K, Negishi K, Okano H, Tanaka KF, Tsubota K, Kurihara T. Starburst amacrine cells amplify optogenetic visual restoration through gap junctions. Mol Ther Methods Clin Dev 2023; 30:1-13. [PMID: 37324975 PMCID: PMC10265492 DOI: 10.1016/j.omtm.2023.05.011] [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: 07/22/2022] [Accepted: 05/09/2023] [Indexed: 06/17/2023]
Abstract
Ectopic induction of optogenetic actuators, such as channelrhodopsin, is a promising approach to restoring vision in the degenerating retina. However, the cell type-specific response of ectopic photoreception has not been well understood. There are limits to obtaining efficient gene expression in a specifically targeted cell population by a transgenic approach. In the present study, we established a murine model with high efficiency of gene induction to retinal ganglion cells (RGCs) and amacrine cells using an improved tetracycline transactivator-operator bipartite system (KENGE-tet system). To investigate the cell type-specific visual restorative effect, we expressed the channelrhodopsin gene into RGCs and amacrine cells using the KENGE-tet system. As a result, enhancement in the visual restorative effect was observed to RGCs and starburst amacrine cells. In conclusion, a photoresponse from amacrine cells may enhance the maintained response of RGCs and further increase or improve the visual restorative effect.
Collapse
Affiliation(s)
- Yusaku Katada
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiromitsu Kunimi
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Naho Serizawa
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, 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, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, 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, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kenji F. Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazuo Tsubota
- Tsubota Laboratory, Inc, Shinjuku-ku, Tokyo 160-0016, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
3
|
Berry MH, Leffler J, Allen CN, Sivyer B. Functional subtypes of rodent melanopsin ganglion cells switch roles between night and day illumination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.26.554902. [PMID: 38168436 PMCID: PMC10760181 DOI: 10.1101/2023.08.26.554902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs), contain the photopigment melanopsin, and influence both image and non-image forming behaviors. Despite being categorized into multiple types (M1-M6), physiological variability within these types suggests our current understanding of ipRGCs is incomplete. We used multi-electrode array (MEA) recordings and unbiased cluster analysis under synaptic blockade to identify 8 functional clusters of ipRGCs, each with distinct photosensitivity and response timing. We used Cre mice to drive the expression of channelrhodopsin in SON-ipRGCs, enabling the localization of distinct ipRGCs in the dorsal retina. Additionally, we conducted a retrospective unbiased cluster analysis of ipRGC photoresponses to light stimuli across scotopic, mesopic, and photopic intensities, aimed at activating both rod and cone inputs to ipRGCs. Our results revealed shared and distinct synaptic inputs to the identified functional clusters, demonstrating that ipRGCs encode visual information with high fidelity at low light intensities, but poorly at photopic light intensities, when melanopsin activation is highest. Collectively, our findings support a framework with at least 8 functional subtypes of ipRGCs, each encoding luminance with distinct spike outputs, highlighting the inherent functional diversity and complexity of ipRGCs and suggesting a reevaluation of their contributions to retinal function and visual perception under varying light conditions.
Collapse
Affiliation(s)
- Michael H. Berry
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
- Medical Scientist Training program, Oregon Health & Science University, Portland, OR, 97239
| | - Joseph Leffler
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
| | - Charles N. Allen
- Oregon Institute of Occupational Health Sciences, Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239
| | - Benjamin Sivyer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239
| |
Collapse
|
4
|
Emanuel AJ, Do MTH. The Multistable Melanopsins of Mammals. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1174255. [PMID: 37994345 PMCID: PMC10664805 DOI: 10.3389/fopht.2023.1174255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 04/10/2023] [Indexed: 11/24/2023]
Abstract
Melanopsin is a light-activated G protein coupled receptor that is expressed widely across phylogeny. In mammals, melanopsin is found in intrinsically photosensitive retinal ganglion cells (ipRGCs), which are especially important for "non-image" visual functions that include the regulation of circadian rhythms, sleep, and mood. Photochemical and electrophysiological experiments have provided evidence that melanopsin has at least two stable conformations and is thus multistable, unlike the monostable photopigments of the classic rod and cone photoreceptors. Estimates of melanopsin's properties vary, challenging efforts to understand how the molecule influences vision. This article seeks to reconcile disparate views of melanopsin and offer a practical guide to melanopsin's complexities.
Collapse
Affiliation(s)
| | - Michael Tri H. Do
- F.M. Kirby Neurobiology Center and Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
5
|
Fu Y, Liu S, Dong Y, Gan Y, Guo X, Liu H, Xu Q, Yuan R, Ning A, Hong W, Peng Y, Yu S. Chronic restraint stress-induced depression-like behavior is mediated by upregulation of melanopsin expression in C57BL/6 mice retina. Psychopharmacology (Berl) 2023; 240:283-293. [PMID: 36580134 DOI: 10.1007/s00213-022-06302-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Depression is associated with circadian disturbances in which melanopsin was a key mechanism. Further studies have demonstrated that melanopsin gene variations are associated with some depressive disorders and aberrant light can impair mood through melanopsin-expressing retinal ganglion cells (mRGCs). The goal of this study was to explore the direct relationship between depression and melanopsin. METHODS Adult C57BL/6 male mice were physically restrained for 16 h in a 50-ml polypropylene centrifuge tube and all behavioral tests were performed after CRS treatment. Western blot analysis and immunofluorescence were used to detect melanopsin expression in the retina of C57BL/6 mice. And we observed the change of the electrophysiological function and release of glutamate of mRGCs. RESULTS The melanopsin expression upregulate in mRGCs of chronic restraint stress (CRS)-treating mice which exhibit depression-like behavior. The frequency of blue light-induced action potentials and light-induced glutamate release mediated by melanopsin also increase significantly. This change of melanopsin is mediated by the CRS-induced glucocorticoid. CONCLUSIONS CRS may induce the depression-like behavior in mice via glucocorticoid-melanopsin pathway. Our findings provide a novel mechanistic link between CRS-induced depression and melanopsin in mice.
Collapse
Affiliation(s)
- Yingmei Fu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Shanshan Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing, China
| | - Yigang Dong
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, School of Physical Education & Health Care, East China Normal University, Shanghai, 200241, China
| | - Yixia Gan
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, School of Physical Education & Health Care, East China Normal University, Shanghai, 200241, China
| | - Xiaoyun Guo
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongmei Liu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingqing Xu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixue Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ailing Ning
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wu Hong
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanmin Peng
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Shunying Yu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Nikonov S, Dolgova N, Sudharsan R, Tochitsky I, Iwabe S, Guzman JM, Van Gelder RN, Kramer RH, Aguirre GD, Beltran WA. Photochemical Restoration of Light Sensitivity in the Degenerated Canine Retina. Pharmaceutics 2022; 14:pharmaceutics14122711. [PMID: 36559205 PMCID: PMC9783220 DOI: 10.3390/pharmaceutics14122711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/11/2022] Open
Abstract
Photopharmacological compounds such as azobenzene-based photoswitches have been shown to control the conductivity of ionic channels in a light-dependent manner and are considered a potential strategy to restore vision in patients with end-stage photoreceptor degeneration. Here, we report the effects of DENAQ, a second-generation azobenzene-based photoswitch on retinal ganglion cells (RGC) in canine retinas using multi-electrode array (MEA) recordings (from nine degenerated and six WT retinas). DENAQ treatment conferred increased light sensitivity to RGCs in degenerated canine retinas. RGC light responses were observed in degenerated retinas following ex vivo application of 1 mM DENAQ (n = 6) or after in vivo DENAQ injection (n = 3, 150 μL, 3-10 mM) using 455 nm light at intensities as low as 0.2 mW/cm2. The number of light-sensitive cells and the per cell response amplitude increased with light intensity up to the maximum tested intensity of 85 mW/cm2. Application of DENAQ to degenerated retinas with partially preserved cone function caused appearance of DENAQ-driven responses both in cone-driven and previously non-responsive RGCs, and disappearance of cone-driven responses. Repeated stimulation slowed activation and accelerated recovery of the DENAQ-driven responses. The latter is likely responsible for the delayed appearance of a response to 4 Hz flicker stimulation. Limited aqueous solubility of DENAQ results in focal drug aggregates associated with ocular toxicity. While this limits the therapeutic potential of DENAQ, more potent third-generation photoswitches may be more promising, especially when delivered in a slow-release formulation that prevents drug aggregation.
Collapse
Affiliation(s)
- Sergei Nikonov
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Natalia Dolgova
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raghavi Sudharsan
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ivan Tochitsky
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 90095, USA
| | - Simone Iwabe
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jose-Manuel Guzman
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Russell N. Van Gelder
- Department of Ophthalmology, Pathology, and Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Richard H. Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 90095, USA
| | - Gustavo D. Aguirre
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William A. Beltran
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence:
| |
Collapse
|
7
|
Takahashi TM, Hirano A, Kanda T, Saito VM, Ashitomi H, Tanaka KZ, Yokoshiki Y, Masuda K, Yanagisawa M, Vogt KE, Tokuda T, Sakurai T. Optogenetic induction of hibernation-like state with modified human Opsin4 in mice. CELL REPORTS METHODS 2022; 2:100336. [PMID: 36452866 PMCID: PMC9701604 DOI: 10.1016/j.crmeth.2022.100336] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 09/01/2022] [Accepted: 10/19/2022] [Indexed: 05/28/2023]
Abstract
We recently determined that the excitatory manipulation of Qrfp-expressing neurons in the preoptic area of the hypothalamus (quiescence-inducing neurons [Q neurons]) induced a hibernation-like hypothermic/hypometabolic state (QIH) in mice. To control the QIH with a higher time resolution, we develop an optogenetic method using modified human opsin4 (OPN4; also known as melanopsin), a G protein-coupled-receptor-type blue-light photoreceptor. C-terminally truncated OPN4 (OPN4dC) stably and reproducibly induces QIH for at least 24 h by illumination with low-power light (3 μW, 473 nm laser) with high temporal resolution. The high sensitivity of OPN4dC allows us to transcranially stimulate Q neurons with blue-light-emitting diodes and non-invasively induce the QIH. OPN4dC-mediated QIH recapitulates the kinetics of the physiological changes observed in natural hibernation, revealing that Q neurons concurrently contribute to thermoregulation and cardiovascular function. This optogenetic method may facilitate identification of the neural mechanisms underlying long-term dormancy states such as sleep, daily torpor, and hibernation.
Collapse
Affiliation(s)
- Tohru M. Takahashi
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- International Integrative Institute for Sleep medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Arisa Hirano
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- International Integrative Institute for Sleep medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- JST PRESTO, Japan
| | - Takeshi Kanda
- International Integrative Institute for Sleep medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Viviane M. Saito
- Memory Research Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa, Japan
| | - Hiroto Ashitomi
- Memory Research Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa, Japan
| | - Kazumasa Z. Tanaka
- Memory Research Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa, Japan
| | - Yasufumi Yokoshiki
- Institute of Innovative Research (IIR), Tokyo Institute of Technology, Tokyo, Japan
| | - Kosaku Masuda
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- International Integrative Institute for Sleep medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Masashi Yanagisawa
- International Integrative Institute for Sleep medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Kaspar E. Vogt
- International Integrative Institute for Sleep medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Takashi Tokuda
- JST PRESTO, Japan
- Institute of Innovative Research (IIR), Tokyo Institute of Technology, Tokyo, Japan
| | - Takeshi Sakurai
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- International Integrative Institute for Sleep medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
8
|
Taghert PH. The incidence of candidate binding sites for β-arrestin in Drosophila neuropeptide GPCRs. PLoS One 2022; 17:e0275410. [PMID: 36318573 PMCID: PMC9624432 DOI: 10.1371/journal.pone.0275410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/14/2022] [Indexed: 01/24/2023] Open
Abstract
To support studies of neuropeptide neuromodulation, I have studied beta-arrestin binding sites (BBS's) by evaluating the incidence of BBS sequences among the C terminal tails (CTs) of each of the 49 Drosophila melanogaster neuropeptide GPCRs. BBS were identified by matches with a prediction derived from structural analysis of rhodopsin:arrestin and vasopressin receptor: arrestin complexes [1]. To increase the rigor of the identification, I determined the conservation of BBS sequences between two long-diverged species D. melanogaster and D. virilis. There is great diversity in the profile of BBS's in this group of GPCRs. I present evidence for conserved BBS's in a majority of the Drosophila neuropeptide GPCRs; notably some have no conserved BBS sequences. In addition, certain GPCRs display numerous conserved compound BBS's, and many GPCRs display BBS-like sequences in their intracellular loop (ICL) domains as well. Finally, 20 of the neuropeptide GPCRs are expressed as protein isoforms that vary in their CT domains. BBS profiles are typically different across related isoforms suggesting a need to diversify and regulate the extent and nature of GPCR:arrestin interactions. This work provides the initial basis to initiate future in vivo, genetic analyses in Drosophila to evaluate the roles of arrestins in neuropeptide GPCR desensitization, trafficking and signaling.
Collapse
Affiliation(s)
- Paul H. Taghert
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, United States of America
- * E-mail:
| |
Collapse
|
9
|
Bipolar cell targeted optogenetic gene therapy restores parallel retinal signaling and high-level vision in the degenerated retina. Commun Biol 2022; 5:1116. [PMID: 36266533 PMCID: PMC9585040 DOI: 10.1038/s42003-022-04016-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 09/21/2022] [Indexed: 11/09/2022] Open
Abstract
Optogenetic gene therapies to restore vision are in clinical trials. Whilst current clinical approaches target the ganglion cells, the output neurons of the retina, new molecular tools enable efficient targeting of the first order retinal interneurons, the bipolar cells, with the potential to restore a higher quality of vision. Here we investigate retinal signaling and behavioral vision in blind mice treated with bipolar cell targeted optogenetic gene therapies. All tested tools, including medium-wave opsin, Opto-mGluR6, and two new melanopsin based chimeras restored visual acuity and contrast sensitivity. The best performing opsin was a melanopsin-mGluR6 chimera, which in some cases restored visual acuities and contrast sensitivities that match wild-type animals. Light responses from the ganglion cells were robust with diverse receptive-field types, inferring elaborate inner retinal signaling. Our results highlight the potential of bipolar cell targeted optogenetics to recover high-level vision in human patients with end-stage retinal degenerations. A chimeric Mela(CTmGluR6) optogenetic tool has the potential to restore vision and signaling in a mouse model of degenerative retinal disease.
Collapse
|
10
|
Dai R, Yu T, Weng D, Li H, Cui Y, Wu Z, Guo Q, Zou H, Wu W, Gao X, Qi Z, Ren Y, Wang S, Li Y, Luo M. A neuropsin-based optogenetic tool for precise control of G q signaling. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1271-1284. [PMID: 35579776 DOI: 10.1007/s11427-022-2122-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
Gq-coupled receptors regulate numerous physiological processes by activating enzymes and inducing intracellular Ca2+ signals. There is a strong need for an optogenetic tool that enables powerful experimental control over Gq signaling. Here, we present chicken opsin 5 (cOpn5) as the long sought-after, single-component optogenetic tool that mediates ultra-sensitive optical control of intracellular Gq signaling with high temporal and spatial resolution. Expressing cOpn5 in HEK 293T cells and primary mouse astrocytes enables blue light-triggered, Gq-dependent Ca2+ release from intracellular stores and protein kinase C activation. Strong Ca2+ transients were evoked by brief light pulses of merely 10 ms duration and at 3 orders lower light intensity of that for common optogenetic tools. Photostimulation of cOpn5-expressing cells at the subcellular and single-cell levels generated fast intracellular Ca2+ transition, thus demonstrating the high spatial precision of cOpn5 optogenetics. The cOpn5-mediated optogenetics could also be applied to activate neurons and control animal behavior in a circuit-dependent manner. cOpn5 optogenetics may find broad applications in studying the mechanisms and functional relevance of Gq signaling in both non-excitable cells and excitable cells in all major organ systems.
Collapse
Affiliation(s)
- Ruicheng Dai
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- School of Life Sciences, Peking University, Beijing, 100871, China
- Peking University-Tsinghua University-NIBS Joint Graduate Program, NIBS, Beijing, 102206, China
| | - Tao Yu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- Peking University-Tsinghua University-NIBS Joint Graduate Program, NIBS, Beijing, 102206, China
| | - Danwei Weng
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- Graduate School of Peking Union Medical College, Beijing, 100730, China
| | - Heng Li
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Beijing, 102206, China
| | - Yuting Cui
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Zhaofa Wu
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, 100871, China
- PKU-McGovern Institute for Brain Research, Beijing, 100871, China
| | - Qingchun Guo
- Chinese Institute for Brain Research, Beijing, 102206, China
- Capital Medical University, Beijing, 102206, China
| | - Haiyue Zou
- Chinese Institute for Brain Research, Beijing, 102206, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Wenting Wu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
- Peking University-Tsinghua University-NIBS Joint Graduate Program, NIBS, Beijing, 102206, China
| | - Xinwei Gao
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Zhongyang Qi
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Yuqi Ren
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Shu Wang
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, 100871, China
- PKU-McGovern Institute for Brain Research, Beijing, 100871, China
| | - Minmin Luo
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China.
- Graduate School of Peking Union Medical College, Beijing, 100730, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research (TIMBR), Beijing, 102206, China.
| |
Collapse
|
11
|
Mansouri M, Fussenegger M. Therapeutic cell engineering: designing programmable synthetic genetic circuits in mammalian cells. Protein Cell 2022; 13:476-489. [PMID: 34586617 PMCID: PMC9226217 DOI: 10.1007/s13238-021-00876-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/02/2021] [Indexed: 12/01/2022] Open
Abstract
Cell therapy approaches that employ engineered mammalian cells for on-demand production of therapeutic agents in the patient's body are moving beyond proof-of-concept in translational medicine. The therapeutic cells can be customized to sense user-defined signals, process them, and respond in a programmable and predictable way. In this paper, we introduce the available tools and strategies employed to design therapeutic cells. Then, various approaches to control cell behaviors, including open-loop and closed-loop systems, are discussed. We also highlight therapeutic applications of engineered cells for early diagnosis and treatment of various diseases in the clinic and in experimental disease models. Finally, we consider emerging technologies such as digital devices and their potential for incorporation into future cell-based therapies.
Collapse
Affiliation(s)
- Maysam Mansouri
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland.
- Faculty of Science, University of Basel, Mattenstrasse 26, 4058, Basel, Switzerland.
| |
Collapse
|
12
|
Gilhooley MJ, Lindner M, Palumaa T, Hughes S, Peirson SN, Hankins MW. A systematic comparison of optogenetic approaches to visual restoration. Mol Ther Methods Clin Dev 2022; 25:111-123. [PMID: 35402632 PMCID: PMC8956963 DOI: 10.1016/j.omtm.2022.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/04/2022] [Indexed: 02/06/2023]
Abstract
During inherited retinal degenerations (IRDs), vision is lost due to photoreceptor cell death; however, a range of optogenetic tools have been shown to restore light responses in animal models. Restored response characteristics vary between tools and the neuronal cell population to which they are delivered: the interplay between these is complex, but targeting upstream neurons (such as retinal bipolar cells) may provide functional benefit by retaining intraretinal signal processing. In this study, our aim was to compare two optogenetic tools: mammalian melanopsin (hOPN4) and microbial red-shifted channelrhodopsin (ReaChR) expressed within two subpopulations of surviving cells in a degenerate retina. Intravitreal adeno-associated viral vectors and mouse models utilising the Cre/lox system restricted expression to populations dominated by bipolar cells or retinal ganglion cells and was compared with non-targeted delivery using the chicken beta actin (CBA) promoter. In summary, we found bipolar-targeted optogenetic tools produced faster kinetics and flatter intensity-response relationships compared with non-targeted or retinal-ganglion-cell-targeted hOPN4. Hence, optogenetic tools of both mammalian and microbial origins show advantages when targeted to bipolar cells. This demonstrates the advantage of bipolar-cell-targeted optogenetics for vision restoration in IRDs. We therefore developed a bipolar-cell-specific gene delivery system employing a compressed promoter with the potential for clinical translation.
Collapse
Affiliation(s)
- Michael J. Gilhooley
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- The Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
- Moorfields Eye Hospital, 162, City Road, London EC1V 2PD, UK
| | - Moritz Lindner
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Institute of Physiology and Pathophysiology, Department of Neurophysiology, Philipps University, Deutschhausstrasse 1-2, Marburg 35037, Germany
| | - Teele Palumaa
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- East Tallinn Central Hospital Eye Clinic, Ravi 18, 10138 Tallinn, Estonia
| | - Steven Hughes
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
| | - Stuart N. Peirson
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
| | - Mark W. Hankins
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Corresponding author Mark W. Hankins, Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK.
| |
Collapse
|
13
|
Li W, Trigg JS, Taghert PH. Regulation of PDF receptor signaling controlling daily locomotor rhythms in Drosophila. PLoS Genet 2022; 18:e1010013. [PMID: 35605015 PMCID: PMC9166358 DOI: 10.1371/journal.pgen.1010013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/03/2022] [Accepted: 04/27/2022] [Indexed: 11/19/2022] Open
Abstract
Each day and in conjunction with ambient daylight conditions, neuropeptide PDF regulates the phase and amplitude of locomotor activity rhythms in Drosophila through its receptor, PDFR, a Family B G protein-coupled receptor (GPCR). We studied the in vivo process by which PDFR signaling turns off, by converting as many as half of the 28 potential sites of phosphorylation in its C terminal tail to a non-phosphorylatable residue (alanine). We report that many such sites are conserved evolutionarily, and their conversion creates a specific behavioral syndrome opposite to loss-of-function phenotypes previously described for pdfr. That syndrome includes increases in the amplitudes of both Morning and Evening behavioral peaks, as well as multi-hour delays of the Evening phase. The precise behavioral effects were dependent on day-length, and most effects mapped to conversion of only a few, specific serine residues near the very end of the protein and specific to its A isoform. Behavioral phase delays of the Evening activity under entraining conditions predicted the phase of activity cycles under constant darkness. The behavioral phenotypes produced by the most severe PDFR variant were ligand-dependent in vivo, and not a consequence of changes to their pharmacological properties, nor of changes in their surface expression, as measured in vitro. The mechanisms underlying termination of PDFR signaling are complex, subject to regulation that is modified by season, and central to a better understanding of the peptidergic modulation of behavior. In multi-cellular organisms, circadian pacemakers create output as a series of phase markers across the 24 hour day to allow other cells to pattern diverse aspects of daily rhythmic physiology and behavior. Within circadian pacemaker circuits, neuropeptide signaling is essential to help promote coherent circadian outputs. In the fruit fly Drosophila 150 neurons are dedicated circadian clocks and they all tell the same time. In spite of such strong synchronization, they provide diverse phasic outputs in the form of their discrete, asynchronous neuronal activity patterns. Neuropeptide signaling breaks the clock-generated symmetry and drives many pacemakers away from their preferred activity period in the morning. Each day, neuropeptide PDF is released by Morning pacemakers and delays the phase of activity of specific other pacemakers to later parts of the day or night. When and how the PDF that is released in the morning stops acting is unknown. Furthermore, timing of signal termination is not fixed because day length changes each day, hence the modulatory delay exerted by PDF must itself be regulated. Here we test a canonical model of G protein-coupled receptor physiology to ask how PDF receptor signaling is normally de-activated. We use behavioral measures to define sequence elements of the receptor whose post-translational modifications (e.g., phosphorylation) may define the duration of receptor signaling.
Collapse
Affiliation(s)
- Weihua Li
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Jennifer S. Trigg
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Paul H. Taghert
- Department of Neuroscience, Washington University School of Medicine, St Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
14
|
Dekens MPS, Fontinha BM, Gallach M, Pflügler S, Tessmar‐Raible K. Melanopsin elevates locomotor activity during the wake state of the diurnal zebrafish. EMBO Rep 2022; 23:e51528. [PMID: 35233929 PMCID: PMC9066073 DOI: 10.15252/embr.202051528] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 01/24/2022] [Accepted: 02/04/2022] [Indexed: 11/28/2022] Open
Abstract
Mammalian and fish pineals play a key role in adapting behaviour to the ambient light conditions through the release of melatonin. In mice, light inhibits nocturnal locomotor activity via the non‐visual photoreceptor Melanopsin. In contrast to the extensively studied function of Melanopsin in the indirect regulation of the rodent pineal, its role in the intrinsically photosensitive zebrafish pineal has not been elucidated. Therefore, it is not evident if the light signalling mechanism is conserved between distant vertebrates, and how Melanopsin could affect diurnal behaviour. A double knockout of melanopsins (opn4.1‐opn4xb) was generated in the diurnal zebrafish, which manifests attenuated locomotor activity during the wake state. Transcriptome sequencing gave insight into pathways downstream of Melanopsin, implying that sustained repression of the melatonin pathway is required to elevate locomotor activity during the diurnal wake state. Moreover, we show that light induces locomotor activity during the diurnal wake state in an intensity‐dependent manner. These observations suggest a common Melanopsin‐driven mechanism between zebrafish and mammals, while the diurnal and nocturnal chronotypes are inversely regulated downstream of melatonin.
Collapse
Affiliation(s)
- Marcus P S Dekens
- Max Perutz Laboratory Centre for Molecular Biology University of Vienna and Medical University of Vienna Vienna Austria
| | - Bruno M Fontinha
- Max Perutz Laboratory Centre for Molecular Biology University of Vienna and Medical University of Vienna Vienna Austria
| | - Miguel Gallach
- Max Perutz Laboratory Centre for Molecular Biology University of Vienna and Medical University of Vienna Vienna Austria
- Max Perutz Laboratory Centre for Integrative Bioinformatics University of Vienna and Medical University of Vienna Vienna Austria
| | - Sandra Pflügler
- Max Perutz Laboratory Centre for Molecular Biology University of Vienna and Medical University of Vienna Vienna Austria
| | - Kristin Tessmar‐Raible
- Max Perutz Laboratory Centre for Molecular Biology University of Vienna and Medical University of Vienna Vienna Austria
- Research Platform “Marine Rhythms of Life” University of Vienna Vienna Austria
| |
Collapse
|
15
|
Sheng Y, Chen L, Ren X, Jiang Z, Yau KW. Molecular determinants of response kinetics of mouse M1 intrinsically-photosensitive retinal ganglion cells. Sci Rep 2021; 11:23424. [PMID: 34873237 PMCID: PMC8648817 DOI: 10.1038/s41598-021-02832-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/19/2021] [Indexed: 11/28/2022] Open
Abstract
Intrinsically-photosensitive retinal ganglion cells (ipRGCs) are non-rod/non-cone retinal photoreceptors expressing the visual pigment, melanopsin, to detect ambient irradiance for various non-image-forming visual functions. The M1-subtype, amongst the best studied, mediates primarily circadian photoentrainment and pupillary light reflex. Their intrinsic light responses are more prolonged than those of rods and cones even at the single-photon level, in accordance with the typically slower time course of non-image-forming vision. The short (OPN4S) and long (OPN4L) alternatively-spliced forms of melanopsin proteins are both present in M1-ipRGCs, but their functional difference is unclear. We have examined this point by genetically removing the Opn4 gene (Opn4-/-) in mouse and re-expressing either OPN4S or OPN4L singly in Opn4-/- mice by using adeno-associated virus, but found no obvious difference in their intrinsic dim-flash responses. Previous studies have indicated that two dominant slow steps in M1-ipRGC phototransduction dictate these cells' intrinsic dim-flash-response kinetics, with time constants (τ1 and τ2) at room temperature of ~ 2 s and ~ 20 s, respectively. Here we found that melanopsin inactivation by phosphorylation or by β-arrestins may not be one of these two steps, because their genetic disruptions did not prolong the two time constants or affect the response waveform. Disruption of GAP (GTPase-Activating-Protein) activity on the effector enzyme, PLCβ4, in M1-ipRGC phototransduction to slow down G-protein deactivation also did not prolong the response decay, but caused its rising phase to become slightly sigmoidal by giving rise to a third time constant, τ3, of ~ 2 s (room temperature). This last observation suggests that GAP-mediated G-protein deactivation does partake in the flash-response termination, although normally with a time constant too short to be visible in the response waveform.
Collapse
Affiliation(s)
- Yanghui Sheng
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA
- Graduate Neuroscience Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Lujing Chen
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA
- Graduate Neuroscience Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA, 02115, USA
| | - Xiaozhi Ren
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA
- Vedere Bio II, Inc., 700 Main St, Cambridge, MA, 02139, USA
| | - Zheng Jiang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA
- Department of Ophthalmology, Baylor College of Medicine, 6565 Fannin St, Houston, TX, 77030, USA
| | - King-Wai Yau
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 725 North Wolfe St, Baltimore, MD, 21205, USA.
| |
Collapse
|
16
|
Contreras E, Nobleman AP, Robinson PR, Schmidt TM. Melanopsin phototransduction: beyond canonical cascades. J Exp Biol 2021; 224:273562. [PMID: 34842918 PMCID: PMC8714064 DOI: 10.1242/jeb.226522] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Melanopsin is a visual pigment that is expressed in a small subset of intrinsically photosensitive retinal ganglion cells (ipRGCs). It is involved in regulating non-image forming visual behaviors, such as circadian photoentrainment and the pupillary light reflex, while also playing a role in many aspects of image-forming vision, such as contrast sensitivity. Melanopsin was initially discovered in the melanophores of the skin of the frog Xenopus, and subsequently found in a subset of ganglion cells in rat, mouse and primate retinas. ipRGCs were initially thought to be a single retinal ganglion cell population, and melanopsin was thought to activate a single, invertebrate-like Gq/transient receptor potential canonical (TRPC)-based phototransduction cascade within these cells. However, in the 20 years since the discovery of melanopsin, our knowledge of this visual pigment and ipRGCs has expanded dramatically. Six ipRGC subtypes have now been identified in the mouse, each with unique morphological, physiological and functional properties. Multiple subtypes have also been identified in other species, suggesting that this cell type diversity is a general feature of the ipRGC system. This diversity has led to a renewed interest in melanopsin phototransduction that may not follow the canonical Gq/TRPC cascade in the mouse or in the plethora of other organisms that express the melanopsin photopigment. In this Review, we discuss recent findings and discoveries that have challenged the prevailing view of melanopsin phototransduction as a single pathway that influences solely non-image forming functions.
Collapse
Affiliation(s)
- Ely Contreras
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA,Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, IL 60208, USA
| | - Alexis P. Nobleman
- University of Maryland Baltimore County, Department of Biological Sciences, Baltimore, MD 21250, USA,Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, NIH, Bethesda, MD 20892, USA
| | - Phyllis R. Robinson
- University of Maryland Baltimore County, Department of Biological Sciences, Baltimore, MD 21250, USA,Authors for correspondence (; )
| | - Tiffany M. Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA,Department of Ophthalmology, Feinberg School of Medicine, Chicago, IL 60611, USA,Authors for correspondence (; )
| |
Collapse
|
17
|
Fasick JI, Algrain H, Samuels C, Mahadevan P, Schweikert LE, Naffaa ZJ, Robinson PR. Spectral tuning and deactivation kinetics of marine mammal melanopsins. PLoS One 2021; 16:e0257436. [PMID: 34653198 PMCID: PMC8519484 DOI: 10.1371/journal.pone.0257436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/31/2021] [Indexed: 02/04/2023] Open
Abstract
In mammals, the photopigment melanopsin (Opn4) is found in a subset of retinal ganglion cells that serve light detection for circadian photoentrainment and pupil constriction (i.e., mydriasis). For a given species, the efficiency of photoentrainment and length of time that mydriasis occurs is determined by the spectral sensitivity and deactivation kinetics of melanopsin, respectively, and to date, neither of these properties have been described in marine mammals. Previous work has indicated that the absorbance maxima (λmax) of marine mammal rhodopsins (Rh1) have diversified to match the available light spectra at foraging depths. However, similar to the melanopsin λmax of terrestrial mammals (~480 nm), the melanopsins of marine mammals may be conserved, with λmax values tuned to the spectrum of solar irradiance at the water's surface. Here, we investigated the Opn4 pigments of 17 marine mammal species inhabiting diverse photic environments including the Infraorder Cetacea, as well as the Orders Sirenia and Carnivora. Both genomic and cDNA sequences were used to deduce amino acid sequences to identify substitutions most likely involved in spectral tuning and deactivation kinetics of the Opn4 pigments. Our results show that there appears to be no amino acid substitutions in marine mammal Opn4 opsins that would result in any significant change in λmax values relative to their terrestrial counterparts. We also found some marine mammal species to lack several phosphorylation sites in the carboxyl terminal domain of their Opn4 pigments that result in significantly slower deactivation kinetics, and thus longer mydriasis, compared to terrestrial controls. This finding was restricted to cetacean species previously found to lack cone photoreceptor opsins, a condition known as rod monochromacy. These results suggest that the rod monochromat whales rely on extended pupillary constriction to prevent photobleaching of the highly photosensitive all-rod retina when moving between photopic and scotopic conditions.
Collapse
Affiliation(s)
- Jeffry I. Fasick
- Department of Biological Sciences, The University of Tampa, Tampa, Florida, United States of America
| | - Haya Algrain
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States of America
| | - Courtland Samuels
- Department of Chemistry, University of South Florida, Tampa, Florida, United States of America
| | - Padmanabhan Mahadevan
- Department of Biological Sciences, The University of Tampa, Tampa, Florida, United States of America
| | - Lorian E. Schweikert
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, North Carolina, United States of America
| | - Zaid J. Naffaa
- Department of Biological Sciences, Kean University, Union, New Jersey, United States of America
| | - Phyllis R. Robinson
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States of America
| |
Collapse
|
18
|
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) signal not only anterogradely to drive behavioral responses, but also retrogradely to some amacrine interneurons to modulate retinal physiology. We previously found that all displaced amacrine cells with spiking, tonic excitatory photoresponses receive gap-junction input from ipRGCs, but the connectivity patterns and functional roles of ipRGC-amacrine coupling remained largely unknown. Here, we injected PoPro1 fluorescent tracer into all six types of mouse ipRGCs to identify coupled amacrine cells, and analyzed the latter's morphological and electrophysiological properties. We also examined how genetically disrupting ipRGC-amacrine coupling affected ipRGC photoresponses. Results showed that ipRGCs couple with not just ON- and ON/OFF-stratified amacrine cells in the ganglion-cell layer as previously reported, but also OFF-stratified amacrine cells in both ganglion-cell and inner nuclear layers. M1- and M3-type ipRGCs couple mainly with ON/OFF-stratified amacrine cells, whereas the other ipRGC types couple almost exclusively with ON-stratified ones. ipRGCs transmit melanopsin-based light responses to at least 93% of the coupled amacrine cells. Some of the ON-stratifying ipRGC-coupled amacrine cells exhibit transient hyperpolarizing light responses. We detected bidirectional electrical transmission between an ipRGC and a coupled amacrine cell, although transmission was asymmetric for this particular cell pair, favoring the ipRGC-to-amacrine direction. We also observed electrical transmission between two amacrine cells coupled to the same ipRGC. In both scenarios of coupling, the coupled cells often spiked synchronously. While ipRGC-amacrine coupling somewhat reduces the peak firing rates of ipRGCs' intrinsic melanopsin-based photoresponses, it renders these responses more sustained and longer-lasting. In summary, ipRGCs' gap junctional network involves more amacrine cell types and plays more roles than previously appreciated.
Collapse
|
19
|
Morita Y, Jounai K, Tomita Y, Maruyama M. Long-term intake of Lactobacillus paracasei KW3110 prevents age-related circadian locomotor activity and changes in gut metabolism in physiologically aged mice. Exp Gerontol 2021; 153:111477. [PMID: 34271136 DOI: 10.1016/j.exger.2021.111477] [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: 04/20/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 11/20/2022]
Abstract
Aging involves age-progressive loss of physiological functions in organs and tissues. We previously showed that Lactobacillus paracasei KW3110 suppressed age-related inflammation and prevented age-related retinal ganglion cell (RGC) loss. As RGCs mediate biological behaviors associated with responses to ambient light, we assessed whether L. paracasei KW3110 affects circadian locomotor activities in physiologically aged mice. The ratio of locomotor activity during the nighttime (active phase) to daytime (inactive phase) significantly decreased in physiologically aged mice compared with young mice: intake of L. paracasei KW3110 prevented this decrease. We also performed metabolomics analysis of cecal contents using both capillary electrophoresis and liquid chromatography time-of-flight mass spectrometry to better understand the benefical effects for aging of L. paracasei KW3110 through a gut retina axis, since our previous study showed that L. paracasei KW3110 mitigated not only age-related expansions of intestinal inflammatory immune cells but age-related alternation of gut microbiome composition. Principal component analysis showed clear changes in metabolites between physiologically aged mice fed a diet containing L. paracasei KW3110 and age-matched control mice. Furthermore, we found that intake of L. paracasei KW3110 mitigated age-related changes in some fatty acids compared with age-matched control mice. Taken together, L. paracasei KW3110 might regulate age-related alternation of metabolites in cecal contents, potentially leading to suppression of age-related decline in physiological functions, including impairment of circadian locomotor activities.
Collapse
Affiliation(s)
- Yuji Morita
- KIRIN Central Research Institute, Kirin Holdings Co., Ltd., Kanagawa, Japan.
| | - Kenta Jounai
- Technical Development Center, Koiwai Dairy Products Co., Ltd., Sayama, Saitama 350-1321, Japan
| | - Yasuyuki Tomita
- KIRIN Central Research Institute, Kirin Holdings Co., Ltd., Kanagawa, Japan
| | - Mitsuo Maruyama
- Department of Inflammation and Immunosenescence, Geroscience Research Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan.; Department of Aging Research, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
| |
Collapse
|
20
|
Aranda ML, Schmidt TM. Diversity of intrinsically photosensitive retinal ganglion cells: circuits and functions. Cell Mol Life Sci 2021; 78:889-907. [PMID: 32965515 PMCID: PMC8650628 DOI: 10.1007/s00018-020-03641-5] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/10/2020] [Accepted: 09/03/2020] [Indexed: 12/25/2022]
Abstract
The melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs) are a relatively recently discovered class of atypical ganglion cell photoreceptor. These ipRGCs are a morphologically and physiologically heterogeneous population that project widely throughout the brain and mediate a wide array of visual functions ranging from photoentrainment of our circadian rhythms, to driving the pupillary light reflex to improve visual function, to modulating our mood, alertness, learning, sleep/wakefulness, regulation of body temperature, and even our visual perception. The presence of melanopsin as a unique molecular signature of ipRGCs has allowed for the development of a vast array of molecular and genetic tools to study ipRGC circuits. Given the emerging complexity of this system, this review will provide an overview of the genetic tools and methods used to study ipRGCs, how these tools have been used to dissect their role in a variety of visual circuits and behaviors in mice, and identify important directions for future study.
Collapse
Affiliation(s)
- Marcos L Aranda
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Tiffany M Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
21
|
Valdez-Lopez JC, Gebreegziabher M, Bailey RJ, Flores J, Awotunde O, Burnett T, Robinson PR. Protein Phosphatase 2A and Clathrin-Mediated Endocytosis Facilitate Robust Melanopsin Light Responses and Resensitization. Invest Ophthalmol Vis Sci 2020; 61:10. [PMID: 33049058 PMCID: PMC7571330 DOI: 10.1167/iovs.61.12.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Intrinsically photosensitive retinal ganglion cells (ipRGCs) that express the visual pigment melanopsin regulate non-image-forming visual tasks, such as circadian photoentrainment and pupil constriction, as well as contrast detection for image formation. Sustained ipRGC function throughout the day is, therefore, of great importance. Melanopsin is a bistable rhabdomeric-type (R-type) visual pigment, which is thought to use light to regenerate its chromophore from all-trans-retinal back to 11-cis-retinal and does not depend on constant chromophore supply to the extent required by visual pigment in rod and cone photoreceptors. Like the majority of photopigments and G-protein-coupled receptors (GPCRs), melanopsin deactivation requires C-terminal phosphorylation and subsequent β-arrestin binding. We hypothesize that melanopsin utilizes canonical GPCR resensitization mechanisms, including dephosphorylation and endocytosis, during the light, and together, they provide a mechanism for prolonged light responses. Methods Here, we examined expression of protein phosphatases from a variety of subfamilies by RT-PCR and immunohistochemical analyses of the mouse retina. The expression of protein phosphatase 2A (PP2A) in ipRGCs was assessed. We also examine the role of phosphatase and endocytic activity in sustaining melanopsin signaling using transiently-transfected HEK293 cells. Results Our analyses suggest that melanopsin-mediated light responses can be rapidly and extensively enhanced by PP2A activity. Light-activated melanopsin undergoes endocytosis in a clathrin-dependent manner. This endocytic activity enhances light responses upon repeated stimulation, implicating a role for endocytic activity in resensitization. Conclusions Thus, we propose that melanopsin phototransduction is maintained by utilizing canonical GPCR resensitization mechanisms rather than reliance on chromophore replenishment from supporting cells.
Collapse
Affiliation(s)
- Juan C Valdez-Lopez
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States
| | - Meheret Gebreegziabher
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States.,National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Robin J Bailey
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States
| | - Jair Flores
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States
| | - Olanike Awotunde
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States
| | - Thomas Burnett
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States.,Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Phyllis R Robinson
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States
| |
Collapse
|
22
|
Kim KY, Rios LC, Le H, Perez AJ, Phan S, Bushong EA, Deerinck TJ, Liu YH, Ellisman MA, Lev-Ram V, Ju S, Panda SA, Yoon S, Hirayama M, Mure LS, Hatori M, Ellisman MH, Panda S. Synaptic Specializations of Melanopsin-Retinal Ganglion Cells in Multiple Brain Regions Revealed by Genetic Label for Light and Electron Microscopy. Cell Rep 2020; 29:628-644.e6. [PMID: 31618632 DOI: 10.1016/j.celrep.2019.09.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 07/01/2019] [Accepted: 09/04/2019] [Indexed: 11/17/2022] Open
Abstract
The form and synaptic fine structure of melanopsin-expressing retinal ganglion cells, also called intrinsically photosensitive retinal ganglion cells (ipRGCs), were determined using a new membrane-targeted version of a genetic probe for correlated light and electron microscopy (CLEM). ipRGCs project to multiple brain regions, and because the method labels the entire neuron, it was possible to analyze nerve terminals in multiple retinorecipient brain regions, including the suprachiasmatic nucleus (SCN), olivary pretectal nucleus (OPN), and subregions of the lateral geniculate. Although ipRGCs provide the only direct retinal input to the OPN and SCN, ipRGC terminal arbors and boutons were found to be remarkably different in each target region. A network of dendro-dendritic chemical synapses (DDCSs) was also revealed in the SCN, with ipRGC axon terminals preferentially synapsing on the DDCS-linked cells. The methods developed to enable this analysis should propel other CLEM studies of long-distance brain circuits at high resolution.
Collapse
Affiliation(s)
- Keun-Young Kim
- Department of Neurosciences, University of California at San Diego School of Medicine, La Jolla, CA, USA; National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Luis C Rios
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hiep Le
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Alex J Perez
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Sébastien Phan
- Department of Neurosciences, University of California at San Diego School of Medicine, La Jolla, CA, USA; National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Eric A Bushong
- Department of Neurosciences, University of California at San Diego School of Medicine, La Jolla, CA, USA; National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Thomas J Deerinck
- Department of Neurosciences, University of California at San Diego School of Medicine, La Jolla, CA, USA; National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Yu Hsin Liu
- Salk Institute for Biological Studies, La Jolla, CA, USA; Medical Scientist Training Program, University of California at San Diego School of Medicine, La Jolla, CA, USA
| | - Maya A Ellisman
- Biological Sciences Graduate Training Program, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Varda Lev-Ram
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Suyeon Ju
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Sneha A Panda
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | - Sanghee Yoon
- National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA
| | | | - Ludovic S Mure
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Megumi Hatori
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Mark H Ellisman
- Department of Neurosciences, University of California at San Diego School of Medicine, La Jolla, CA, USA; National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA, USA; Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA.
| | | |
Collapse
|
23
|
Valdez-Lopez JC, Petr ST, Donohue MP, Bailey RJ, Gebreeziabher M, Cameron EG, Wolf JB, Szalai VA, Robinson PR. The C-Terminus and Third Cytoplasmic Loop Cooperatively Activate Mouse Melanopsin Phototransduction. Biophys J 2020; 119:389-401. [PMID: 32621866 PMCID: PMC7376183 DOI: 10.1016/j.bpj.2020.06.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/28/2020] [Accepted: 06/10/2020] [Indexed: 11/30/2022] Open
Abstract
Melanopsin, an atypical vertebrate visual pigment, mediates non-image-forming light responses including circadian photoentrainment and pupillary light reflexes and contrast detection for image formation. Melanopsin-expressing intrinsically photosensitive retinal ganglion cells are characterized by sluggish activation and deactivation of their light responses. The molecular determinants of mouse melanopsin's deactivation have been characterized (i.e., C-terminal phosphorylation and β-arrestin binding), but a detailed analysis of melanopsin's activation is lacking. We propose that an extended third cytoplasmic loop is adjacent to the proximal C-terminal region of mouse melanopsin in the inactive conformation, which is stabilized by the ionic interaction of these two regions. This model is supported by site-directed spin labeling and electron paramagnetic resonance spectroscopy of melanopsin, the results of which suggests a high degree of steric freedom at the third cytoplasmic loop, which is increased upon C-terminus truncation, supporting the idea that these two regions are close in three-dimensional space in wild-type melanopsin. To test for a functionally critical C-terminal conformation, calcium imaging of melanopsin mutants including a proximal C-terminus truncation (at residue 365) and proline mutation of this proximal region (H377P, L380P, Y382P) delayed melanopsin's activation rate. Mutation of all potential phosphorylation sites, including a highly conserved tyrosine residue (Y382), into alanines also delayed the activation rate. A comparison of mouse melanopsin with armadillo melanopsin-which has substitutions of various potential phosphorylation sites and a substitution of the conserved tyrosine-indicates that substitution of these potential phosphorylation sites and the tyrosine residue result in dramatically slower activation kinetics, a finding that also supports the role of phosphorylation in signaling activation. We therefore propose that melanopsin's C-terminus is proximal to intracellular loop 3, and C-terminal phosphorylation permits the ionic interaction between these two regions, thus forming a stable structural conformation that is critical for initiating G-protein signaling.
Collapse
Affiliation(s)
- Juan C Valdez-Lopez
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland
| | - Stephen T Petr
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland
| | - Matthew P Donohue
- Center for Nanoscale and Technology, National Institutes of Standards and Technology, Gaithersburg, Maryland; Maryland NanoCenter, University of Maryland College Park, College Park, Maryland
| | - Robin J Bailey
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland
| | - Meheret Gebreeziabher
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland
| | - Evan G Cameron
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland
| | - Julia B Wolf
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland
| | - Veronika A Szalai
- Center for Nanoscale and Technology, National Institutes of Standards and Technology, Gaithersburg, Maryland
| | - Phyllis R Robinson
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland.
| |
Collapse
|
24
|
Valdez-Lopez JC, Gulati S, Ortiz EA, Palczewski K, Robinson PR. Melanopsin Carboxy-terminus phosphorylation plasticity and bulk negative charge, not strict site specificity, achieves phototransduction deactivation. PLoS One 2020; 15:e0228121. [PMID: 32236094 PMCID: PMC7112210 DOI: 10.1371/journal.pone.0228121] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/06/2020] [Indexed: 11/19/2022] Open
Abstract
Melanopsin is a visual pigment expressed in a small subset of ganglion cells in the mammalian retina known as intrinsically photosensitive retinal ganglion cells (ipRGCs) and is implicated in regulating non-image forming functions such as circadian photoentrainment and pupil constriction and contrast sensitivity in image formation. Mouse melanopsin's Carboxy-terminus (C-terminus) possesses 38 serine and threonine residues, which can potentially serve as phosphorylation sites for a G-protein Receptor Kinase (GRK) and be involved in the deactivation of signal transduction. Previous studies suggest that S388, T389, S391, S392, S394, S395 on the proximal region of the C-terminus of mouse melanopsin are necessary for melanopsin deactivation. We expressed a series of mouse melanopsin C-terminal mutants in HEK293 cells and using calcium imaging, and we found that the necessary cluster of six serine and threonine residues, while being critical, are insufficient for proper melanopsin deactivation. Interestingly, the additional six serine and threonine residues adjacent to the required six sites, in either proximal or distal direction, are capable of restoring wild-type deactivation of melanopsin. These findings suggest an element of plasticity in the molecular basis of melanopsin phosphorylation and deactivation. In addition, C-terminal chimeric mutants and molecular modeling studies support the idea that the initial steps of deactivation and β-arrestin binding are centered around these critical phosphorylation sites (S388-S395). The degree of functional versatility described in this study, along with ipRGC biophysical heterogeneity and the possible use of multiple signal transduction cascades, might contribute to the diverse ipRGC light responses for use in non-image and image forming behaviors, even though all six sub types of ipRGCs express the same melanopsin gene OPN4.
Collapse
MESH Headings
- HEK293 Cells
- Humans
- Light Signal Transduction/physiology
- Models, Molecular
- Mutagenesis, Site-Directed
- Mutation
- Phosphorylation/physiology
- Protein Binding
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Rod Opsins/chemistry
- Rod Opsins/genetics
- Rod Opsins/metabolism
- Serine/genetics
- Serine/metabolism
- Threonine/genetics
- Threonine/metabolism
- beta-Arrestin 1/chemistry
- beta-Arrestin 1/metabolism
Collapse
Affiliation(s)
- Juan C. Valdez-Lopez
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States of America
| | - Sahil Gulati
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, California, United States of America
- Gatan Inc, Pleasanton, California, United States of America
| | - Elelbin A. Ortiz
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States of America
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Krzysztof Palczewski
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, California, United States of America
| | - Phyllis R. Robinson
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland, United States of America
| |
Collapse
|
25
|
Vanderstraeten J, Gailly P, Malkemper EP. Light entrainment of retinal biorhythms: cryptochrome 2 as candidate photoreceptor in mammals. Cell Mol Life Sci 2020; 77:875-884. [PMID: 31982933 PMCID: PMC11104904 DOI: 10.1007/s00018-020-03463-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 12/31/2022]
Abstract
The mechanisms that synchronize the biorhythms of the mammalian retina with the light/dark cycle are independent of those synchronizing the rhythms in the central pacemaker, the suprachiasmatic nucleus. The identity of the photoreceptor(s) responsible for the light entrainment of the retina of mammals is still a matter of debate, and recent studies have reported contradictory results in this respect. Here, we suggest that cryptochromes (CRY), in particular CRY 2, are involved in that light entrainment. CRY are highly conserved proteins that are a key component of the cellular circadian clock machinery. In plants and insects, they are responsible for the light entrainment of these biorhythms, mediated by the light response of their flavin cofactor (FAD). In mammals, however, no light-dependent role is currently assumed for CRY in light-exposed tissues, including the retina. It has been reported that FAD influences the function of mammalian CRY 2 and that human CRY 2 responds to light in Drosophila, suggesting that mammalian CRY 2 keeps the ability to respond to light. Here, we hypothesize that CRY 2 plays a role in the light entrainment of retinal biorhythms, at least in diurnal mammals. Indeed, published data shows that the light intensity dependence and the wavelength sensitivity commonly reported for that light entrainment fits the light sensitivity and absorption spectrum of light-responsive CRY. We propose experiments to test our hypothesis and to further explore the still-pending question of the function of CRY 2 in the mammalian retina.
Collapse
Affiliation(s)
- Jacques Vanderstraeten
- Faculty of Medicine, School of Public Health, Environmental and Work Health Research Center, Université Libre de Bruxelles, CP593, Route de Lennik, 808, 1070, Brussels, Belgium.
- , Avenue Constant Montald, 11, 1200, Brussels, Belgium.
| | - Philippe Gailly
- Faculty of Medicine, Institute of Neuroscience (IONS), Cellular and Molecular Pole (CEMO), Catholic University of Louvain, Avenue Mounier 53/B1.53.17, 1200, Brussels, Belgium
| | - E Pascal Malkemper
- Center of Advanced European Studies and Research (CAESAR), Ludwig-Erhard-Allee 2, Bonn, 53175, Germany
| |
Collapse
|
26
|
Mansouri M, Lichtenstein S, Strittmatter T, Buchmann P, Fussenegger M. Construction of a Multiwell Light-Induction Platform for Traceless Control of Gene Expression in Mammalian Cells. Methods Mol Biol 2020; 2173:189-199. [PMID: 32651919 DOI: 10.1007/978-1-0716-0755-8_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Mammalian cells can be engineered to incorporate light-responsive elements that reliably sense stimulation by light and activate endogenous pathways, such as the cAMP or Ca2+ pathway, to control gene expression. Light-inducible gene expression systems offer high spatiotemporal resolution, and are also traceless, reversible, tunable, and inexpensive. Melanopsin, a well-known representative of the animal opsins, is a G-protein-coupled receptor that triggers a Gαq-dependent signaling cascade upon activation with blue light (≈470 nm). Here, we describe how to rewire melanopsin activation by blue light to transgene expression in mammalian cells, with detailed instructions for constructing a 96-LED array platform with multiple tunable parameters for illumination of the engineered cells in multiwell plates.
Collapse
Affiliation(s)
- Maysam Mansouri
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Samson Lichtenstein
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Tobias Strittmatter
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Peter Buchmann
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland. .,Faculty of Science, University of Basel, Basel, Switzerland.
| |
Collapse
|
27
|
Mure LS, Hatori M, Ruda K, Benegiamo G, Demas J, Panda S. Sustained Melanopsin Photoresponse Is Supported by Specific Roles of β-Arrestin 1 and 2 in Deactivation and Regeneration of Photopigment. Cell Rep 2019; 25:2497-2509.e4. [PMID: 30485815 PMCID: PMC6396282 DOI: 10.1016/j.celrep.2018.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/04/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
Melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) are indispensable for non-image-forming visual responses that sustain under prolonged illumination. For sustained signaling of ipRGCs, the melanopsin photopigment must continuously regenerate. The underlying mechanism is unknown. We discovered that a cluster of Ser/Thr sites within the C-terminal region of mammalian melanopsin is phosphorylated after a light pulse. This forms a binding site for β-arrestin 1 (βARR1) and β-arrestin 2. β-arrestin 2 primarily regulates the deactivation of melanopsin; accordingly, βαrr2–/–mice exhibit prolonged ipRGC responses after cessation of a light pulse. β-arrestin 1 primes melanopsin for regeneration. Therefore, βαrr1–/– ipRGCs become desensitized after repeated or prolonged photostimulation. The lack of either β-arrestin atten-uates ipRGC response under prolonged illumination, suggesting that β-arrestin 2-mediated deactivation and β-arrestin 1-dependent regeneration of melanopsin function in sequence. In conclusion, we discovered a molecular mechanism by which β-arrestins regulate different aspects of melanopsin photoresponses and allow ipRGC-sustained responses under prolonged illumination. The mechanism by which melanopsin-expressing retinal ganglion cells (mRGCs) tonically respond to continuous illumination is unknown. Mure et al. show that phosphorylation-dependent binding of β-arrestin 1 and 2 coordinately deactivate and regenerate melanopsin photopigment to enable sustained firing of mRGCs in response to prolonged illumination.
Collapse
Affiliation(s)
- Ludovic S Mure
- Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Megumi Hatori
- Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; Keio University School of Medicine, Tokyo, Japan
| | - Kiersten Ruda
- St. Olaf College, 1520 St. Olaf Avenue, Northfield, MN 55057, USA
| | - Giorgia Benegiamo
- Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - James Demas
- St. Olaf College, 1520 St. Olaf Avenue, Northfield, MN 55057, USA
| | - Satchidananda Panda
- Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
28
|
Long-term intake of Lactobacillus paracasei KW3110 prevents age-related chronic inflammation and retinal cell loss in physiologically aged mice. Aging (Albany NY) 2019; 10:2723-2740. [PMID: 30341255 PMCID: PMC6224250 DOI: 10.18632/aging.101583] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 09/26/2018] [Indexed: 01/10/2023]
Abstract
Age-related chronic inflammation is a major risk factor for the incidence and prevalence of age-related diseases, including infectious and neurodegenerative diseases. We previously reported that a lactic acid bacteria, Lactobacillus paracasei KW3110, activated macrophages and suppressed inflammation in mice and humans. In this study, we investigated whether long-term intake of heat-killed L. paracasei KW3110 modulated age-related inflammation and altered the gut microbiota in physiologically aged mice. Compared with age-matched control mice, fecal analyses of gut microbiota revealed that intake of L. paracasei KW3110 mitigated age-related changes of beneficial bacterial composition, including the Bifidobacteriaceae family. L. paracasei KW3110 intake also mitigated age-related immune defects by reducing the prevalence of interferon-gamma (IFN-γ) -producing inflammatory CD4-positive T cells in the lamina propia of the small intestine, and reduced serum levels of proinflammatory cytokines. Furthermore, L. paracasei KW3110 intake suppressed retinal inflammation by reducing proinflammatory cytokine-producing macrophage, and age-related retinal cell loss. Taken together, these findings suggested that L. paracasei KW3110 mitigated age-related chronic inflammation through modulation of gut microbiota composition and immune system functions in aged mice, and also reduced age-related retinal ganglion cell (RGC) loss. Further studies are needed to evaluate the effect in age-related senescent changes of the retina.
Collapse
|
29
|
Do MTH. Melanopsin and the Intrinsically Photosensitive Retinal Ganglion Cells: Biophysics to Behavior. Neuron 2019; 104:205-226. [PMID: 31647894 PMCID: PMC6944442 DOI: 10.1016/j.neuron.2019.07.016] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/19/2019] [Accepted: 07/12/2019] [Indexed: 12/19/2022]
Abstract
The mammalian visual system encodes information over a remarkable breadth of spatiotemporal scales and light intensities. This performance originates with its complement of photoreceptors: the classic rods and cones, as well as the intrinsically photosensitive retinal ganglion cells (ipRGCs). IpRGCs capture light with a G-protein-coupled receptor called melanopsin, depolarize like photoreceptors of invertebrates such as Drosophila, discharge electrical spikes, and innervate dozens of brain areas to influence physiology, behavior, perception, and mood. Several visual responses rely on melanopsin to be sustained and maximal. Some require ipRGCs to occur at all. IpRGCs fulfill their roles using mechanisms that include an unusual conformation of the melanopsin protein, an extraordinarily slow phototransduction cascade, divisions of labor even among cells of a morphological type, and unorthodox configurations of circuitry. The study of ipRGCs has yielded insight into general topics that include photoreceptor evolution, cellular diversity, and the steps from biophysical mechanisms to behavior.
Collapse
Affiliation(s)
- Michael Tri H Do
- F.M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital and Harvard Medical School, Center for Life Science 12061, 3 Blackfan Circle, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Mederos S, Hernández-Vivanco A, Ramírez-Franco J, Martín-Fernández M, Navarrete M, Yang A, Boyden ES, Perea G. Melanopsin for precise optogenetic activation of astrocyte-neuron networks. Glia 2019; 67:915-934. [PMID: 30632636 DOI: 10.1002/glia.23580] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022]
Abstract
Optogenetics has been widely expanded to enhance or suppress neuronal activity and it has been recently applied to glial cells. Here, we have used a new approach based on selective expression of melanopsin, a G-protein-coupled photopigment, in astrocytes to trigger Ca2+ signaling. Using the genetically encoded Ca2+ indicator GCaMP6f and two-photon imaging, we show that melanopsin is both competent to stimulate robust IP3-dependent Ca2+ signals in astrocyte fine processes, and to evoke an ATP/Adenosine-dependent transient boost of hippocampal excitatory synaptic transmission. Additionally, under low-frequency light stimulation conditions, melanopsin-transfected astrocytes can trigger long-term synaptic changes. In vivo, melanopsin-astrocyte activation enhances episodic-like memory, suggesting melanopsin as an optical tool that could recapitulate the wide range of regulatory actions of astrocytes on neuronal networks in behaving animals. These results describe a novel approach using melanopsin as a precise trigger for astrocytes that mimics their endogenous G-protein signaling pathways, and present melanopsin as a valuable optical tool for neuron-glia studies.
Collapse
Affiliation(s)
- Sara Mederos
- Department of Functional and Systems Neurobiology, Instituto Cajal, CSIC, Madrid, Spain
| | | | - Jorge Ramírez-Franco
- Department of Functional and Systems Neurobiology, Instituto Cajal, CSIC, Madrid, Spain
| | | | - Marta Navarrete
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Aimei Yang
- Media Lab, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Edward S Boyden
- Media Lab, Massachusetts Institute of Technology, Cambridge, Massachusetts.,McGovern Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Gertrudis Perea
- Department of Functional and Systems Neurobiology, Instituto Cajal, CSIC, Madrid, Spain
| |
Collapse
|
31
|
Mansouri M, Strittmatter T, Fussenegger M. Light-Controlled Mammalian Cells and Their Therapeutic Applications in Synthetic Biology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1800952. [PMID: 30643713 PMCID: PMC6325585 DOI: 10.1002/advs.201800952] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/21/2018] [Indexed: 05/12/2023]
Abstract
The ability to remote control the expression of therapeutic genes in mammalian cells in order to treat disease is a central goal of synthetic biology-inspired therapeutic strategies. Furthermore, optogenetics, a combination of light and genetic sciences, provides an unprecedented ability to use light for precise control of various cellular activities with high spatiotemporal resolution. Recent work to combine optogenetics and therapeutic synthetic biology has led to the engineering of light-controllable designer cells, whose behavior can be regulated precisely and noninvasively. This Review focuses mainly on non-neural optogenetic systems, which are often used in synthetic biology, and their applications in genetic programing of mammalian cells. Here, a brief overview of the optogenetic tool kit that is available to build light-sensitive mammalian cells is provided. Then, recently developed strategies for the control of designer cells with specific biological functions are summarized. Recent translational applications of optogenetically engineered cells are also highlighted, ranging from in vitro basic research to in vivo light-controlled gene therapy. Finally, current bottlenecks, possible solutions, and future prospects for optogenetics in synthetic biology are discussed.
Collapse
Affiliation(s)
- Maysam Mansouri
- Department of Biosystems Science and EngineeringETH ZurichMattenstrasse 26CH‐4058BaselSwitzerland
| | - Tobias Strittmatter
- Department of Biosystems Science and EngineeringETH ZurichMattenstrasse 26CH‐4058BaselSwitzerland
| | - Martin Fussenegger
- Department of Biosystems Science and EngineeringETH ZurichMattenstrasse 26CH‐4058BaselSwitzerland
- Faculty of ScienceUniversity of BaselMattenstrasse 26CH‐4058BaselSwitzerland
| |
Collapse
|
32
|
Rodgers J, Peirson SN, Hughes S, Hankins MW. Functional characterisation of naturally occurring mutations in human melanopsin. Cell Mol Life Sci 2018; 75:3609-3624. [PMID: 29700553 PMCID: PMC6133154 DOI: 10.1007/s00018-018-2813-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/06/2018] [Accepted: 04/05/2018] [Indexed: 12/12/2022]
Abstract
Melanopsin is a blue light-sensitive opsin photopigment involved in a range of non-image forming behaviours, including circadian photoentrainment and the pupil light response. Many naturally occurring genetic variants exist within the human melanopsin gene (OPN4), yet it remains unclear how these variants affect melanopsin protein function and downstream physiological responses to light. Here, we have used bioinformatic analysis and in vitro expression systems to determine the functional phenotypes of missense human OPN4 variants. From 1242 human OPN4 variants collated in the NCBI Short Genetic Variation database (dbSNP), we identified 96 that lead to non-synonymous amino acid substitutions. These 96 missense mutations were screened using sequence alignment and comparative approaches to select 16 potentially deleterious variants for functional characterisation using calcium imaging of melanopsin-driven light responses in HEK293T cells. We identify several previously uncharacterised OPN4 mutations with altered functional properties, including attenuated or abolished light responses, as well as variants demonstrating abnormal response kinetics. These data provide valuable insight into the structure-function relationships of human melanopsin, including several key functional residues of the melanopsin protein. The identification of melanopsin variants with significantly altered function may serve to detect individuals with disrupted melanopsin-based light perception, and potentially highlight those at increased risk of sleep disturbance, circadian dysfunction, and visual abnormalities.
Collapse
Affiliation(s)
- Jessica Rodgers
- Nuffield Laboratory of Ophthalmology, Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, Sir William Dunn School of Pathology, University of Oxford, OMPI G, South Parks Road, Oxford, OX1 3RE, UK
| | - Stuart N Peirson
- Nuffield Laboratory of Ophthalmology, Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, Sir William Dunn School of Pathology, University of Oxford, OMPI G, South Parks Road, Oxford, OX1 3RE, UK
| | - Steven Hughes
- Nuffield Laboratory of Ophthalmology, Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, Sir William Dunn School of Pathology, University of Oxford, OMPI G, South Parks Road, Oxford, OX1 3RE, UK.
| | - Mark W Hankins
- Nuffield Laboratory of Ophthalmology, Sleep and Circadian Neuroscience Institute, Nuffield Department of Clinical Neurosciences, Sir William Dunn School of Pathology, University of Oxford, OMPI G, South Parks Road, Oxford, OX1 3RE, UK.
| |
Collapse
|
33
|
Stachurska A, Sarna T. Regulation of Melanopsin Signaling: Key Interactions of the Nonvisual Photopigment. Photochem Photobiol 2018; 95:83-94. [DOI: 10.1111/php.12995] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/26/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Anna Stachurska
- Labolatory of Imaging and Force Spectroscopy; Malopolska Centre of Biotechnology; Jagiellonian University; Krakow Poland
| | - Tadeusz Sarna
- Department of Biophysics; Faculty of Biochemistry, Biophysics and Biotechnology; Jagiellonian University; Krakow Poland
| |
Collapse
|
34
|
Katz B, Minke B. The Drosophila light-activated TRP and TRPL channels - Targets of the phosphoinositide signaling cascade. Prog Retin Eye Res 2018; 66:200-219. [DOI: 10.1016/j.preteyeres.2018.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/29/2018] [Accepted: 05/02/2018] [Indexed: 01/28/2023]
|
35
|
Rodgers J, Hughes S, Pothecary CA, Brown LA, Hickey DG, Peirson SN, Hankins MW. Defining the impact of melanopsin missense polymorphisms using in vivo functional rescue. Hum Mol Genet 2018; 27:2589-2603. [PMID: 29718372 PMCID: PMC6048994 DOI: 10.1093/hmg/ddy150] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 04/11/2018] [Accepted: 04/16/2018] [Indexed: 12/02/2022] Open
Abstract
Melanopsin (OPN4) is an opsin photopigment expressed within intrinsically photosensitive retinal ganglion cells (ipRGCs) that mediate non-image forming (NIF) responses to light. Two single-nucleotide polymorphisms (SNPs) in human melanopsin (hOPN4), Pro10Leu and Thr394Ile, have recently been associated with abnormal NIF responses to light, including seasonal affective disorder. It has been suggested these behavioural changes are due to altered melanopsin signalling. However, there is currently no direct evidence to support this. Here we have used ipRGC-specific delivery of hOPN4 wild-type (WT), Pro10Leu or Thr394Ile adeno-associated viruses (AAV) to determine the functional consequences of hOPN4 SNPs on melanopsin-driven light responses and associated behaviours. Immunohistochemistry confirmed hOPN4 AAVs exclusively transduced mouse ipRGCs. Behavioural phenotyping performed before and after AAV injection demonstrated that both hOPN4 Pro10Leu and Thr394Ile could functionally rescue pupillary light responses and circadian photoentrainment in Opn4-/- mice, with no differences in NIF behaviours detected for animals expressing either SNP compared to hOPN4 WT. Multi-electrode array recordings revealed that ipRGCs expressing hOPN4 Thr394Ile exhibit melanopsin-driven light responses with significantly attenuated response amplitude, decreased sensitivity and faster offset kinetics compared to hOPN4 WT. IpRGCs expressing hOpn4 Pro10Leu also showed reduced response amplitude. Collectively these data suggest Thr394Ile and Pro10Leu may be functionally significant SNPs, which result in altered melanopsin signalling. To our knowledge, this study provides the first direct evidence for the effects of hOPN4 polymorphisms on melanopsin-driven light responses and NIF behaviours in vivo, providing further insight into the role of these SNPs in melanopsin function and human physiology.
Collapse
Affiliation(s)
- Jessica Rodgers
- Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute, OMPI G, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Steven Hughes
- Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute, OMPI G, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Carina A Pothecary
- Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute, OMPI G, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Laurence A Brown
- Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute, OMPI G, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Doron G Hickey
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Stuart N Peirson
- Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute, OMPI G, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Mark W Hankins
- Nuffield Department of Clinical Neurosciences, Sleep and Circadian Neuroscience Institute, OMPI G, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
36
|
Melanopsin mRNA in the Iris of Red-Eared Slider Turtles (Trachemys scripta elegans). J HERPETOL 2017. [DOI: 10.1670/16-046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
37
|
Abstract
In this issue of Neuron, Mure et al. (2016) demonstrate that two mechanisms-phosphorylation of a C-terminal intracellular region, and mechanism involving the whole of the C terminus-oppositely shape the kinetics and sensitivity of the nonvisual photoreceptor melanopsin.
Collapse
Affiliation(s)
- Russell N Van Gelder
- Department of Ophthalmology, University of Washington School of Medicine, Seattle, WA 98104, USA; Department of Biological Structure, University of Washington School of Medicine, Seattle, WA 98104, USA; Department of Pathology, University of Washington School of Medicine, Seattle, WA 98104, USA.
| | - Ethan D Buhr
- Department of Ophthalmology, University of Washington School of Medicine, Seattle, WA 98104, USA
| |
Collapse
|
38
|
Yasin B, Kohn E, Peters M, Zaguri R, Weiss S, Schopf K, Katz B, Huber A, Minke B. Ectopic Expression of Mouse Melanopsin in Drosophila Photoreceptors Reveals Fast Response Kinetics and Persistent Dark Excitation. J Biol Chem 2017; 292:3624-3636. [PMID: 28119450 PMCID: PMC5339748 DOI: 10.1074/jbc.m116.754770] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 01/19/2017] [Indexed: 01/10/2023] Open
Abstract
The intrinsically photosensitive M1 retinal ganglion cells (ipRGC) initiate non-image-forming light-dependent activities and express the melanopsin (OPN4) photopigment. Several features of ipRGC photosensitivity are characteristic of fly photoreceptors. However, the light response kinetics of ipRGC is much slower due to unknown reasons. Here we used transgenic Drosophila, in which the mouse OPN4 replaced the native Rh1 photopigment of Drosophila R1-6 photoreceptors, resulting in deformed rhabdomeric structure. Immunocytochemistry revealed OPN4 expression at the base of the rhabdomeres, mainly at the rhabdomeral stalk. Measurements of the early receptor current, a linear manifestation of photopigment activation, indicated large expression of OPN4 in the plasma membrane. Comparing the early receptor current amplitude and action spectra between WT and the Opn4-expressing Drosophila further indicated that large quantities of a blue absorbing photopigment were expressed, having a dark stable blue intermediate state. Strikingly, the light-induced current of the Opn4-expressing fly photoreceptors was ∼40-fold faster than that of ipRGC. Furthermore, an intense white flash induced a small amplitude prolonged dark current composed of discrete unitary currents similar to the Drosophila single photon responses. The induction of prolonged dark currents by intense blue light could be suppressed by a following intense green light, suggesting induction and suppression of prolonged depolarizing afterpotential. This is the first demonstration of heterologous functional expression of mammalian OPN4 in the genetically emendable Drosophila photoreceptors. Moreover, the fast OPN4-activated ionic current of Drosophila photoreceptors relative to that of mouse ipRGC, indicates that the slow light response of ipRGC does not arise from an intrinsic property of melanopsin.
Collapse
Affiliation(s)
- Bushra Yasin
- From the Department of Medical Neurobiology, Institute for Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel and
| | - Elkana Kohn
- From the Department of Medical Neurobiology, Institute for Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel and
| | - Maximilian Peters
- From the Department of Medical Neurobiology, Institute for Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel and
| | - Rachel Zaguri
- From the Department of Medical Neurobiology, Institute for Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel and
| | - Shirley Weiss
- From the Department of Medical Neurobiology, Institute for Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel and
| | - Krystina Schopf
- the Department of Biosensorics, Institute of Physiology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Ben Katz
- From the Department of Medical Neurobiology, Institute for Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel and
| | - Armin Huber
- the Department of Biosensorics, Institute of Physiology, University of Hohenheim, 70599 Stuttgart, Germany
| | - Baruch Minke
- From the Department of Medical Neurobiology, Institute for Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel and
| |
Collapse
|
39
|
C-terminal phosphorylation regulates the kinetics of a subset of melanopsin-mediated behaviors in mice. Proc Natl Acad Sci U S A 2017; 114:2741-2746. [PMID: 28223508 DOI: 10.1073/pnas.1611893114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) express the photopigment melanopsin and mediate several non-image-forming visual functions, including circadian photoentrainment and the pupillary light reflex (PLR). ipRGCs act as autonomous photoreceptors via the intrinsic melanopsin-based phototransduction pathway and as a relay for rod/cone input via synaptically driven responses. Under low light intensities, where only synaptically driven rod/cone input activates ipRGCs, the duration of the ipRGC response will be determined by the termination kinetics of the rod/cone circuits. Little is known, however, about the termination kinetics of the intrinsic melanopsin-based phototransduction pathway and its contribution to several melanopsin-mediated behaviors. Here, we show that C-terminal phosphorylation of melanopsin determines the recovery kinetics of the intrinsic melanopsin-based photoresponse in ipRGCs, the duration of the PLR, and the speed of reentrainment. In contrast, circadian phase alignment and direct effects of light on activity (masking) are not influenced by C-terminal phosphorylation of melanopsin. Electrophysiological measurements demonstrate that expression of a virally encoded melanopsin lacking all C-terminal phosphorylation sites (C terminus phosphonull) leads to a prolonged intrinsic light response. In addition, mice expressing the C terminus phosphonull in ipRGCs reentrain faster to a delayed light/dark cycle compared with mice expressing virally encoded WT melanopsin; however, the phase angle of entrainment and masking were indistinguishable. Importantly, a sustained PLR in the phosphonull animals is only observed at brighter light intensities that activate melanopsin phototransduction, but not at dimmer light intensities that activate only the rod/cone pathway. Taken together, our results highlight how the kinetics of the melanopsin photoresponse differentially regulate distinct light-mediated behaviors.
Collapse
|
40
|
Zhao X, Reifler AN, Schroeder MM, Jaeckel ER, Chervenak AP, Wong KY. Mechanisms creating transient and sustained photoresponses in mammalian retinal ganglion cells. J Gen Physiol 2017; 149:335-353. [PMID: 28153865 PMCID: PMC5339512 DOI: 10.1085/jgp.201611720] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/24/2016] [Accepted: 12/30/2016] [Indexed: 11/20/2022] Open
Abstract
Visual stimuli of different frequencies are encoded in the retina using transient and sustained responses. Zhao et al. describe the different strategies that are used by four types of retinal ganglion cells to shape photoresponse kinetics. Retinal neurons use sustained and transient light responses to encode visual stimuli of different frequency ranges, but the underlying mechanisms remain poorly understood. In particular, although earlier studies in retinal ganglion cells (RGCs) proposed seven potential mechanisms, all seven have since been disputed, and it remains unknown whether different RGC types use different mechanisms or how many mechanisms are used by each type. Here, we conduct a comprehensive survey in mice and rats of 12 candidate mechanisms that could conceivably produce tonic rod/cone-driven ON responses in intrinsically photosensitive RGCs (ipRGCs) and transient ON responses in three types of direction-selective RGCs (TRHR+, Hoxd10+ ON, and Hoxd10+ ON-OFF cells). We find that the tonic kinetics of ipRGCs arises from their substantially above-threshold resting potentials, input from sustained ON bipolar cells, absence of amacrine cell inhibition of presynaptic ON bipolar cells, and mGluR7-mediated maintenance of light-evoked glutamatergic input. All three types of direction-selective RGCs receive input from transient ON bipolar cells, and each type uses additional strategies to promote photoresponse transience: presynaptic inhibition and dopaminergic modulation for TRHR+ cells, center/surround antagonism and relatively negative resting potentials for Hoxd10+ ON cells, and presynaptic inhibition for Hoxd10+ ON-OFF cells. We find that the sustained nature of ipRGCs’ rod/cone-driven responses depends neither on melanopsin nor on N-methyl-d-aspartate (NMDA) receptors, whereas the transience of the direction-selective cells’ responses is influenced neither by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/kainate receptor desensitization nor by glutamate uptake. For all cells, we further rule out spike frequency adaptation and intracellular Ca2+ as determinants of photoresponse kinetics. In conclusion, different RGC types use diverse mechanisms to produce sustained or transient light responses. Parenthetically, we find evidence in both mice and rats that the kinetics of light-induced mGluR6 deactivation determines whether an ON bipolar cell responds tonically or transiently to light.
Collapse
Affiliation(s)
- Xiwu Zhao
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105
| | - Aaron N Reifler
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105
| | - Melanie M Schroeder
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105
| | - Elizabeth R Jaeckel
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105
| | - Andrew P Chervenak
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105
| | - Kwoon Y Wong
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105 .,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48105
| |
Collapse
|