1
|
Avilés EC, Wang SK, Patel S, Cordero S, Shi S, Lin L, Kefalov VJ, Goodrich LV, Cepko CL, Xue Y. ERG responses to high-frequency flickers require FAT3 signaling in mouse retinal bipolar cells. J Gen Physiol 2025; 157:e202413642. [PMID: 39903280 PMCID: PMC11793021 DOI: 10.1085/jgp.202413642] [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: 07/12/2024] [Revised: 11/17/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
Vision is initiated by the reception of light by photoreceptors and subsequent processing via downstream retinal neurons. Proper circuit organization depends on the multifunctional tissue polarity protein FAT3, which is required for amacrine cell connectivity and retinal lamination. Here, we investigated the retinal function of Fat3 mutant mice and found decreases in both electroretinography and perceptual responses to high-frequency flashes. These defects did not correlate with abnormal amacrine cell wiring, pointing instead to a role in bipolar cell subtypes that also express FAT3. The role of FAT3 in the response to high temporal frequency flashes depends upon its ability to transduce an intracellular signal. Mechanistically, FAT3 binds to the synaptic protein PTPσ intracellularly and is required to localize GRIK1 to OFF-cone bipolar cell synapses with cone photoreceptors. These findings expand the repertoire of FAT3's functions and reveal its importance in bipolar cells for high-frequency light response.
Collapse
Affiliation(s)
- Evelyn C. Avilés
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sean K. Wang
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Sarina Patel
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Sebastian Cordero
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Shuxiang Shi
- Lingang Laboratory, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lucas Lin
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Vladimir J. Kefalov
- Gavin Herbert Eye Institute and Center for Translational Vision Research, University of California, Irvine, Irvine, CA, USA
| | - Lisa V. Goodrich
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Constance L. Cepko
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Yunlu Xue
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA, USA
- Lingang Laboratory, Shanghai, China
| |
Collapse
|
2
|
Kim SY, Park CH, Moon BH, Seabold GK. Murine Retina Outer Plexiform Layer Development and Transcriptome Analysis of Pre-Synapses in Photoreceptors. Life (Basel) 2024; 14:1103. [PMID: 39337887 PMCID: PMC11433150 DOI: 10.3390/life14091103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Photoreceptors in the mammalian retina convert light signals into electrical and molecular signals through phototransduction and transfer the visual inputs to second-order neurons via specialized ribbon synapses. Two kinds of photoreceptors, rods and cones, possess distinct morphology and function. Currently, we have limited knowledge about rod versus (vs.) cone synapse development and the associated genes. The transcription factor neural retina leucine zipper (NRL) determines the rod vs. cone photoreceptor cell fate and is critical for rod differentiation. Nrl knockout mice fail to form rods, generating all cone or S-cone-like (SCL) photoreceptors in the retina, whereas ectopic expression of Nrl using a cone-rod homeobox (Crx) promoter (CrxpNrl) forms all rods. Here, we examined rod and cone pre-synapse development, including axonal elongation, terminal shaping, and synaptic lamination in the outer plexiform layer (OPL) in the presence or absence of Nrl. We show that NRL loss and knockdown result in delayed OPL maturation and plasticity with aberrant dendrites of bipolar neurons. The integrated analyses of the transcriptome in developing rods and SCLs with NRL CUT&RUN and synaptic gene ontology analyses identified G protein subunit beta (Gnb) 1 and p21 (RAC1) activated kinase 5 (Pak5 or Pak7) transcripts were upregulated in developing rods and down-regulated in developing SCLs. Notably, Gnb1 and Gnb5 are rod dominant, and Gnb3 is enriched in cones. NRL binds to the genes of Gnb1, Gnb3, and Gnb5. NRL also regulates pre-synapse ribbon genes, and their expression is altered in rods and SCLs. Our study of histological and gene analyses provides new insights into the morphogenesis of photoreceptor pre-synapse development and regulation of associated genes in the developing retina.
Collapse
Affiliation(s)
- Soo-Young Kim
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christine Haewon Park
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bo-Hyun Moon
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Gail K Seabold
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
3
|
Stevens-Sostre WA, Hoon M. Cellular and Molecular Mechanisms Regulating Retinal Synapse Development. Annu Rev Vis Sci 2024; 10:377-402. [PMID: 39292551 PMCID: PMC12022667 DOI: 10.1146/annurev-vision-102122-105721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Synapse formation within the retinal circuit ensures that distinct neuronal types can communicate efficiently to process visual signals. Synapses thus form the core of the visual computations performed by the retinal circuit. Retinal synapses are diverse but can be broadly categorized into multipartner ribbon synapses and 1:1 conventional synapses. In this article, we review our current understanding of the cellular and molecular mechanisms that regulate the functional establishment of mammalian retinal synapses, including the role of adhesion proteins, synaptic proteins, extracellular matrix and cytoskeletal-associated proteins, and activity-dependent cues. We outline future directions and areas of research that will expand our knowledge of these mechanisms. Understanding the regulators moderating synapse formation and function not only reveals the integrated developmental processes that establish retinal circuits, but also divulges the identity of mechanisms that could be engaged during disease and degeneration.
Collapse
Affiliation(s)
- Whitney A Stevens-Sostre
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA;
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mrinalini Hoon
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA;
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
4
|
Avilés EC, Wang SK, Patel S, Shi S, Lin L, Kefalov VJ, Goodrich LV, Cepko CL, Xue Y. High temporal frequency light response in mouse retina requires FAT3 signaling in bipolar cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.02.565326. [PMID: 37961274 PMCID: PMC10635074 DOI: 10.1101/2023.11.02.565326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Vision is initiated by the reception of light by photoreceptors and subsequent processing via downstream retinal neurons. Proper cellular organization depends on the multi-functional tissue polarity protein FAT3, which is required for amacrine cell connectivity and retinal lamination. Here we investigated the retinal function of Fat3 mutant mice and found decreases in physiological and perceptual responses to high frequency flashes. These defects did not correlate with abnormal amacrine cell wiring, pointing instead to a role in bipolar cell subtypes that also express FAT3. The role of FAT3 in the response to high temporal frequency flashes depends upon its ability to transduce an intracellular signal. Mechanistically, FAT3 binds to the synaptic protein PTPσ, intracellularly, and is required to localize GRIK1 to OFF-cone bipolar cell synapses with cone photoreceptors. These findings expand the repertoire of FAT3's functions and reveal its importance in bipolar cells for high frequency light response.
Collapse
Affiliation(s)
- Evelyn C. Avilés
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
- Facultad de Ciencias Biologicas, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Sean K. Wang
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA 02115
- Howard Hughes Medical Institute, Boston, MA 02115
| | - Sarina Patel
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Shuxiang Shi
- Lingang Laboratory, Shanghai, China, 200031
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China, 201210
| | - Lucas Lin
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA 02115
| | - Vladimir J. Kefalov
- Gavin Herbert Eye Institute & Center for Translational Vision Research, University of California, Irvine, CA 92697
| | - Lisa V. Goodrich
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Constance L. Cepko
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA 02115
- Howard Hughes Medical Institute, Boston, MA 02115
| | - Yunlu Xue
- Departments of Genetics and Ophthalmology, Harvard Medical School, Boston, MA 02115
- Lingang Laboratory, Shanghai, China, 200031
- Lead contact
| |
Collapse
|
5
|
Hanke-Gogokhia C, Zapadka TE, Finkelstein S, Klingeborn M, Maugel TK, Singer JH, Arshavsky VY, Demb JB. The Structural and Functional Integrity of Rod Photoreceptor Ribbon Synapses Depends on Redundant Actions of Dynamins 1 and 3. J Neurosci 2024; 44:e1379232024. [PMID: 38641407 PMCID: PMC11209669 DOI: 10.1523/jneurosci.1379-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 04/02/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024] Open
Abstract
Vertebrate vision begins with light absorption by rod and cone photoreceptors, which transmit signals from their synaptic terminals to second-order neurons: bipolar and horizontal cells. In mouse rods, there is a single presynaptic ribbon-type active zone at which the release of glutamate occurs tonically in the dark. This tonic glutamatergic signaling requires continuous exo- and endocytosis of synaptic vesicles. At conventional synapses, endocytosis commonly requires dynamins: GTPases encoded by three genes (Dnm1-3), which perform membrane scission. Disrupting endocytosis by dynamin deletions impairs transmission at conventional synapses, but the impact of disrupting endocytosis and the role(s) of specific dynamin isoforms at rod ribbon synapses are understood incompletely. Here, we used cell-specific knock-outs (KOs) of the neuron-specific Dnm1 and Dnm3 to investigate the functional roles of dynamin isoforms in rod photoreceptors in mice of either sex. Analysis of synaptic protein expression, synapse ultrastructure, and retinal function via electroretinograms (ERGs) showed that dynamins 1 and 3 act redundantly and are essential for supporting the structural and functional integrity of rod ribbon synapses. Single Dnm3 KO showed no phenotype, and single Dnm1 KO only modestly reduced synaptic vesicle density without affecting vesicle size and overall synapse integrity, whereas double Dnm1/Dnm3 KO impaired vesicle endocytosis profoundly, causing enlarged vesicles, reduced vesicle density, reduced ERG responses, synaptic terminal degeneration, and disassembly and degeneration of postsynaptic processes. Concurrently, cone function remained intact. These results show the fundamental redundancy of dynamins 1 and 3 in regulating the structure and function of rod ribbon synapses.
Collapse
Affiliation(s)
- Christin Hanke-Gogokhia
- Departments of Ophthalmology & Visual Science, Yale University, New Haven, Connecticut 06511
| | - Thomas E Zapadka
- Departments of Ophthalmology & Visual Science, Yale University, New Haven, Connecticut 06511
- Cellular & Molecular Physiology, Yale University, New Haven, Connecticut 06511
| | - Stella Finkelstein
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina 27705
| | - Mikael Klingeborn
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina 27705
| | - Timothy K Maugel
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | - Joshua H Singer
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina 27705
| | - Jonathan B Demb
- Departments of Ophthalmology & Visual Science, Yale University, New Haven, Connecticut 06511
- Cellular & Molecular Physiology, Yale University, New Haven, Connecticut 06511
- Department of Neuroscience, Yale University, New Haven, Connecticut 06511
- Wu Tsai Institute, Yale University, New Haven, Connecticut 06511
| |
Collapse
|
6
|
Hölzel MB, Kamermans W, Winkelman BHJ, Howlett MHC, De Zeeuw CI, Kamermans M. A common cause for nystagmus in different congenital stationary night blindness mouse models. J Physiol 2023; 601:5317-5340. [PMID: 37864560 DOI: 10.1113/jp284965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 09/22/2023] [Indexed: 10/23/2023] Open
Abstract
In Nyxnob mice, a model for congenital nystagmus associated with congenital stationary night blindness (CSNB), synchronous oscillating retinal ganglion cells (RGCs) lead to oscillatory eye movements, i.e. nystagmus. Given the specific expression of mGluR6 and Cav 1.4 in the photoreceptor to bipolar cell synapses, as well as their clinical association with CSNB, we hypothesize that Grm6nob3 and Cav 1.4-KO mutants show, like the Nyxnob mouse, oscillations in both their RGC activity and eye movements. Using multi-electrode array recordings of RGCs and measurements of the eye movements, we demonstrate that Grm6nob3 and Cav 1.4-KO mice also show oscillations of their RGCs as well as a nystagmus. Interestingly, the preferred frequencies of RGC activity as well as the eye movement oscillations of the Grm6nob3 , Cav 1.4-KO and Nyxnob mice differ among mutants, but the neuronal activity and eye movement behaviour within a strain remain aligned in the same frequency domain. Model simulations indicate that mutations affecting the photoreceptor-bipolar cell synapse can form a common cause of the nystagmus of CSNB by driving oscillations in RGCs via AII amacrine cells. KEY POINTS: In Nyxnob mice, a model for congenital nystagmus associated with congenital stationary night blindness (CSNB), their oscillatory eye movements (i.e. nystagmus) are caused by synchronous oscillating retinal ganglion cells. Here we show that the same mechanism applies for two other CSNB mouse models - Grm6nob3 and Cav 1.4-KO mice. We propose that the retinal ganglion cell oscillations originate in the AII amacrine cells. Model simulations show that by only changing the input to ON-bipolar cells, all phenotypical differences between the various genetic mouse models can be reproduced.
Collapse
Affiliation(s)
- Maj-Britt Hölzel
- Netherlands Institute for Neuroscience Amsterdam, Amsterdam, the Netherlands
| | - Wouter Kamermans
- Netherlands Institute for Neuroscience Amsterdam, Amsterdam, the Netherlands
| | - Beerend H J Winkelman
- Netherlands Institute for Neuroscience Amsterdam, Amsterdam, the Netherlands
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Marcus H C Howlett
- Netherlands Institute for Neuroscience Amsterdam, Amsterdam, the Netherlands
| | - Chris I De Zeeuw
- Netherlands Institute for Neuroscience Amsterdam, Amsterdam, the Netherlands
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Maarten Kamermans
- Netherlands Institute for Neuroscience Amsterdam, Amsterdam, the Netherlands
- Department of Biomedical Physics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Yadav P, Podia M, Kumari SP, Mani I. Glutamate receptor endocytosis and signaling in neurological conditions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:167-207. [PMID: 36813358 DOI: 10.1016/bs.pmbts.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The non-essential amino acid glutamate acts as a major excitatory neurotransmitter and plays a significant role in the central nervous system (CNS). It binds with two different types of receptors, ionotropic glutamate receptors (iGluRs) and metabotropic glutamate receptors (mGluRs), responsible for the postsynaptic excitation of neurons. They are important for memory, neural development and communication, and learning. Endocytosis and subcellular trafficking of the receptor are essential for the regulation of receptor expression on the cell membrane and excitation of the cells. The endocytosis and trafficking of the receptor are dependent on its type, ligand, agonist, and antagonist present. This chapter discusses the types of glutamate receptors, their subtypes, and the regulation of their internalization and trafficking. The roles of glutamate receptors in neurological diseases are also briefly discussed.
Collapse
Affiliation(s)
- Prerna Yadav
- Department of Microbiology, University of Delhi, New Delhi, India
| | - Mansi Podia
- Department of Microbiology, University of Delhi, New Delhi, India
| | - Shashi Prabha Kumari
- Department of Microbiology, Ram Lal Anand College, University of Delhi, New Delhi, India
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
8
|
Križaj D, Cordeiro S, Strauß O. Retinal TRP channels: Cell-type-specific regulators of retinal homeostasis and multimodal integration. Prog Retin Eye Res 2023; 92:101114. [PMID: 36163161 PMCID: PMC9897210 DOI: 10.1016/j.preteyeres.2022.101114] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 02/05/2023]
Abstract
Transient receptor potential (TRP) channels are a widely expressed family of 28 evolutionarily conserved cationic ion channels that operate as primary detectors of chemical and physical stimuli and secondary effectors of metabotropic and ionotropic receptors. In vertebrates, the channels are grouped into six related families: TRPC, TRPV, TRPM, TRPA, TRPML, and TRPP. As sensory transducers, TRP channels are ubiquitously expressed across the body and the CNS, mediating critical functions in mechanosensation, nociception, chemosensing, thermosensing, and phototransduction. This article surveys current knowledge about the expression and function of the TRP family in vertebrate retinas, which, while dedicated to transduction and transmission of visual information, are highly susceptible to non-visual stimuli. Every retinal cell expresses multiple TRP subunits, with recent evidence establishing their critical roles in paradigmatic aspects of vertebrate vision that include TRPM1-dependent transduction of ON bipolar signaling, TRPC6/7-mediated ganglion cell phototransduction, TRP/TRPL phototransduction in Drosophila and TRPV4-dependent osmoregulation, mechanotransduction, and regulation of inner and outer blood-retina barriers. TRP channels tune light-dependent and independent functions of retinal circuits by modulating the intracellular concentration of the 2nd messenger calcium, with emerging evidence implicating specific subunits in the pathogenesis of debilitating diseases such as glaucoma, ocular trauma, diabetic retinopathy, and ischemia. Elucidation of TRP channel involvement in retinal biology will yield rewards in terms of fundamental understanding of vertebrate vision and therapeutic targeting to treat diseases caused by channel dysfunction or over-activation.
Collapse
Affiliation(s)
- David Križaj
- Departments of Ophthalmology, Neurobiology, and Bioengineering, University of Utah, Salt Lake City, USA
| | - Soenke Cordeiro
- Institute of Physiology, Faculty of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Olaf Strauß
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, a Corporate Member of Freie Universität, Humboldt-University, The Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
9
|
Cao Y, Fajardo D, Guerrero-Given D, Samuel MA, Ohtsuka T, Boye SE, Kamasawa N, Martemyanov KA. Post-developmental plasticity of the primary rod pathway allows restoration of visually guided behaviors. Curr Biol 2022; 32:4783-4796.e3. [PMID: 36179691 PMCID: PMC9691582 DOI: 10.1016/j.cub.2022.09.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/17/2022] [Accepted: 09/08/2022] [Indexed: 01/24/2023]
Abstract
The formation of neural circuits occurs in a programmed fashion, but proper activity in the circuit is essential for refining the organization necessary for driving complex behavioral tasks. In the retina, sensory deprivation during the critical period of development is well known to perturb the organization of the visual circuit making the animals unable to use vision for behavior. However, the extent of plasticity, molecular factors involved, and malleability of individual channels in the circuit to manipulations outside of the critical period are not well understood. In this study, we selectively disconnected and reconnected rod photoreceptors in mature animals after completion of the retina circuit development. We found that introducing synaptic rod photoreceptor input post-developmentally allowed their integration into the circuit both anatomically and functionally. Remarkably, adult mice with newly integrated rod photoreceptors gained high-sensitivity vision, even when it was absent from birth. These observations reveal plasticity of the retina circuit organization after closure of the critical period and encourage the development of vision restoration strategies for congenital blinding disorders.
Collapse
Affiliation(s)
- Yan Cao
- Department of Neuroscience, UF Scripps Biomedical Research, Jupiter, FL 33458, USA
| | - Diego Fajardo
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Debbie Guerrero-Given
- The Imaging Center, Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Melanie A Samuel
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Toshihisa Ohtsuka
- Department of Biochemistry, Graduate School of Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Shannon E Boye
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Naomi Kamasawa
- The Imaging Center, Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, UF Scripps Biomedical Research, Jupiter, FL 33458, USA.
| |
Collapse
|
10
|
Ganczer A, Szarka G, Balogh M, Hoffmann G, Tengölics ÁJ, Kenyon G, Kovács-Öller T, Völgyi B. Transience of the Retinal Output Is Determined by a Great Variety of Circuit Elements. Cells 2022; 11:cells11050810. [PMID: 35269432 PMCID: PMC8909309 DOI: 10.3390/cells11050810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
Abstract
Retinal ganglion cells (RGCs) encrypt stimulus features of the visual scene in action potentials and convey them toward higher visual centers in the brain. Although there are many visual features to encode, our recent understanding is that the ~46 different functional subtypes of RGCs in the retina share this task. In this scheme, each RGC subtype establishes a separate, parallel signaling route for a specific visual feature (e.g., contrast, the direction of motion, luminosity), through which information is conveyed. The efficiency of encoding depends on several factors, including signal strength, adaptational levels, and the actual efficacy of the underlying retinal microcircuits. Upon collecting inputs across their respective receptive field, RGCs perform further analysis (e.g., summation, subtraction, weighting) before they generate the final output spike train, which itself is characterized by multiple different features, such as the number of spikes, the inter-spike intervals, response delay, and the rundown time (transience) of the response. These specific kinetic features are essential for target postsynaptic neurons in the brain in order to effectively decode and interpret signals, thereby forming visual perception. We review recent knowledge regarding circuit elements of the mammalian retina that participate in shaping RGC response transience for optimal visual signaling.
Collapse
Affiliation(s)
- Alma Ganczer
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Gergely Szarka
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Márton Balogh
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Gyula Hoffmann
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Ádám Jonatán Tengölics
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Garrett Kenyon
- Los Alamos National Laboratory, Computer & Computational Science Division, Los Alamos, NM 87545, USA;
| | - Tamás Kovács-Öller
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Hungary; (A.G.); (G.S.); (M.B.); (G.H.); (Á.J.T.); (T.K.-Ö.)
- Department of Experimental Zoology and Neurobiology, University of Pécs, H-7624 Pécs, Hungary
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, H-7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, H-7624 Pécs, Hungary
- Correspondence:
| |
Collapse
|
11
|
Tsukamoto Y, Iseki K, Omi N. Helical Fasciculation of Bipolar and Horizontal Cell Neurites for Wiring With Photoreceptors in Macaque and Mouse Retinas. Invest Ophthalmol Vis Sci 2021; 62:31. [PMID: 33507230 PMCID: PMC7846946 DOI: 10.1167/iovs.62.1.31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The three-dimensional configurations of rod and cone bipolar cell (BC) dendrites and horizontal cell (HC) processes outside rod and cone synaptic terminals have not been fully elucidated. We reveal how these neurites are mutually arranged to coordinate formation and maintenance of the postsynaptic complex of ribbon synapses in mouse and monkey retinas. Methods Serial section transmission electron microscopy was utilized to reconstruct BC and HC neurites in macaque monkey and mouse, including metabotropic glutamate receptor 6 (mGluR6)-knockout mice. Results Starting from sporadically distributed branching points, rod BC and HC neurites (B and H, respectively) took specific paths to rod spherules by gradually adjusting their mutual positions, which resulted in a closed alternating pattern of H‒B‒H‒B neurites at the rod spherule aperture. This order corresponded to the array of elements constituting the postsynaptic complex of ribbon synapses. We identified novel helical coils of HC processes surrounding the rod BC dendrite in both mouse and macaque retinas, and these structures occurred more frequently in mGluR6-knockout than wild-type mouse retinas. Horizontal cell processes also formed hook-like protrusions that encircled cone BC and HC neurites below the cone pedicles in the macaque retina. Conclusions Bipolar and horizontal cell neurites take specific paths to adjust their mutual positions at the rod spherule aperture. Some HC processes are helically coiled around rod BC dendrites or form hook-like protrusions around cone BC dendrites and HC processes. Loss of mGluR6 signaling may be one factor promoting unbalanced neurite growth and compensatory neurite coiling.
Collapse
Affiliation(s)
- Yoshihiko Tsukamoto
- Department of Biology, Hyogo College of Medicine, Mukogawa, Nishinomiya, Hyogo, Japan.,Studio EM-Retina, Satonaka, Nishinomiya, Hyogo, Japan
| | - Kyoko Iseki
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, Japan
| | - Naoko Omi
- Studio EM-Retina, Satonaka, Nishinomiya, Hyogo, Japan
| |
Collapse
|
12
|
Fina ME, Wang J, Nikonov SS, Sterling S, Vardi N, Kashina A, Dong DW. Arginyltransferase (Ate1) regulates the RGS7 protein level and the sensitivity of light-evoked ON-bipolar responses. Sci Rep 2021; 11:9376. [PMID: 33931669 PMCID: PMC8087773 DOI: 10.1038/s41598-021-88628-3] [Citation(s) in RCA: 5] [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: 02/05/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Regulator of G-protein signaling 7 (RGS7) is predominately present in the nervous system and is essential for neuronal signaling involving G-proteins. Prior studies in cultured cells showed that RGS7 is regulated via proteasomal degradation, however no protein is known to facilitate proteasomal degradation of RGS7 and it has not been shown whether this regulation affects G-protein signaling in neurons. Here we used a knockout mouse model with conditional deletion of arginyltransferase (Ate1) in the nervous system and found that in retinal ON bipolar cells, where RGS7 modulates a G-protein to signal light increments, deletion of Ate1 raised the level of RGS7. Electroretinographs revealed that lack of Ate1 leads to increased light-evoked response sensitivities of ON-bipolar cells, as well as their downstream neurons. In cultured mouse embryonic fibroblasts (MEF), RGS7 was rapidly degraded via proteasome pathway and this degradation was abolished in Ate1 knockout MEF. Our results indicate that Ate1 regulates RGS7 protein level by facilitating proteasomal degradation of RGS7 and thus affects G-protein signaling in neurons.
Collapse
Affiliation(s)
- Marie E Fina
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Junling Wang
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sergei S Nikonov
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Stephanie Sterling
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Noga Vardi
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anna Kashina
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Dawei W Dong
- Department of Biomedical Sciences, School of Veterinary Medicines, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
13
|
Orhan E, Neuillé M, de Sousa Dias M, Pugliese T, Michiels C, Condroyer C, Antonio A, Sahel JA, Audo I, Zeitz C. A New Mouse Model for Complete Congenital Stationary Night Blindness Due to Gpr179 Deficiency. Int J Mol Sci 2021; 22:ijms22094424. [PMID: 33922602 PMCID: PMC8122890 DOI: 10.3390/ijms22094424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 01/24/2023] Open
Abstract
Mutations in GPR179 lead to autosomal recessive complete congenital stationary night blindness (cCSNB). This condition represents a signal transmission defect from the photoreceptors to the ON-bipolar cells. To confirm the phenotype, better understand the pathogenic mechanism in vivo, and provide a model for therapeutic approaches, a Gpr179 knock-out mouse model was genetically and functionally characterized. We confirmed that the insertion of a neo/lac Z cassette in intron 1 of Gpr179 disrupts the same gene. Spectral domain optical coherence tomography reveals no obvious retinal structure abnormalities. Gpr179 knock-out mice exhibit a so-called no-b-wave (nob) phenotype with severely reduced b-wave amplitudes in the electroretinogram. Optomotor tests reveal decreased optomotor responses under scotopic conditions. Consistent with the genetic disruption of Gpr179, GPR179 is absent at the dendritic tips of ON-bipolar cells. While proteins of the same signal transmission cascade (GRM6, LRIT3, and TRPM1) are correctly localized, other proteins (RGS7, RGS11, and GNB5) known to regulate GRM6 are absent at the dendritic tips of ON-bipolar cells. These results add a new model of cCSNB, which is important to better understand the role of GPR179, its implication in patients with cCSNB, and its use for the development of therapies.
Collapse
Affiliation(s)
- Elise Orhan
- Institut de la Vision, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Sorbonne Université, F-75012 Paris, France; (E.O.); (M.N.); (M.d.S.D.); (T.P.); (C.M.); (C.C.); (A.A.); (J.-A.S.); (I.A.)
| | - Marion Neuillé
- Institut de la Vision, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Sorbonne Université, F-75012 Paris, France; (E.O.); (M.N.); (M.d.S.D.); (T.P.); (C.M.); (C.C.); (A.A.); (J.-A.S.); (I.A.)
| | - Miguel de Sousa Dias
- Institut de la Vision, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Sorbonne Université, F-75012 Paris, France; (E.O.); (M.N.); (M.d.S.D.); (T.P.); (C.M.); (C.C.); (A.A.); (J.-A.S.); (I.A.)
| | - Thomas Pugliese
- Institut de la Vision, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Sorbonne Université, F-75012 Paris, France; (E.O.); (M.N.); (M.d.S.D.); (T.P.); (C.M.); (C.C.); (A.A.); (J.-A.S.); (I.A.)
| | - Christelle Michiels
- Institut de la Vision, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Sorbonne Université, F-75012 Paris, France; (E.O.); (M.N.); (M.d.S.D.); (T.P.); (C.M.); (C.C.); (A.A.); (J.-A.S.); (I.A.)
| | - Christel Condroyer
- Institut de la Vision, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Sorbonne Université, F-75012 Paris, France; (E.O.); (M.N.); (M.d.S.D.); (T.P.); (C.M.); (C.C.); (A.A.); (J.-A.S.); (I.A.)
| | - Aline Antonio
- Institut de la Vision, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Sorbonne Université, F-75012 Paris, France; (E.O.); (M.N.); (M.d.S.D.); (T.P.); (C.M.); (C.C.); (A.A.); (J.-A.S.); (I.A.)
| | - José-Alain Sahel
- Institut de la Vision, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Sorbonne Université, F-75012 Paris, France; (E.O.); (M.N.); (M.d.S.D.); (T.P.); (C.M.); (C.C.); (A.A.); (J.-A.S.); (I.A.)
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC1423, F-75012 Paris, France
- Fondation Ophtalmologique Adolphe de Rothschild, F-75019 Paris, France
- Academie des Sciences, Institut de France, F-75006 Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Isabelle Audo
- Institut de la Vision, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Sorbonne Université, F-75012 Paris, France; (E.O.); (M.N.); (M.d.S.D.); (T.P.); (C.M.); (C.C.); (A.A.); (J.-A.S.); (I.A.)
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC1423, F-75012 Paris, France
- Institute of Ophthalmology, University College of London, London EC1V 9EL, UK
| | - Christina Zeitz
- Institut de la Vision, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Sorbonne Université, F-75012 Paris, France; (E.O.); (M.N.); (M.d.S.D.); (T.P.); (C.M.); (C.C.); (A.A.); (J.-A.S.); (I.A.)
- Correspondence: ; Tel.: +33-1-53-46-25-40
| |
Collapse
|
14
|
Loss of the ER membrane protein complex subunit Emc3 leads to retinal bipolar cell degeneration in aged mice. PLoS One 2020; 15:e0238435. [PMID: 32886670 PMCID: PMC7473584 DOI: 10.1371/journal.pone.0238435] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 08/17/2020] [Indexed: 02/05/2023] Open
Abstract
The endoplasmic reticulum (ER) membrane protein complex (EMC) is a conserved protein complex involved in inserting the transmembrane domain of membrane proteins into membranes in the ER. EMC3 is an essential component of EMC and is important for rhodopsin synthesis in photoreceptor cells. However, the in vivo function of Emc3 in bipolar cells (BCs) has not been determined. To explore the role of Emc3 in BCs, we generated a BC-specific Emc3 knockout mouse model (named Emc3 cKO) using the Purkinje cell protein 2 (Pcp2) Cre line. Although normal electroretinography (ERG) b-waves were observed in Emc3 cKO mice at 6 months of age, Emc3 cKO mice exhibited reduced b-wave amplitudes at 12 months of age, as determined by scotopic and photopic ERG, and progressive death of BCs, whereas the ERG a-wave amplitudes were preserved. PKCa staining of retinal cryosections from Emc3 cKO mice revealed death of rod BCs. Loss of Emc3 led to the presence of the synaptic protein mGLuR6 in the outer nuclear layer (ONL). Immunostaining analysis of presynaptic protein postsynaptic density protein 95 (PSD95) revealed rod terminals retracted to the ONL in Emc3 cKO mice at 12 months of age. In addition, deletion of Emc3 resulted in elevated glial fibrillary acidic protein, indicating reactive gliosis in the retina. Our data demonstrate that loss of Emc3 in BCs leads to decreased ERG response, increased astrogliosis and disruption of the retinal inner nuclear layer in mice of 12 months of age. Taken together, our studies indicate that Emc3 is not required for the development of BCs but is important for long-term survival of BCs.
Collapse
|
15
|
Agosto MA, Wensel TG. LRRTM4 is a member of the transsynaptic complex between rod photoreceptors and bipolar cells. J Comp Neurol 2020; 529:221-233. [PMID: 32390181 DOI: 10.1002/cne.24944] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/09/2020] [Accepted: 05/01/2020] [Indexed: 12/22/2022]
Abstract
Leucine rich repeat transmembrane (LRRTM) proteins are synaptic adhesion molecules with roles in synapse formation and signaling. LRRTM4 transcripts were previously shown to be enriched in rod bipolar cells (BCs), secondary neurons of the retina that form synapses with rod photoreceptors. Using two different antibodies, LRRTM4 was found to reside primarily at rod BC dendritic tips, where it colocalized with the transduction channel protein, TRPM1. LRRTM4 was not detected at dendritic tips of ON-cone BCs. Following somatic knockout of LRRTM4 in BCs by subretinal injection and electroporation of CRISPR/Cas9, LRRTM4 was abolished or reduced in the dendritic tips of transfected cells. Knockout cells had a normal complement of TRPM1 at their dendritic tips, while GPR179 accumulation was partially reduced. In experiments with heterologously expressed protein, the extracellular domain of LRRTM4 was found to engage in heparan-sulfate dependent binding with pikachurin. These results implicate LRRTM4 in the GPR179-pikachurin-dystroglycan transsynaptic complex at rod synapses.
Collapse
Affiliation(s)
- Melina A Agosto
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Theodore G Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
16
|
Homeostatic Plasticity Shapes the Retinal Response to Photoreceptor Degeneration. Curr Biol 2020; 30:1916-1926.e3. [PMID: 32243858 DOI: 10.1016/j.cub.2020.03.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/28/2020] [Accepted: 03/12/2020] [Indexed: 11/21/2022]
Abstract
Homeostatic plasticity stabilizes input and activity levels during neural development, but whether it can restore connectivity and preserve circuit function during neurodegeneration is unknown. Photoreceptor degeneration is the most common cause of blindness in the industrialized world. Visual deficits are dominated by cone loss, which progresses slowly, leaving a window during which rewiring of second-order neurons (i.e., bipolar cells) could preserve function. Here we establish a transgenic model to induce cone degeneration with precise control and analyze bipolar cell responses and their effects on vision through anatomical reconstructions, in vivo electrophysiology, and behavioral assays. In young retinas, we find that three bipolar cell types precisely restore input synapse numbers when 50% of cones degenerate but one does not. Of the three bipolar cell types that rewire, two contact new cones within stable dendritic territories, whereas one expands its dendrite arbors to reach new partners. In mature retinas, only one of four bipolar cell types rewires homeostatically. This steep decline in homeostatic plasticity is accompanied by reduced light responses of bipolar cells and deficits in visual behaviors. By contrast, light responses and behavioral performance are preserved when cones degenerate in young mice. Our results reveal unexpected cell type specificity and a steep maturational decline of homeostatic plasticity. The effect of homeostatic plasticity on functional outcomes identify it as a promising therapeutic target for retinal and other neurodegenerative diseases.
Collapse
|
17
|
Furukawa T, Ueno A, Omori Y. Molecular mechanisms underlying selective synapse formation of vertebrate retinal photoreceptor cells. Cell Mol Life Sci 2020; 77:1251-1266. [PMID: 31586239 PMCID: PMC11105113 DOI: 10.1007/s00018-019-03324-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/21/2019] [Accepted: 09/25/2019] [Indexed: 11/29/2022]
Abstract
In vertebrate central nervous systems (CNSs), highly diverse neurons are selectively connected via synapses, which are essential for building an intricate neural network. The vertebrate retina is part of the CNS and is comprised of a distinct laminar organization, which serves as a good model system to study developmental synapse formation mechanisms. In the retina outer plexiform layer, rods and cones, two types of photoreceptor cells, elaborate selective synaptic contacts with ON- and/or OFF-bipolar cell terminals as well as with horizontal cell terminals. In the mouse retina, three photoreceptor subtypes and at least 15 bipolar subtypes exist. Previous and recent studies have significantly progressed our understanding of how selective synapse formation, between specific subtypes of photoreceptor and bipolar cells, is designed at the molecular level. In the ON pathway, photoreceptor-derived secreted and transmembrane proteins directly interact in trans with the GRM6 (mGluR6) complex, which is localized to ON-bipolar cell dendritic terminals, leading to selective synapse formation. Here, we review our current understanding of the key factors and mechanisms underlying selective synapse formation of photoreceptor cells with bipolar and horizontal cells in the retina. In addition, we describe how defects/mutations of the molecules involved in photoreceptor synapse formation are associated with human retinal diseases and visual disorders.
Collapse
Affiliation(s)
- Takahisa Furukawa
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Akiko Ueno
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshihiro Omori
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
18
|
Orlandi C, Omori Y, Wang Y, Cao Y, Ueno A, Roux MJ, Condomitti G, de Wit J, Kanagawa M, Furukawa T, Martemyanov KA. Transsynaptic Binding of Orphan Receptor GPR179 to Dystroglycan-Pikachurin Complex Is Essential for the Synaptic Organization of Photoreceptors. Cell Rep 2020; 25:130-145.e5. [PMID: 30282023 PMCID: PMC6203450 DOI: 10.1016/j.celrep.2018.08.068] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 08/16/2018] [Accepted: 08/23/2018] [Indexed: 01/05/2023] Open
Abstract
Establishing synaptic contacts between neurons is paramount for nervous system function. This process involves transsynaptic interactions between a host of cell adhesion molecules that act in cooperation with the proteins of the extracellular matrix to specify uniquephysiological propertiesofindividual synaptic connections. However, understanding of the molecular mechanisms that generate functional diversity in an input-specific fashion is limited. In this study, we identify that major components of the extracellular matrix proteins present in the synaptic cleft—members oftheheparansulfateproteoglycan (HSPG) family—associate with the GPR158/179 group of orphan receptors. Using the mammalian retina as a model system, we demonstrate that the HSPG member Pikachurin, released by photoreceptors, recruits a key post-synaptic signaling complex of downstream ON-bipolar neurons in coordination with the presynaptic dystroglycan glycoprotein complex. We further demonstrate that this transsynaptic assembly plays an essential role in synaptic transmission of photoreceptor signals. Orlandi et al. identify transsynaptic assembly at photoreceptor synapses involving pre-synaptic dystrophindystroglycan complex and the postsynaptic orphan receptor GPR179 bridged by HSPG protein Pikachurin in the cleft and demonstrate its role in shaping transmission of photoreceptor signals.
Collapse
Affiliation(s)
- Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Yoshihiro Omori
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Yuchen Wang
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Yan Cao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Akiko Ueno
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Michel J Roux
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Centre National de la Recherche Scientifique, UMR7104, INSERM, U1258, Illkirch, France
| | - Giuseppe Condomitti
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Herestraat 49, 3000 Leuven, Belgium
| | - Joris de Wit
- VIB Center for Brain & Disease Research, Herestraat 49, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Herestraat 49, 3000 Leuven, Belgium
| | - Motoi Kanagawa
- Division of Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takahisa Furukawa
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
19
|
Hellmer CB, Clemons MR, Nawy S, Ichinose T. A group I metabotropic glutamate receptor controls synaptic gain between rods and rod bipolar cells in the mouse retina. Physiol Rep 2018; 6:e13885. [PMID: 30338673 PMCID: PMC6194217 DOI: 10.14814/phy2.13885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 09/10/2018] [Accepted: 09/16/2018] [Indexed: 11/24/2022] Open
Abstract
The canonical mGluR6-Trpm1 pathway that generates the sign-inverting signal between photoreceptors and ON bipolar cells has been well described. However, one type of ON bipolar cell, the rod bipolar cell (RBC), additionally is thought to express the group I mGluRs whose function is unknown. We examined the role of group I mGluRs in mouse RBCs and here provide evidence that it controls synaptic gain between rods and RBCs. In dark-adapted conditions, the mGluR1 antagonists LY367385 and (RS)-1-Aminoindan-1,5-dicarboxylic acid, but not the mGluR5 antagonist 2-Methyl-6-(phenylethynyl)pyridine hydrochloride reduced the light-evoked responses in RBCs indicating that mGluR1, but not mGluR5, serves to potentiate RBC responses. Perturbing the downstream phospholipase C (PLC)-protein kinase C (PKC) pathway by inhibiting PLC, tightly buffering intracellular Ca2+ , or preventing its release from intracellular stores reduced the synaptic potentiation by mGluR1. The effect of mGluR1 activation was dependent upon adaptation state, strongly increasing the synaptic gain in dark-, but not in light-adapted retinas, or in the presence of a moderate background light, consistent with the idea that mGluR1 activation requires light-dependent glutamate release from rods. Moreover, immunostaining revealed that protein kinase Cα (PKCα) is more strongly expressed in RBC dendrites in dark-adapted conditions, revealing an additional mechanism behind the loss of mGluR1 potentiation. In light-adapted conditions, exogenous activation of mGluR1 with the agonist 3,5-Dihydroxyphenylglycine increased the mGluR6 currents in some RBCs and decreased it in others, suggesting an additional action of mGluR1 that is unmasked in the light-adapted state. Elevating intracellular free Ca2+ , consistently resulted in a decrease in synaptic gain. Our results provide evidence that mGluR1 controls the synaptic gain in RBCs.
Collapse
Affiliation(s)
- Chase B. Hellmer
- Department of Ophthalmology, Visual and Anatomical SciencesWayne State University School of MedicineDetroitMichigan48201
| | - Melissa Rampino Clemons
- Dominic P Purpura Dept. of NeuroscienceAlbert Einstein College of Medicine BronxBronxNew York10461
| | - Scott Nawy
- Dominic P Purpura Dept. of NeuroscienceAlbert Einstein College of Medicine BronxBronxNew York10461
- Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaNebraska68198
| | - Tomomi Ichinose
- Department of Ophthalmology, Visual and Anatomical SciencesWayne State University School of MedicineDetroitMichigan48201
| |
Collapse
|
20
|
Qutob N, Masuho I, Alon M, Emmanuel R, Cohen I, Di Pizio A, Madore J, Elkahloun A, Ziv T, Levy R, Gartner JJ, Hill VK, Lin JC, Hevroni Y, Greenberg P, Brodezki A, Rosenberg SA, Kosloff M, Hayward NK, Admon A, Niv MY, Scolyer RA, Martemyanov KA, Samuels Y. RGS7 is recurrently mutated in melanoma and promotes migration and invasion of human cancer cells. Sci Rep 2018; 8:653. [PMID: 29330521 PMCID: PMC5766496 DOI: 10.1038/s41598-017-18851-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 12/19/2017] [Indexed: 11/14/2022] Open
Abstract
Analysis of 501 melanoma exomes revealed RGS7, which encodes a GTPase-accelerating protein (GAP), to be a tumor-suppressor gene. RGS7 was mutated in 11% of melanomas and was found to harbor three recurrent mutations (p.R44C, p.E383K and p.R416Q). Structural modeling of the most common recurrent mutation of the three (p.R44C) predicted that it destabilizes the protein due to the loss of an H-bond and salt bridge network between the mutated position and the serine and aspartic acid residues at positions 58 as 61, respectively. We experimentally confirmed this prediction showing that the p.R44C mutant protein is indeed destabilized. We further show RGS7 p.R44C has weaker catalytic activity for its substrate Gαo, thus providing a dual mechanism for its loss of function. Both of these effects are expected to contribute to loss of function of RGS7 resulting in increased anchorage-independent growth, migration and invasion of melanoma cells. By mutating position 56 in the R44C mutant from valine to cysteine, thereby enabling the formation of a disulfide bridge between the two mutated positions, we slightly increased the catalytic activity and reinstated protein stability, leading to the rescue of RGS7′s function as a tumor suppressor. Our findings identify RGS7 as a novel melanoma driver and point to the clinical relevance of using strategies to stabilize the protein and, thereby, restore its function.
Collapse
Affiliation(s)
- Nouar Qutob
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Ikuo Masuho
- Department of Neuroscience, The Scripps Research Institute, FL, 33458, USA
| | - Michal Alon
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Rafi Emmanuel
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Isadora Cohen
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Antonella Di Pizio
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University, Rehovot, Israel
| | - Jason Madore
- Melanoma Institute Australia, University of Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW, Australia
| | - Abdel Elkahloun
- National Human Genome Research Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Tamar Ziv
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ronen Levy
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Jared J Gartner
- National Cancer Institute, Surgery Branch, US National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Victoria K Hill
- National Human Genome Research Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Jimmy C Lin
- National Human Genome Research Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Yael Hevroni
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Polina Greenberg
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Alexandra Brodezki
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Steven A Rosenberg
- National Human Genome Research Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Mickey Kosloff
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Nicholas K Hayward
- Melanoma Institute Australia, University of Sydney, NSW, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Arie Admon
- Department of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Masha Y Niv
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University, Rehovot, Israel
| | - Richard A Scolyer
- Melanoma Institute Australia, University of Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW, Australia.,Disciplines of Surgery and Pathology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | | | - Yardena Samuels
- Molecular Cell Biology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
21
|
Homeostatic plasticity shapes the visual system's first synapse. Nat Commun 2017; 8:1220. [PMID: 29089553 PMCID: PMC5663853 DOI: 10.1038/s41467-017-01332-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 09/08/2017] [Indexed: 11/19/2022] Open
Abstract
Vision in dim light depends on synapses between rods and rod bipolar cells (RBCs). Here, we find that these synapses exist in multiple configurations, in which single release sites of rods are apposed by one to three postsynaptic densities (PSDs). Single RBCs often form multiple PSDs with one rod; and neighboring RBCs share ~13% of their inputs. Rod-RBC synapses develop while ~7% of RBCs undergo programmed cell death (PCD). Although PCD is common throughout the nervous system, its influences on circuit development and function are not well understood. We generate mice in which ~53 and ~93% of RBCs, respectively, are removed during development. In these mice, dendrites of the remaining RBCs expand in graded fashion independent of light-evoked input. As RBC dendrites expand, they form fewer multi-PSD contacts with rods. Electrophysiological recordings indicate that this homeostatic co-regulation of neurite and synapse development preserves retinal function in dim light. Retinal rod bipolar cells (RBCs) partially undergo programmed cell death triggering cell density-dependent plasticity. This study shows that increased removal of RBCs using genetic approaches causes dendrites of the remaining RBCs to expand and contact more rod photoreceptors while reducing connectivity with each.
Collapse
|
22
|
The TRPM1 Channel Is Required for Development of the Rod ON Bipolar Cell-AII Amacrine Cell Pathway in the Retinal Circuit. J Neurosci 2017; 37:9889-9900. [PMID: 28899920 DOI: 10.1523/jneurosci.0824-17.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 09/01/2017] [Accepted: 09/06/2017] [Indexed: 12/28/2022] Open
Abstract
Neurotransmission plays an essential role in neural circuit formation in the central nervous system (CNS). Although neurotransmission has been recently clarified as a key modulator of retinal circuit development, the roles of individual synaptic transmissions are not yet fully understood. In the current study, we investigated the role of neurotransmission from photoreceptor cells to ON bipolar cells in development using mutant mouse lines of both sexes in which this transmission is abrogated. We found that deletion of the ON bipolar cation channel TRPM1 results in the abnormal contraction of rod bipolar terminals and a decreased number of their synaptic connections with amacrine cells. In contrast, these histological alterations were not caused by a disruption of total glutamate transmission due to loss of the ON bipolar glutamate receptor mGluR6 or the photoreceptor glutamate transporter VGluT1. In addition, TRPM1 deficiency led to the reduction of total dendritic length, branch numbers, and cell body size in AII amacrine cells. Activated Goα, known to close the TRPM1 channel, interacted with TRPM1 and induced the contraction of rod bipolar terminals. Furthermore, overexpression of Channelrhodopsin-2 partially rescued rod bipolar cell development in the TRPM1-/- retina, whereas the rescue effect by a constitutively closed form of TRPM1 was lower than that by the native form. Our results suggest that TRPM1 channel opening is essential for rod bipolar pathway establishment in development.SIGNIFICANCE STATEMENT Neurotransmission has been recognized recently as a key modulator of retinal circuit development in the CNS. However, the roles of individual synaptic transmissions are not yet fully understood. In the current study, we focused on neurotransmission between rod photoreceptor cells and rod bipolar cells in the retina. We used genetically modified mouse models which abrogate each step of neurotransmission: presynaptic glutamate release, postsynaptic glutamate reception, or transduction channel function. We found that the TRPM1 transduction channel is required for the development of rod bipolar cells and their synaptic formation with subsequent neurons, independently of glutamate transmission. This study advances our understanding of neurotransmission-mediated retinal circuit refinement.
Collapse
|
23
|
Martemyanov KA, Sampath AP. The Transduction Cascade in Retinal ON-Bipolar Cells: Signal Processing and Disease. Annu Rev Vis Sci 2017; 3:25-51. [PMID: 28715957 DOI: 10.1146/annurev-vision-102016-061338] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Our robust visual experience is based on the reliable transfer of information from our photoreceptor cells, the rods and cones, to higher brain centers. At the very first synapse of the visual system, information is split into two separate pathways, ON and OFF, which encode increments and decrements in light intensity, respectively. The importance of this segregation is borne out in the fact that receptive fields in higher visual centers maintain a separation between ON and OFF regions. In the past decade, the molecular mechanisms underlying the generation of ON signals have been identified, which are unique in their use of a G-protein signaling cascade. In this review, we consider advances in our understanding of G-protein signaling in ON-bipolar cell (BC) dendrites and how insights about signaling have emerged from visual deficits, mostly night blindness. Studies of G-protein signaling in ON-BCs reveal an intricate mechanism that permits the regulation of visual sensitivity over a wide dynamic range.
Collapse
Affiliation(s)
| | - Alapakkam P Sampath
- Jules Stein Eye Institute, University of California, Los Angeles, California 90095;
| |
Collapse
|
24
|
Neuillé M, Cao Y, Caplette R, Guerrero-Given D, Thomas C, Kamasawa N, Sahel JA, Hamel CP, Audo I, Picaud S, Martemyanov KA, Zeitz C. LRIT3 Differentially Affects Connectivity and Synaptic Transmission of Cones to ON- and OFF-Bipolar Cells. Invest Ophthalmol Vis Sci 2017; 58:1768-1778. [PMID: 28334377 PMCID: PMC5374884 DOI: 10.1167/iovs.16-20745] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose Mutations in LRIT3 lead to complete congenital stationary night blindness (cCSNB). Using a cCSNB mouse model lacking Lrit3 (nob6), we recently have shown that LRIT3 has a role in the correct localization of TRPM1 (transient receptor potential melastatin 1) to the dendritic tips of ON-bipolar cells (BCs), contacting both rod and cone photoreceptors. Furthermore, postsynaptic clustering of other mGluR6 cascade components is selectively eliminated at the dendritic tips of cone ON-BCs. The purpose of this study was to further define the role of LRIT3 in structural and functional organization of cone synapses. Methods Exhaustive electroretinogram analysis was performed in a patient with LRIT3 mutations. Multielectrode array recordings were performed at the level of retinal ganglion cells in nob6 mice. Targeting of GluR1 and GluR5 at the dendritic tips of OFF-BCs in nob6 retinas was assessed by immunostaining and confocal microscopy. The ultrastructure of photoreceptor synapses was evaluated by electron microscopy in nob6 mice. Results The patient with LRIT3 mutations had a selective ON-BC dysfunction with relatively preserved OFF-BC responses. In nob6 mice, complete lack of ON-pathway function with robust, yet altered signaling processing in OFF-pathways was detected. Consistent with these observations, molecules essential for the OFF-BC signaling were normally targeted to the synapse. Finally, synaptic contacts made by ON-BC but not OFF-BC neurons with the cone pedicles were disorganized without ultrastructural alterations in cone terminals, horizontal cell processes, or synaptic ribbons. Conclusions These results suggest that LRIT3 is likely involved in coordination of the transsynaptic communication between cones and ON-BCs during synapse formation and function.
Collapse
Affiliation(s)
- Marion Neuillé
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Yan Cao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida United States
| | - Romain Caplette
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | | | - Connon Thomas
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida United States
| | - Naomi Kamasawa
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida United States
| | - José-Alain Sahel
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France 4CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France 5Institute of Ophthalmology, University College of London, London, United Kingdom 6Fondation Ophtalmologique Adolphe de Rothschild, Paris, France 8Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Christian P Hamel
- INSERM U583, Physiopathologie et thérapie des déficits sensoriels et moteurs, Institut des Neurosciences de Montpellier, Hôpital Saint-Eloi, Montpellier, France
| | - Isabelle Audo
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France 4CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DHOS CIC1423, Paris, France 5Institute of Ophthalmology, University College of London, London, United Kingdom
| | - Serge Picaud
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida United States
| | - Christina Zeitz
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| |
Collapse
|
25
|
Daniele LL, Emran F, Lobo GP, Gaivin RJ, Perkins BD. Mutation of wrb, a Component of the Guided Entry of Tail-Anchored Protein Pathway, Disrupts Photoreceptor Synapse Structure and Function. Invest Ophthalmol Vis Sci 2017; 57:2942-54. [PMID: 27273592 PMCID: PMC4898200 DOI: 10.1167/iovs.15-18996] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Tail-anchored (TA) proteins contain a single hydrophobic domain at the C-terminus and are posttranslationally inserted into the ER membrane via the GET (guided entry of tail-anchored proteins) pathway. The role of the GET pathway in photoreceptors is unexplored. The goal of this study was to characterize the zebrafish pinball wizard mutant, which disrupts Wrb, a core component of the GET pathway. METHODS Electroretinography, optokinetic response measurements (OKR), immunohistochemistry, and electron microscopy analyses were employed to assess ribbon synapse function, protein expression, and ultrastructure in 5-day-old zebrafish larvae. Expression of wrb was investigated with real-time qRT-PCR and in situ hybridization. RESULTS Mutation of wrb abolished the OKR and greatly diminished the ERG b-wave, but not the a-wave. Ribeye and SV2 were partially mislocalized in both photoreceptors and hair cells of wrb mutants. Fewer contacts were seen between photoreceptors and bipolar cells in wrb-/- mutants. Expression of wrb was observed throughout the nervous system and Wrb localized to the ER and synaptic region of photoreceptors. Morpholino knockdown of the cytosolic ATPase trc40, which targets TA proteins to the ER, also diminished the OKR. Overexpression of wrb fully restored contrast sensitivity in mutants, while overexpression of mutant wrbR73A, which cannot bind Trc40, did not. CONCLUSIONS Proteins Wrb and Trc40 are required for synaptic transmission between photoreceptors and bipolar cells, indicating that TA protein insertion by the TRC pathway is a critical step in ribbon synapse assembly and function.
Collapse
Affiliation(s)
- Lauren L Daniele
- Department of Ophthalmic Research Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Farida Emran
- Centre for Research in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Glenn P Lobo
- Department of Ophthalmic Research Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Robert J Gaivin
- Department of Ophthalmic Research Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Brian D Perkins
- Department of Ophthalmic Research Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| |
Collapse
|
26
|
Wang Y, Fehlhaber KE, Sarria I, Cao Y, Ingram NT, Guerrero-Given D, Throesch B, Baldwin K, Kamasawa N, Ohtsuka T, Sampath AP, Martemyanov KA. The Auxiliary Calcium Channel Subunit α2δ4 Is Required for Axonal Elaboration, Synaptic Transmission, and Wiring of Rod Photoreceptors. Neuron 2017; 93:1359-1374.e6. [PMID: 28262416 PMCID: PMC5364038 DOI: 10.1016/j.neuron.2017.02.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 12/31/2016] [Accepted: 02/08/2017] [Indexed: 11/24/2022]
Abstract
Neural circuit wiring relies on selective synapse formation whereby a presynaptic release apparatus is matched with its cognate postsynaptic machinery. At metabotropic synapses, the molecular mechanisms underlying this process are poorly understood. In the mammalian retina, rod photoreceptors form selective contacts with rod ON-bipolar cells by aligning the presynaptic voltage-gated Ca2+ channel directing glutamate release (CaV1.4) with postsynaptic mGluR6 receptors. We show this coordination requires an extracellular protein, α2δ4, which complexes with CaV1.4 and the rod synaptogenic mediator, ELFN1, for trans-synaptic alignment with mGluR6. Eliminating α2δ4 in mice abolishes rod synaptogenesis and synaptic transmission to rod ON-bipolar cells, and disrupts postsynaptic mGluR6 clustering. We further find that in rods, α2δ4 is crucial for organizing synaptic ribbons and setting CaV1.4 voltage sensitivity. In cones, α2δ4 is essential for CaV1.4 function, but is not required for ribbon organization, synaptogenesis, or synaptic transmission. These findings offer insights into retinal pathologies associated with α2δ4 dysfunction.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Katherine E Fehlhaber
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ignacio Sarria
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Yan Cao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Norianne T Ingram
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Debbie Guerrero-Given
- Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Ben Throesch
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92121, USA
| | - Kristin Baldwin
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92121, USA
| | - Naomi Kamasawa
- Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Toshihisa Ohtsuka
- Department of Biochemistry, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Alapakkam P Sampath
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
27
|
Intermolecular Interaction between Anchoring Subunits Specify Subcellular Targeting and Function of RGS Proteins in Retina ON-Bipolar Neurons. J Neurosci 2016; 36:2915-25. [PMID: 26961947 DOI: 10.1523/jneurosci.3833-15.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In vertebrate retina, light responses generated by the rod photoreceptors are transmitted to the second-order neurons, the ON-bipolar cells (ON-BC), and this communication is indispensible for vision in dim light. In ON-BCs, synaptic transmission is initiated by the metabotropic glutamate receptor, mGluR6, that signals via the G-protein Go to control opening of the effector ion channel, TRPM1. A key role in this process belongs to the GTPase Activating Protein (GAP) complex that catalyzes Go inactivation upon light-induced suppression of glutamate release in rod photoreceptors, thereby driving ON-BC depolarization to changes in synaptic input. The GAP complex has a striking molecular complexity. It contains two Regulator of G-protein Signaling (RGS) proteins RGS7 and RGS11 that directly act on Go and two adaptor subunits: RGS Anchor Protein (R9AP) and the orphan receptor, GPR179. Here we examined the organizational principles of the GAP complex in ON-BCs. Biochemical experiments revealed that RGS7 binds to a conserved site in GPR179 and that RGS11 in vivo forms a complex only with R9AP. R9AP and GPR179 are further integrated via direct protein-protein interactions involving their cytoplasmic domains. Elimination of GPR179 prevents postsynaptic accumulation of R9AP. Furthermore, concurrent knock-out of both R9AP and RGS7 does not reconfigure the GAP complex and completely abolishes synaptic transmission, resulting in a novel mouse model of night blindness. Based on these results, we propose a model of hierarchical assembly and function of the GAP complex that supports ON-BCs visual signaling.
Collapse
|
28
|
Dinet V, Ciccotosto GD, Delaunay K, Borras C, Ranchon-Cole I, Kostic C, Savoldelli M, El Sanharawi M, Jonet L, Pirou C, An N, Abitbol M, Arsenijevic Y, Behar-Cohen F, Cappai R, Mascarelli F. Amyloid Precursor-Like Protein 2 deletion-induced retinal synaptopathy related to congenital stationary night blindness: structural, functional and molecular characteristics. Mol Brain 2016; 9:64. [PMID: 27267879 PMCID: PMC4897877 DOI: 10.1186/s13041-016-0245-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/30/2016] [Indexed: 12/03/2022] Open
Abstract
Background Amyloid precursor protein knockout mice (APP-KO) have impaired differentiation of amacrine and horizontal cells. APP is part of a gene family and its paralogue amyloid precursor-like protein 2 (APLP2) has both shared as well as distinct expression patterns to APP, including in the retina. Given the impact of APP in the retina we investigated how APLP2 expression affected the retina using APLP2 knockout mice (APLP2-KO). Results Using histology, morphometric analysis with noninvasive imaging technique and electron microscopy, we showed that APLP2-KO retina displayed abnormal formation of the outer synaptic layer, accompanied with greatly impaired photoreceptor ribbon synapses in adults. Moreover, APLP2-KO displayed a significant decease in ON-bipolar, rod bipolar and type 2 OFF-cone bipolar cells (36, 21 and 63 %, respectively). Reduction of the number of bipolar cells was accompanied with disrupted dendrites, reduced expression of metabotropic glutamate receptor 6 at the dendritic tips and alteration of axon terminals in the OFF laminae of the inner plexiform layer. In contrast, the APP-KO photoreceptor ribbon synapses and bipolar cells were intact. The APLP2-KO retina displayed numerous phenotypic similarities with the congenital stationary night blindness, a non-progressive retinal degeneration disease characterized by the loss of night vision. The pathological phenotypes in the APLP2-KO mouse correlated to altered transcription of genes involved in pre- and postsynatic structure/function, including CACNA1F, GRM6, TRMP1 and Gα0, and a normal scotopic a-wave electroretinogram amplitude, markedly reduced scotopic electroretinogram b-wave and modestly reduced photopic cone response. This confirmed the impaired function of the photoreceptor ribbon synapses and retinal bipolar cells, as is also observed in congenital stationary night blindness. Since congenital stationary night blindness present at birth, we extended our analysis to retinal differentiation and showed impaired differentiation of different bipolar cell subtypes and an altered temporal sequence of development from OFF to ON laminae in the inner plexiform layer. This was associated with the altered expression patterns of bipolar cell generation and differentiation factors, including MATH3, CHX10, VSX1 and OTX2. Conclusions These findings demonstrate that APLP2 couples retina development and synaptic genes and present the first evidence that APLP2 expression may be linked to synaptic disease. Electronic supplementary material The online version of this article (doi:10.1186/s13041-016-0245-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Virginie Dinet
- Centre de Recherche des Cordeliers, Université Paris Descartes, Université Pierre et Marie Curie, Paris, France
| | - Giuseppe D Ciccotosto
- Department of Pathology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Australia
| | - Kimberley Delaunay
- Centre de Recherche des Cordeliers, Université Paris Descartes, Université Pierre et Marie Curie, Paris, France
| | - Céline Borras
- Centre de Recherche des Cordeliers, Université Paris Descartes, Université Pierre et Marie Curie, Paris, France
| | - Isabelle Ranchon-Cole
- Laboratoire de Biophysique Sensorielle, Université Clermont 1, Clermont-Ferrand, France
| | - Corinne Kostic
- Unit of Gene Therapy & Stem Cell Biology, University of Lausanne, Jules-Gonin Eye Hospital, Lausanne, Switzerland
| | - Michèle Savoldelli
- Centre de Recherche des Cordeliers, Université Paris Descartes, Université Pierre et Marie Curie, Paris, France
| | - Mohamed El Sanharawi
- Centre de Recherche des Cordeliers, Université Paris Descartes, Université Pierre et Marie Curie, Paris, France
| | - Laurent Jonet
- Centre de Recherche des Cordeliers, Université Paris Descartes, Université Pierre et Marie Curie, Paris, France
| | - Caroline Pirou
- Centre de Recherche des Cordeliers, Université Paris Descartes, Université Pierre et Marie Curie, Paris, France
| | - Na An
- Centre de Recherche des Cordeliers, Université Paris Descartes, Université Pierre et Marie Curie, Paris, France
| | - Marc Abitbol
- Centre de Recherche des Cordeliers, Université Paris Descartes, Université Pierre et Marie Curie, Paris, France
| | - Yvan Arsenijevic
- Unit of Gene Therapy & Stem Cell Biology, University of Lausanne, Jules-Gonin Eye Hospital, Lausanne, Switzerland
| | - Francine Behar-Cohen
- Centre de Recherche des Cordeliers, Université Paris Descartes, Université Pierre et Marie Curie, Paris, France
| | - Roberto Cappai
- Department of Pathology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Australia
| | - Frédéric Mascarelli
- Centre de Recherche des Cordeliers, Université Paris Descartes, Université Pierre et Marie Curie, Paris, France.
| |
Collapse
|
29
|
Glasauer SMK, Wäger R, Gesemann M, Neuhauss SCF. mglur6b:EGFP Transgenic zebrafish suggest novel functions of metabotropic glutamate signaling in retina and other brain regions. J Comp Neurol 2016; 524:2363-78. [PMID: 27121676 DOI: 10.1002/cne.24029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/21/2016] [Accepted: 04/21/2016] [Indexed: 02/04/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are mainly known for regulating excitability of neurons. However, mGluR6 at the photoreceptor-ON bipolar cell synapse mediates sign inversion through glutamatergic inhibition. Although this is currently the only confirmed function of mGluR6, other functions have been suggested. Here we present Tg(mglur6b:EGFP)zh1, a new transgenic zebrafish line recapitulating endogenous expression of one of the two mglur6 paralogs in zebrafish. Investigating transgene as well as endogenous mglur6b expression within the zebrafish retina indicates that EGFP and mglur6b mRNA are not only expressed in bipolar cells, but also in a subset of ganglion and amacrine cells. The amacrine cells labeled in Tg(mglur6b:EGFP)zh1 constitute a novel cholinergic, non-GABAergic, non-starburst amacrine cell type described for the first time in teleost fishes. Apart from the retina, we found transgene expression in subsets of periventricular neurons of the hypothalamus, Purkinje cells of the cerebellum, various cell types of the optic tectum, and mitral/ruffed cells of the olfactory bulb. These findings suggest novel functions of mGluR6 besides sign inversion at ON bipolar cell dendrites, opening up the possibility that inhibitory glutamatergic signaling may be more prevalent than currently thought. J. Comp. Neurol. 524:2363-2378, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Stella M K Glasauer
- University of Zurich, Institute of Molecular Life Sciences, Zurich, Switzerland.,Life Science Zurich Graduate School, Ph.D. Program in Molecular Life Sciences, Zurich, Switzerland
| | - Robert Wäger
- University of Zurich, Institute of Molecular Life Sciences, Zurich, Switzerland
| | - Matthias Gesemann
- University of Zurich, Institute of Molecular Life Sciences, Zurich, Switzerland
| | - Stephan C F Neuhauss
- University of Zurich, Institute of Molecular Life Sciences, Zurich, Switzerland.,Life Science Zurich Graduate School, Ph.D. Program in Molecular Life Sciences, Zurich, Switzerland
| |
Collapse
|
30
|
Tummala SR, Dhingra A, Fina ME, Li JJ, Ramakrishnan H, Vardi N. Lack of mGluR6-related cascade elements leads to retrograde trans-synaptic effects on rod photoreceptor synapses via matrix-associated proteins. Eur J Neurosci 2016; 43:1509-22. [PMID: 27037829 DOI: 10.1111/ejn.13243] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/29/2016] [Indexed: 12/23/2022]
Abstract
Heterotrimeric G-proteins couple metabotropic receptors to downstream effectors. In retinal ON bipolar cells, Go couples the metabotropic receptor mGluR6 to the TRPM1 channel and closes it in the dark, thus hyperpolarizing the cell. Light, via GTPase-activating proteins, deactivates Go , opens TRPM1 and depolarizes the cell. Go comprises Gαo1 , Gβ3 and Gγ13; all are necessary for efficient coupling. In addition, Gβ3 contributes to trafficking of certain cascade proteins and to maintaining the synaptic structure. The goal of this study was to determine the role of Gαo1 in maintaining the cascade and synaptic integrity. Using mice lacking Gαo1 , we quantified the immunostaining of certain mGluR6-related components. Deleting Gαo1 greatly reduced staining for Gβ3, Gγ13, Gβ5, RGS11, RGS7 and R9AP. Deletion of Gαo1 did not affect mGluR6, TRPM1 or PCP2. In addition, deleting Gαo1 reduced the number of rod bipolar dendrites that invaginate the rod terminal, similar to the effect seen in the absence of mGluR6, Gβ3 or the matrix-associated proteins, pikachurin, dystroglycan and dystrophin, which are localized presynaptically to the rod bipolar cell. We therefore tested mice lacking mGluR6, Gαo1 and Gβ3 for expression of these matrix-associated proteins. In all three genotypes, staining intensity for these proteins was lower than in wild type, suggesting a retrograde trans-synaptic effect. We propose that the mGluR6 macromolecular complex is connected to the presynaptic rod terminal via a protein chain that includes the matrix-associated proteins. When a component of the macromolecular chain is missing, the chain may fall apart and loosen the dendritic tip adherence within the invagination.
Collapse
Affiliation(s)
- Shanti R Tummala
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anuradha Dhingra
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Marie E Fina
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jian J Li
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Noga Vardi
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
31
|
The TRPM1 channel in ON-bipolar cells is gated by both the α and the βγ subunits of the G-protein Go. Sci Rep 2016; 6:20940. [PMID: 26883481 PMCID: PMC4756708 DOI: 10.1038/srep20940] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/16/2015] [Indexed: 12/25/2022] Open
Abstract
Transmission from photoreceptors to ON bipolar cells in mammalian retina is mediated by a sign-inverting cascade. Upon binding glutamate, the metabotropic glutamate receptor mGluR6 activates the heterotrimeric G-protein Gαoβ3γ13, and this leads to closure of the TRPM1 channel (melastatin). TRPM1 is thought to be constitutively open, but the mechanism that leads to its closure is unclear. We investigated this question in mouse rod bipolar cells by dialyzing reagents that modify the activity of either Gαo or Gβγ and then observing their effects on the basal holding current. After opening the TRPM1 channels with light, a constitutively active mutant of Gαo closed the channel, but wild-type Gαo did not. After closing the channels by dark adaptation, phosducin or inactive Gαo (both sequester Gβγ) opened the channel while the active mutant of Gαo did not. Co-immunoprecipitation showed that TRPM1 interacts with Gβ3 and with the active and inactive forms of Gαo. Furthermore, bioluminescent energy transfer assays indicated that while Gαo interacts with both the N- and the C- termini of TRPM1, Gβγ interacts only with the N-terminus. Our physiological and biochemical results suggest that both Gαo and Gβγ bind TRPM1 channels and cooperate to close them.
Collapse
|
32
|
Cao Y, Sarria I, Fehlhaber KE, Kamasawa N, Orlandi C, James KN, Hazen JL, Gardner MR, Farzan M, Lee A, Baker S, Baldwin K, Sampath AP, Martemyanov KA. Mechanism for Selective Synaptic Wiring of Rod Photoreceptors into the Retinal Circuitry and Its Role in Vision. Neuron 2015; 87:1248-1260. [PMID: 26402607 PMCID: PMC4583715 DOI: 10.1016/j.neuron.2015.09.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/11/2015] [Accepted: 08/31/2015] [Indexed: 01/06/2023]
Abstract
In the retina, rod and cone photoreceptors form distinct connections with different classes of downstream bipolar cells. However, the molecular mechanisms responsible for their selective connectivity are unknown. Here we identify a cell-adhesion protein, ELFN1, to be essential for the formation of synapses between rods and rod ON-bipolar cells in the primary rod pathway. ELFN1 is expressed selectively in rods where it is targeted to the axonal terminals by the synaptic release machinery. At the synapse, ELFN1 binds in trans to mGluR6, the postsynaptic receptor on rod ON-bipolar cells. Elimination of ELFN1 in mice prevents the formation of synaptic contacts involving rods, but not cones, allowing a dissection of the contributions of primary and secondary rod pathways to retinal circuit function and vision. We conclude that ELFN1 is necessary for the selective wiring of rods into the primary rod pathway and is required for high sensitivity of vision.
Collapse
Affiliation(s)
- Yan Cao
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Ignacio Sarria
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Katherine E Fehlhaber
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, 100 Stein Plaza, Los Angeles, CA 90095, USA
| | - Naomi Kamasawa
- Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Kiely N James
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92121, USA
| | - Jennifer L Hazen
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92121, USA
| | - Matthew R Gardner
- Department of Infectious Disease, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Michael Farzan
- Department of Infectious Disease, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, 51 Newton Road, Iowa City, IA 52242, USA
| | - Sheila Baker
- Department of Biochemistry, University of Iowa, 51 Newton Road, Iowa City, IA 52242, USA
| | - Kristin Baldwin
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92121, USA
| | - Alapakkam P Sampath
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, 100 Stein Plaza, Los Angeles, CA 90095, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA.
| |
Collapse
|
33
|
A Naturally Occurring Canine Model of Autosomal Recessive Congenital Stationary Night Blindness. PLoS One 2015; 10:e0137072. [PMID: 26368928 PMCID: PMC4569341 DOI: 10.1371/journal.pone.0137072] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/12/2015] [Indexed: 11/20/2022] Open
Abstract
Congenital stationary night blindness (CSNB) is a non-progressive, clinically and genetically heterogeneous disease of impaired night vision. We report a naturally-occurring, stationary, autosomal recessive phenotype in beagle dogs with normal daylight vision but absent night vision. Affected dogs had normal retinas on clinical examination, but showed no detectable rod responses. They had “negative-type” mixed rod and cone responses in full-field ERGs. Their photopic long-flash ERGs had normal OFF-responses associated with severely reduced ON-responses. The phenotype is similar to the Schubert-Bornschein form of complete CSNB in humans. Homozygosity mapping ruled out most known CSNB candidates as well as CACNA2D4 and GNB3. Three remaining genes were excluded based on sequencing the open reading frame and intron-exon boundaries (RHO, NYX), causal to a different form of CSNB (RHO) or X-chromosome (NYX, CACNA1F) location. Among the genes expressed in the photoreceptors and their synaptic terminals, and mGluR6 cascade and modulators, reduced expression of GNAT1, CACNA2D4 and NYX was observed by qRT-PCR in both carrier (n = 2) and affected (n = 2) retinas whereas CACNA1F was down-regulated only in the affecteds. Retinal morphology revealed normal cellular layers and structure, and electron microscopy showed normal rod spherules and synaptic ribbons. No difference from normal was observed by immunohistochemistry (IHC) for antibodies labeling rods, cones and their presynaptic terminals. None of the retinas showed any sign of stress. Selected proteins of mGluR6 cascade and its modulators were examined by IHC and showed that PKCα weakly labeled the rod bipolar somata in the affected, but intensely labeled axonal terminals that appeared thickened and irregular. Dendritic terminals of ON-bipolar cells showed increased Goα labeling. Both PKCα and Goα labeled the more prominent bipolar dendrites that extended into the OPL in affected but not normal retinas. Interestingly, RGS11 showed no labeling in the affected retina. Our results indicate involvement of a yet unknown gene in this canine model of complete CSNB.
Collapse
|
34
|
Neuillé M, Morgans CW, Cao Y, Orhan E, Michiels C, Sahel JA, Audo I, Duvoisin RM, Martemyanov KA, Zeitz C. LRIT3 is essential to localize TRPM1 to the dendritic tips of depolarizing bipolar cells and may play a role in cone synapse formation. Eur J Neurosci 2015; 42:1966-75. [PMID: 25997951 DOI: 10.1111/ejn.12959] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 04/30/2015] [Accepted: 05/17/2015] [Indexed: 02/06/2023]
Abstract
Mutations in LRIT3 lead to complete congenital stationary night blindness (cCSNB). The exact role of LRIT3 in ON-bipolar cell signaling cascade remains to be elucidated. Recently, we have characterized a novel mouse model lacking Lrit3 [no b-wave 6, (Lrit3(nob6/nob6) )], which displays similar abnormalities to patients with cCSNB with LRIT3 mutations. Here we compare the localization of components of the ON-bipolar cell signaling cascade in wild-type and Lrit3(nob6/nob6) retinal sections by immunofluorescence confocal microscopy. An anti-LRIT3 antibody was generated. Immunofluorescent staining of LRIT3 in wild-type mice revealed a specific punctate labeling in the outer plexiform layer (OPL), which was absent in Lrit3(nob6/nob6) mice. LRIT3 did not co-localize with ribeye or calbindin but co-localized with mGluR6. TRPM1 staining was severely decreased at the dendritic tips of all depolarizing bipolar cells in Lrit3(nob6/nob6) mice. mGluR6, GPR179, RGS7, RGS11 and Gβ5 immunofluorescence was absent at the dendritic tips of cone ON-bipolar cells in Lrit3(nob6/nob6) mice, while it was present at the dendritic tips of rod bipolar cells. Furthermore, peanut agglutinin (PNA) labeling was severely reduced in the OPL in Lrit3(nob6/nob6) mice. This study confirmed the localization of LRIT3 at the dendritic tips of depolarizing bipolar cells in mouse retina and demonstrated the dependence of TRPM1 localization on the presence of LRIT3. As tested components of the ON-bipolar cell signaling cascade and PNA revealed disrupted localization, an additional function of LRIT3 in cone synapse formation is suggested. These results point to a possibly different regulation of the mGluR6 signaling cascade between rod and cone ON-bipolar cells.
Collapse
Affiliation(s)
- Marion Neuillé
- INSERM, U968, Paris, F-75012, France.,CNRS, UMR_7210, Paris, F-75012, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France
| | - Catherine W Morgans
- Department of Physiology & Pharmacology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Yan Cao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Elise Orhan
- INSERM, U968, Paris, F-75012, France.,CNRS, UMR_7210, Paris, F-75012, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France
| | - Christelle Michiels
- INSERM, U968, Paris, F-75012, France.,CNRS, UMR_7210, Paris, F-75012, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France
| | - José-Alain Sahel
- INSERM, U968, Paris, F-75012, France.,CNRS, UMR_7210, Paris, F-75012, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.,Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, DHU ViewMaintain, INSERM-DHOS CIC, 1423, Paris, F-75012, France.,Institute of Ophthalmology, University College of London, London, EC1V 9EL, UK.,Fondation Ophtalmologique Adolphe de Rothschild, Paris, F-75019, France.,Académie des Sciences-Institut de France, Paris, F-75006, France
| | - Isabelle Audo
- INSERM, U968, Paris, F-75012, France.,CNRS, UMR_7210, Paris, F-75012, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.,Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, DHU ViewMaintain, INSERM-DHOS CIC, 1423, Paris, F-75012, France.,Institute of Ophthalmology, University College of London, London, EC1V 9EL, UK
| | - Robert M Duvoisin
- Department of Physiology & Pharmacology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Christina Zeitz
- INSERM, U968, Paris, F-75012, France.,CNRS, UMR_7210, Paris, F-75012, France.,Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France
| |
Collapse
|
35
|
Schneider FM, Mohr F, Behrendt M, Oberwinkler J. Properties and functions of TRPM1 channels in the dendritic tips of retinal ON-bipolar cells. Eur J Cell Biol 2015; 94:420-7. [PMID: 26111660 DOI: 10.1016/j.ejcb.2015.06.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
An increase in light intensity induces a depolarization in retinal ON-bipolar cells via a reduced glutamate release from presynaptic photoreceptor cells. The underlying transduction cascade in the dendritic tips of ON-bipolar cells involves mGluR6 glutamate receptors signaling to TRPM1 proteins that are an indispensable part of the transduction channel. Several other proteins are recognized to participate in the transduction machinery. Deficiency in many of these leads to congenital stationary night blindness, because rod bipolar cells, a subgroup of ON-bipolar cells, constitute the main route for sensory information under scotopic conditions. Here, we review the current knowledge about TRPM1 ion channels and how their activity is regulated within the postsynaptic compartment of ON-bipolar cells. The functional properties of TRPM1 channels in the dendritic compartment are not well understood as they differ substantially from those of recombinant TRPM1 channels. Critical evaluation of possible explanations of these discrepancies indicates that some key components of this transduction pathway might still not be known. The continued exploration of this pathway will yield further clinically useful insights.
Collapse
Affiliation(s)
- Franziska M Schneider
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Deutschhausstr. 1-2, D-35037 Marburg, Germany
| | - Florian Mohr
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Deutschhausstr. 1-2, D-35037 Marburg, Germany
| | - Marc Behrendt
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Deutschhausstr. 1-2, D-35037 Marburg, Germany
| | - Johannes Oberwinkler
- Institut für Physiologie und Pathophysiologie, Philipps-Universität Marburg, Deutschhausstr. 1-2, D-35037 Marburg, Germany.
| |
Collapse
|
36
|
Sarria I, Pahlberg J, Cao Y, Kolesnikov AV, Kefalov VJ, Sampath AP, Martemyanov KA. Sensitivity and kinetics of signal transmission at the first visual synapse differentially impact visually-guided behavior. eLife 2015; 4:e06358. [PMID: 25879270 PMCID: PMC4412108 DOI: 10.7554/elife.06358] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 04/11/2015] [Indexed: 12/29/2022] Open
Abstract
In the retina, synaptic transmission between photoreceptors and downstream ON-bipolar neurons (ON-BCs) is mediated by a GPCR pathway, which plays an essential role in vision. However, the mechanisms that control signal transmission at this synapse and its relevance to behavior remain poorly understood. In this study we used a genetic system to titrate the rate of GPCR signaling in ON-BC dendrites by varying the concentration of key RGS proteins and measuring the impact on transmission of signal between photoreceptors and ON-BC neurons using electroretinography and single cell recordings. We found that sensitivity, onset timing, and the maximal amplitude of light-evoked responses in rod- and cone-driven ON-BCs are determined by different RGS concentrations. We further show that changes in RGS concentration differentially impact visually guided-behavior mediated by rod and cone ON pathways. These findings illustrate that neuronal circuit properties can be modulated by adjusting parameters of GPCR-based neurotransmission at individual synapses. DOI:http://dx.doi.org/10.7554/eLife.06358.001 At the back of the eye, a structure called the retina contains several types of cell that convert light into the electrical signals that the brain interprets to produce vision. Cells called rods and cones detect the light, and then signal to other neurons in the retina that relay this information to the brain. Rods and cones are specialized to respond best to different visual features: cones detect color and can track rapid movement; whereas rods are more sensitive to low light levels and so enable night vision. All rods and cones communicate with particular types of neuron called an ‘ON bipolar cell’: rods send their information to rod-specific ON bipolar cells and cones to cone ON-bipolar cells. To maintain the differences in how visual features are detected, the signals sent by the rod or cone cells need to be tuned separately. Previous studies showed that bipolar cells rely on the action of proteins called RGSs to control how information is passed from rods and cones to ON bipolar cells. However, how the RGS proteins produce their effects is not well understood, and neither is their impact on vision or behavior. Sarria et al. used a genetic approach to create mice that progressively lost RGS proteins from their retina over the course of several weeks. Recording the nerve impulses produced by the bipolar cells as light shone on the retina revealed that RGS depletion affects these neurons in three ways: how sensitive they are to the signals sent by the rod and cone cells, how quickly they respond to a signal, and the size of the electrical response that they produce. Sarria et al. then investigated how these changes affected the behavior of the mice. To test the response of the rod cells, the mice performed tasks in dim light. This revealed that it was only when the sensitivity of the bipolar cells decreased that the mice performed worse. However, in a task involving fast-moving objects that investigated the response of cone cells, only changes to the speed of the response affected vision. Therefore, the RGS protein has different effects on the signals from rod cells and cone cells. These findings will be useful for understanding how different light sensitive cells in the retina communicate their signals to extract important visual features, allowing us to both see well at night and track rapid changes in scenery on a bright sunny day. DOI:http://dx.doi.org/10.7554/eLife.06358.002
Collapse
Affiliation(s)
- Ignacio Sarria
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Johan Pahlberg
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, United States
| | - Yan Cao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| | - Alexander V Kolesnikov
- Department of Ophthalmology and Visual Sciences, Washington University in St.Louis, St. Louis, United States
| | - Vladimir J Kefalov
- Department of Ophthalmology and Visual Sciences, Washington University in St.Louis, St. Louis, United States
| | - Alapakkam P Sampath
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, United States
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, United States
| |
Collapse
|
37
|
Orlandi C, Xie K, Masuho I, Fajardo-Serrano A, Lujan R, Martemyanov KA. Orphan Receptor GPR158 Is an Allosteric Modulator of RGS7 Catalytic Activity with an Essential Role in Dictating Its Expression and Localization in the Brain. J Biol Chem 2015; 290:13622-39. [PMID: 25792749 DOI: 10.1074/jbc.m115.645374] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Indexed: 11/06/2022] Open
Abstract
Regulators of G protein signaling control the duration and extent of signaling via G protein-coupled receptor (GPCR) pathways by accelerating the GTP hydrolysis on G protein α subunits thereby promoting termination of GPCR signaling. A member of this family, RGS7, plays a critical role in the nervous system where it regulates multiple neurotransmitter GPCRs that mediate vision, memory, and the action of addictive drugs. Previous studies have established that in vivo RGS7 forms mutually exclusive complexes with the membrane protein RGS7-binding protein or the orphan receptor GPR158. In this study, we examine the impact of GPR158 on RGS7 in the brain. We report that knock-out of GPR158 in mice results in marked post-transcriptional destabilization of RGS7 and substantial loss of its association with membranes in several brain regions. We further identified the RGS7-binding site in the C terminus of GPR158 and found that it shares significant homology with the RGS7-binding protein. The proximal portion of the GPR158 C terminus additionally contained a conserved sequence that was capable of enhancing RGS7 GTPase-activating protein activity in solution by an allosteric mechanism acting in conjunction with the regulators of the G protein signaling-binding domain. The distal portion of the GPR158 C terminus contained several phosphodiesterase E γ-like motifs and selectively recruited G proteins in their activated state. The results of this study establish GPR158 as an essential regulator of RGS7 in the native nervous system with a critical role in controlling its expression, membrane localization, and catalytic activity.
Collapse
Affiliation(s)
- Cesare Orlandi
- From the Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458 and
| | - Keqiang Xie
- From the Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458 and
| | - Ikuo Masuho
- From the Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458 and
| | - Ana Fajardo-Serrano
- the Instituto de Investigación en Descapacidades Neuronales (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, 02006 Albacete, Spain
| | - Rafael Lujan
- the Instituto de Investigación en Descapacidades Neuronales (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, 02006 Albacete, Spain
| | - Kirill A Martemyanov
- From the Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458 and
| |
Collapse
|
38
|
Woodard GE, Jardín I, Berna-Erro A, Salido GM, Rosado JA. Regulators of G-protein-signaling proteins: negative modulators of G-protein-coupled receptor signaling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 317:97-183. [PMID: 26008785 DOI: 10.1016/bs.ircmb.2015.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Regulators of G-protein-signaling (RGS) proteins are a category of intracellular proteins that have an inhibitory effect on the intracellular signaling produced by G-protein-coupled receptors (GPCRs). RGS along with RGS-like proteins switch on through direct contact G-alpha subunits providing a variety of intracellular functions through intracellular signaling. RGS proteins have a common RGS domain that binds to G alpha. RGS proteins accelerate GTPase and thus enhance guanosine triphosphate hydrolysis through the alpha subunit of heterotrimeric G proteins. As a result, they inactivate the G protein and quickly turn off GPCR signaling thus terminating the resulting downstream signals. Activity and subcellular localization of RGS proteins can be changed through covalent molecular changes to the enzyme, differential gene splicing, and processing of the protein. Other roles of RGS proteins have shown them to not be solely committed to being inhibitors but behave more as modulators and integrators of signaling. RGS proteins modulate the duration and kinetics of slow calcium oscillations and rapid phototransduction and ion signaling events. In other cases, RGS proteins integrate G proteins with signaling pathways linked to such diverse cellular responses as cell growth and differentiation, cell motility, and intracellular trafficking. Human and animal studies have revealed that RGS proteins play a vital role in physiology and can be ideal targets for diseases such as those related to addiction where receptor signaling seems continuously switched on.
Collapse
Affiliation(s)
- Geoffrey E Woodard
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Isaac Jardín
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - A Berna-Erro
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - Gines M Salido
- Department of Physiology, University of Extremadura, Caceres, Spain
| | - Juan A Rosado
- Department of Physiology, University of Extremadura, Caceres, Spain
| |
Collapse
|
39
|
Abstract
Across the nervous system, neurons form highly stereotypic patterns of synaptic connections that are designed to serve specific functions. Mature wiring patterns are often attained upon the refinement of early, less precise connectivity. Much work has led to the prevailing view that many developing circuits are sculpted by activity-dependent competition among converging afferents, which results in the elimination of unwanted synapses and the maintenance and strengthening of desired connections. Studies of the vertebrate retina, however, have recently revealed that activity can play a role in shaping developing circuits without engaging competition among converging inputs that differ in their activity levels. Such neurotransmission-mediated processes can produce stereotypic wiring patterns by promoting selective synapse formation rather than elimination. We discuss how the influence of transmission may also be limited by circuit design and further highlight the importance of transmission beyond development in maintaining wiring specificity and synaptic organization of neural circuits.
Collapse
|
40
|
Effects of mGluR6-deficiency on photoreceptor ribbon synapse formation: comparison of electron microscopic analysis of serial sections with random sections. Vis Neurosci 2015; 31:39-46. [PMID: 24801622 DOI: 10.1017/s0952523813000473] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
This study examined the effects of metabotropic glutamate receptor 6 (mGluR6) deficiency on ribbon synapse formation in rod spherules and cone pedicles using serial-section electron microscopy. In a wild-type (WT) mouse, only 3% of spherules had one invaginating bipolar dendrite (1B-type) and 97% of spherules were 2B-type. In contrast, in an mGluR6-knockout (KO) mouse, 29% of spherules were 1B-type and 71% of spherules were 2B-type. Spherules without bipolar invagination were not observed in either genotype. The single invaginating dendrites in 1B-type spherules were larger and the surface areas of synaptic ribbons were 23% smaller in the mGluR6-KO mouse than in the WT mouse. In cones, the number of invaginating bipolar dendrites decreased from 12 in the WT mouse to 9.5 in the mGluR6-KO mouse. This decrease correlated with a decrease in the number of cone synaptic ribbons from 10 in the WT mouse to 8 in the mGluR6-KO mouse. The mGluR6-KO phenotype showed negative effects on ribbon synapse formation. This negativity was similar to those in mGluR6-nob4, Gβ3-KO, Gβ5-KO, and RGS-7:RGS-11 double-KO mice, but the detailed manners and degrees of alterations appeared to vary depending on different missing components. Two published morphological assessments of the RGS-7:RGS-11 double-KO phenotype reported conflicting data; therefore, we tested the statistical techniques used in the two analyses. One statistical evaluation measure was effective in identifying a significant difference in structure between the mutant and WT phenotypes, whereas the other measure was ineffective. Conventional random section analysis using the effective measure provided sufficient data for a statistical test of the occurrence of structural changes. However, serial section analysis was required to determine the absolute numbers of ribbons and invaginating dendrites and to estimate structural parameters such as ribbon surface area.
Collapse
|
41
|
Ramakrishnan H, Dhingra A, Tummala SR, Fina ME, Li JJ, Lyubarsky A, Vardi N. Differential function of Gγ13 in rod bipolar and ON cone bipolar cells. J Physiol 2015; 593:1531-50. [PMID: 25416620 DOI: 10.1113/jphysiol.2014.281196] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 11/12/2014] [Indexed: 01/17/2023] Open
Abstract
Heterotrimeric G-proteins (comprising Gα and Gβγ subunits) are critical for coupling of metabotropic receptors to their downstream effectors. In the retina, glutamate released from photoreceptors in the dark activates metabotropic glutamate receptor 6 (mGluR6) receptors in ON bipolar cells; this leads to activation of Go , closure of transient receptor potential melastatin 1 channels and hyperpolarization of these cells. Go comprises Gαo , Gβ3 and a Gγ. The best Gγ candidate is Gγ13, although functional data to support this are lacking. Thus, we tested Gγ13 function by generating Gng13(-/-) knockout (KO) mice, recording electroretinograms (ERG) and performing immunocytochemical staining. The amplitude of scotopic ERG b-waves in KO mice was lower than in wild-type (WT) mice. Furthermore, in both KO and WT mice, the ERG b-wave decreased with age; this decrease was much more pronounced in KO mice. By contrast, the photopic ERG b-waves in KO mice were hardly affected at any age. In KO mice retinas, immunostaining for Gβ3 and for the GTPase activating proteins RGS7, RGS11, R9AP and Gβ5 decreased significantly in rod bipolar cells but not in ON cone bipolar cells. Staining for Gαo and certain other cascade elements decreased only slightly. Analysis of our ON bipolar cDNA library showed that these cells express mRNAs for Gγ5, Gγ10 and Gγ11. Quantitative RT-PCR of retinal cDNA showed greater values for these transcripts in retinas of KO mice, although the difference was not significant. Our results suggest that Gγ13 contributes to mGluR6 signalling in rod bipolar cells more than in ON cone bipolar cells, and that this contribution includes both coupling the receptor and maintaining a stable localization of the mGluR6-related cascade elements.
Collapse
Affiliation(s)
- Hariharasubramanian Ramakrishnan
- Department of Neuroscience, Department of Neurology and Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Martemyanov KA. G protein signaling in the retina and beyond: the Cogan lecture. Invest Ophthalmol Vis Sci 2014; 55:8201-7. [PMID: 25511392 PMCID: PMC4541486 DOI: 10.1167/iovs.14-15928] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Kirill A. Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States
| |
Collapse
|
43
|
Agosto MA, Zhang Z, He F, Anastassov IA, Wright SJ, McGehee J, Wensel TG. Oligomeric state of purified transient receptor potential melastatin-1 (TRPM1), a protein essential for dim light vision. J Biol Chem 2014; 289:27019-27033. [PMID: 25112866 PMCID: PMC4175340 DOI: 10.1074/jbc.m114.593780] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/03/2014] [Indexed: 11/06/2022] Open
Abstract
Transient receptor potential melastatin-1 (TRPM1) is essential for the light-induced depolarization of retinal ON bipolar cells. TRPM1 likely forms a multimeric channel complex, although almost nothing is known about the structure or subunit composition of channels formed by TRPM1 or any of its close relatives. Recombinant TRPM1 was robustly expressed in insect cells, but only a small fraction was localized to the plasma membrane. Similar intracellular localization was observed when TRPM1 was heterologously expressed in mammalian cells. TRPM1 was affinity-purified from Sf9 cells and complexed with amphipol, followed by detergent removal. In blue native gels and size exclusion chromatography, TRPM1 migrated with a mobility consistent with detergent- or amphipol-bound dimers. Cross-linking experiments were also consistent with a dimeric subunit stoichiometry, and cryoelectron microscopy and single particle analysis without symmetry imposition yielded a model with approximate 2-fold symmetrical features. Finally, electron microscopy of TRPM1-antibody complexes revealed a large particle that can accommodate TRPM1 and two antibody molecules. Taken together, these data indicate that purified TRPM1 is mostly dimeric. The three-dimensional structure of TRPM1 dimers is characterized by a small putative transmembrane domain and a larger domain with a hollow cavity. Blue native gels of solubilized mouse retina indicate that TRPM1 is present in two distinct complexes: one similar in size to the recombinant protein and one much larger. Because dimers are likely not functional ion channels, these results suggest that additional partner subunits participate in forming the transduction channel required for dim light vision and the ON pathway.
Collapse
Affiliation(s)
- Melina A Agosto
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Zhixian Zhang
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Feng He
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Ivan A Anastassov
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Sara J Wright
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Jennifer McGehee
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Theodore G Wensel
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030.
| |
Collapse
|
44
|
D'Orazi FD, Suzuki SC, Wong RO. Neuronal remodeling in retinal circuit assembly, disassembly, and reassembly. Trends Neurosci 2014; 37:594-603. [PMID: 25156327 DOI: 10.1016/j.tins.2014.07.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 07/03/2014] [Accepted: 07/27/2014] [Indexed: 10/24/2022]
Abstract
Developing neuronal circuits often undergo a period of refinement to eliminate aberrant synaptic connections. Inappropriate connections can also form among surviving neurons during neuronal degeneration. The laminar organization of the vertebrate retina enables synaptic reorganization to be readily identified. Synaptic rearrangements are shown to help sculpt developing retinal circuits, although the mechanisms involved remain debated. Structural changes in retinal diseases can also lead to functional rewiring. This poses a major challenge to retinal repair because it may be necessary to untangle the miswired connections before reconnecting with proper synaptic partners. Here, we review our current understanding of the mechanisms that underlie circuit remodeling during retinal development, and discuss how alterations in connectivity during damage could impede circuit repair.
Collapse
Affiliation(s)
- Florence D D'Orazi
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Sachihiro C Suzuki
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Rachel O Wong
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA.
| |
Collapse
|
45
|
|
46
|
Pearring JN, Lieu EC, Winter JR, Baker SA, Arshavsky VY. R9AP targeting to rod outer segments is independent of rhodopsin and is guided by the SNARE homology domain. Mol Biol Cell 2014; 25:2644-9. [PMID: 25009288 PMCID: PMC4148253 DOI: 10.1091/mbc.e14-02-0747] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
R9AP, the membrane anchor for transducin's GTPase-activating complex, contains targeting information within its SNARE homology domain that is both necessary and sufficient for R9AP delivery to photoreceptor outer segments. R9AP's targeting is independent of rhodopsin, the most abundant protein residing in the outer segment organelle. In vertebrate photoreceptor cells, rapid recovery from light excitation is dependent on the RGS9⋅Gβ5 GTPase-activating complex located in the light-sensitive outer segment organelle. RGS9⋅Gβ5 is tethered to the outer segment membranes by its membrane anchor, R9AP. Recent studies indicated that RGS9⋅Gβ5 possesses targeting information that excludes it from the outer segment and that this information is overridden by association with R9AP, which allows outer segment targeting of the entire complex. It was also proposed that R9AP itself does not contain specific targeting information and instead is delivered to the outer segment in the same post-Golgi vesicles as rhodopsin, because they are the most abundant transport vesicles in photoreceptor cells. In this study, we revisited this concept by analyzing R9AP targeting in rods of wild-type and rhodopsin-knockout mice. We found that the R9AP targeting mechanism does not require the presence of rhodopsin and further demonstrated that R9AP is actively targeted in rods by its SNARE homology domain.
Collapse
Affiliation(s)
- Jillian N Pearring
- Albert Eye Research Institute, Duke Eye Center, Duke University, Durham, NC 27710
| | - Eric C Lieu
- Albert Eye Research Institute, Duke Eye Center, Duke University, Durham, NC 27710
| | - Joan R Winter
- Albert Eye Research Institute, Duke Eye Center, Duke University, Durham, NC 27710
| | - Sheila A Baker
- Department of Biochemistry and Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Vadim Y Arshavsky
- Albert Eye Research Institute, Duke Eye Center, Duke University, Durham, NC 27710
| |
Collapse
|
47
|
GPR179 is required for high sensitivity of the mGluR6 signaling cascade in depolarizing bipolar cells. J Neurosci 2014; 34:6334-43. [PMID: 24790204 DOI: 10.1523/jneurosci.4044-13.2014] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Parallel visual pathways are initiated at the first retinal synapse by signaling between the rod and cone photoreceptors and two general classes of bipolar cells. For normal function, ON or depolarizing bipolar cells (DBCs) require the G-protein-coupled receptor, mGluR6, an intact G-protein-coupled cascade and the transient receptor potential melastatin 1 (TRPM1) cation channel. In addition, another seven transmembrane protein, GPR179, is required for DBC function and recruits the regulators of G-protein signaling (RGS) proteins, RGS7 and RGS11, to the dendritic tips of the DBCs. Here we use the Gpr179(nob5) mouse, which lacks GPR179 and has a no b-wave electroretinogram (ERG) phenotype, to demonstrate that despite the absence of both GPR179 and RGS7/RGS11, a small dark-adapted ERG b-wave remains and can be enhanced with long duration flashes. Consistent with the ERG, the mGluR6-mediated gating of TRPM1 can be evoked pharmacologically in Gpr179(nob5) and RGS7(-/-)/RGS11(-/-) rod BCs if strong stimulation conditions are used. In contrast, direct gating of TRPM1 by capsaicin in RGS7(-/-)/RGS11(-/-) and WT rod BCs is similar, but severely compromised in Gpr179(nob5) rod BCs. Noise and standing current analyses indicate that the remaining channels in Gpr179(nob5) and RGS7(-/-)/RGS11(-/-) rod BCs have a very low open probability. We propose that GPR179 along with RGS7 and RGS11 controls the ability of the mGluR6 cascade to gate TRPM1. In addition to its role in localizing RGS7 and RGS11 to the dendritic tips, GPR179 via a direct interaction with the TRPM1 channel alters its ability to be gated directly by capsaicin.
Collapse
|
48
|
Karpinsky-Semper D, Volmar CH, Brothers SP, Slepak VZ. Differential effects of the Gβ5-RGS7 complex on muscarinic M3 receptor-induced Ca2+ influx and release. Mol Pharmacol 2014; 85:758-68. [PMID: 24586057 PMCID: PMC4170115 DOI: 10.1124/mol.114.091843] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 02/28/2014] [Indexed: 11/22/2022] Open
Abstract
The G protein β subunit Gβ5 uniquely forms heterodimers with R7 family regulators of G protein signaling (RGS) proteins (RGS6, RGS7, RGS9, and RGS11) instead of Gγ. Although the Gβ5-RGS7 complex attenuates Ca(2+) signaling mediated by the muscarinic M3 receptor (M3R), the route of Ca(2+) entry (i.e., release from intracellular stores and/or influx across the plasma membrane) is unknown. Here, we show that, in addition to suppressing carbachol-stimulated Ca(2+) release, Gβ5-RGS7 enhanced Ca(2+) influx. This novel effect of Gβ5-RGS7 was blocked by nifedipine and 2-aminoethoxydiphenyl borate. Experiments with pertussis toxin, an RGS domain-deficient mutant of RGS7, and UBO-QIC {L-threonine,(3R)-N-acetyl-3-hydroxy-L-leucyl-(aR)-a-hydroxybenzenepropanoyl-2,3-idehydro-N-methylalanyl-L-alanyl-N-methyl-L-alanyl-(3R)-3-[[(2S,3R)-3-hydroxy-4- methyl-1-oxo-2-[(1-oxopropyl)amino]pentyl]oxy]-L-leucyl-N,O-dimethyl-,(7→1)-lactone (9CI)}, a novel inhibitor of Gq, showed that Gβ5-RGS7 modulated a Gq-mediated pathway. These studies indicate that Gβ5-RGS7, independent of RGS7 GTPase-accelerating protein activity, couples M3R to a nifedipine-sensitive Ca(2+) channel. We also compared the action of Gβ5-RGS7 on M3R-induced Ca(2+) influx and release elicited by different muscarinic agonists. Responses to Oxo-M [oxotremorine methiodide N,N,N,-trimethyl-4-(2-oxo-1-pyrrolidinyl)-2-butyn-1-ammonium iodide] were insensitive to Gβ5-RGS7. Pilocarpine responses consisted of a large release and modest influx components, of which the former was strongly inhibited whereas the latter was insensitive to Gβ5-RGS7. McN-A-343 [(4-hydroxy-2-butynyl)-1-trimethylammonium-3-chlorocarbanilate chloride] was the only compound whose total Ca(2+) response was enhanced by Gβ5-RGS7, attributed to, in part, by the relatively small Ca(2+) release this partial agonist stimulated. Together, these results show that distinct agonists not only have differential M3R functional selectivity, but also confer specific sensitivity to the Gβ5-RGS7 complex.
Collapse
Affiliation(s)
- Darla Karpinsky-Semper
- Department of Molecular and Cellular Pharmacology (D.K.-S., V.Z.S.) and Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences (C.-H.V., S.P.B.), University of Miami Miller School of Medicine, Miami, Florida
| | | | | | | |
Collapse
|
49
|
Tummala SR, Neinstein A, Fina ME, Dhingra A, Vardi N. Localization of Cacna1s to ON bipolar dendritic tips requires mGluR6-related cascade elements. Invest Ophthalmol Vis Sci 2014; 55:1483-92. [PMID: 24519419 DOI: 10.1167/iovs.13-13766] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE L-type voltage gated calcium channels in retina localize primarily at the presynaptic active zones of photoreceptors and bipolar cells where they modulate glutamate release. However, the pore forming subunit Cacna1s of certain L-type channels is also expressed postsynaptically at the tips of ON bipolar cell dendrites where it colocalizes with mGluR6, but has an unknown function. At these dendritic tips, the components of the mGluR6 signaling cascade cluster together in a macromolecular complex, and each one's localization often depends on that of the others. Thus, we explored if Cacna1s is part of the mGluR6 complex. METHODS We determined Cacna1s expression by PCR using an ON bipolar library, by Western blotting, and by standard immunohistochemistry. RESULTS The PCR amplification confirmed expression of the transcript in ON bipolar cells, and Western blotting showed the expected bands. Immunostaining for Cacna1s was stronger in the dendritic tips of rod bipolar cells than in those of ON cone bipolar cells. This staining severely decreased in mice missing various mGluR6 cascade elements (Grm6(-/-), Gnao1(-/-), Gnb3(-/-), Gng13(-/-), and Trpm1(-/-)). During development, the ratio of the number of Cacna1s puncta to the number of presynaptic ribbons followed a sigmoidal pattern, rising rapidly from P13 to P17. The mGluR6 expression preceded that of Cacna1s and RGS11. CONCLUSIONS Our results show that the localization and stability of Cacna1s depend on the expression of mGluR6 and its cascade components, and they suggest that Cacna1s is part of the mGluR6 complex. We hypothesize that Cacna1s contributes to light adaptation by permeating calcium.
Collapse
Affiliation(s)
- Shanti R Tummala
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | |
Collapse
|
50
|
Abstract
The b-wave is a major component of the electroretinogram that reflects the activity of depolarizing bipolar cells (DBCs). The b-wave is used diagnostically to identify patients with defects in DBC signaling or in transmission from photoreceptors to DBCs. In mouse models, an abnormal b-wave has been used to demonstrate a critical role of a particular protein in the release of glutamate from photoreceptor terminals, in establishing the structure of the photoreceptor-to-DBC synapse, in DBC signal transduction, and also in DBC development, survival, or metabolic support. The purpose of this review is to summarize these models and how they have advanced our understanding of outer retinal function.
Collapse
|