1
|
Zhao C, Cao Y, Ibrahim N, Wang Y, Martemyanov KA. Efficient in vivo labeling of endogenous proteins with SMART delineates retina cellular and synaptic organization. Nat Commun 2025; 16:3768. [PMID: 40263339 PMCID: PMC12015494 DOI: 10.1038/s41467-025-58945-6] [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: 12/27/2023] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
A key application of CRISPR/Cas9-based genomic editing is modification of genes to introduce engineered sequences. However, the editing flexibility is severely constrained by the requirement for targeting sites in proximity to the desired modification site, which makes many modifications intractable. Here, we develop a strategy that overcomes this key limitation to allow CRISPR-based editing at any position with high efficiency. It relies on reconstructing the targeted gene using Silently Mutate And Repair Template (SMART) where we mutate the gap sequence in the repair template to prevent its base pairing with the target DNA while maintaining the same amino acid coding. Using vertebrate retina as a neuronal model system we document the application of SMART editing for labeling endogenous proteins in vivo with high efficiency. We show that SMART editing allows us to access numerous cell types in the retina and address fundamental cell biological questions pertaining to its organization. We propose that this approach will facilitate functional genomic studies in a wide range of systems and increase the precision of corrective gene therapies.
Collapse
Affiliation(s)
- Chuanping Zhao
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
- Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL, USA
| | - Yan Cao
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Noor Ibrahim
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Yuchen Wang
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA.
- Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL, USA.
| |
Collapse
|
2
|
Pindwarawala M, Abid FA, Lee J, Miller ML, Noppers JS, Rideout AP, Agosto MA. Defective glycosylation and ELFN1 binding of mGluR6 congenital stationary night blindness mutants. Life Sci Alliance 2025; 8:e202403118. [PMID: 39681475 DOI: 10.26508/lsa.202403118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/18/2024] Open
Abstract
Synaptic transmission from photoreceptors to ON-bipolar cells (BCs) requires the postsynaptic metabotropic glutamate receptor mGluR6, located at BC dendritic tips. Binding of the neurotransmitter glutamate initiates G protein signaling that regulates the TRPM1 transduction channel. mGluR6 also interacts with presynaptic ELFN adhesion proteins, and these interactions are important for mGluR6 synaptic localization. The mechanisms of mGluR6 trafficking and synaptic targeting remain poorly understood. In this study, we investigated mGluR6 missense mutations from patients with congenital stationary night blindness (CSNB), which is associated with loss of synaptic transmission to ON-BCs. We found that multiple CSNB mutations in the extracellular ligand-binding domain of mGluR6 impart a trafficking defect leading to lack of complex N-glycosylation but efficient plasma membrane insertion, suggesting a Golgi bypass mechanism. These mutants fail to bind ELFN1, consistent with lack of a necessary modification normally acquired in the Golgi. The same mutants were mislocalized in bipolar cells, explaining the loss of function in CSNB. The results reveal a key role of Golgi trafficking in mGluR6 function, and suggest a role of the extracellular domain in Golgi sorting.
Collapse
Affiliation(s)
| | - Faiyaz Ak Abid
- Department of Microbiology and Immunology, Faculty of Science, Dalhousie University, Halifax, Canada
| | - Jaeeun Lee
- Medical Sciences Program, Faculty of Science, Dalhousie University, Halifax, Canada
| | - Michael L Miller
- Medical Sciences Program, Faculty of Science, Dalhousie University, Halifax, Canada
| | - Juliet S Noppers
- Medical Sciences Program, Faculty of Science, Dalhousie University, Halifax, Canada
| | - Andrew P Rideout
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Canada
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
| | - Melina A Agosto
- Retina and Optic Nerve Research Laboratory, Dalhousie University, Halifax, Canada
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Canada
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, Canada
| |
Collapse
|
3
|
Griffis KG, Fehlhaber KE, Rieke F, Sampath AP. Light Adaptation of Retinal Rod Bipolar Cells. J Neurosci 2023; 43:4379-4389. [PMID: 37208176 PMCID: PMC10278674 DOI: 10.1523/jneurosci.0444-23.2023] [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: 03/10/2023] [Revised: 04/07/2023] [Accepted: 05/02/2023] [Indexed: 05/21/2023] Open
Abstract
The sensitivity of retinal cells is altered in background light to optimize the detection of contrast. For scotopic (rod) vision, substantial adaptation occurs in the first two cells, the rods and rod bipolar cells (RBCs), through sensitivity adjustments in rods and postsynaptic modulation of the transduction cascade in RBCs. To study the mechanisms mediating these components of adaptation, we made whole-cell, voltage-clamp recordings from retinal slices of mice from both sexes. Adaptation was assessed by fitting the Hill equation to response-intensity relationships with the parameters of half-maximal response (I1/2 ), Hill coefficient (n), and maximum response amplitude (Rmax ). We show that rod sensitivity decreases in backgrounds according to the Weber-Fechner relation with an I1/2 of ∼50 R* s-1 The sensitivity of RBCs follows a near-identical function, indicating that changes in RBC sensitivity in backgrounds bright enough to adapt the rods are mostly derived from the rods themselves. Backgrounds too dim to adapt the rods can however alter n, relieving a synaptic nonlinearity likely through entry of Ca2+ into the RBCs. There is also a surprising decrease of Rmax , indicating that a step in RBC synaptic transduction is desensitized or that the transduction channels became reluctant to open. This effect is greatly reduced after dialysis of BAPTA at a membrane potential of +50 mV to impede Ca2+ entry. Thus the effects of background illumination in RBCs are in part the result of processes intrinsic to the photoreceptors and in part derive from additional Ca2+-dependent processes at the first synapse of vision.SIGNIFICANCE STATEMENT Light adaptation adjusts the sensitivity of vision as ambient illumination changes. Adaptation for scotopic (rod) vision is known to occur partly in the rods and partly in the rest of the retina from presynaptic and postsynaptic mechanisms. We recorded light responses of rods and rod bipolar cells to identify different components of adaptation and study their mechanisms. We show that bipolar-cell sensitivity largely follows adaptation of the rods but that light too dim to adapt the rods produces a linearization of the bipolar-cell response and a surprising decrease in maximum response amplitude, both mediated by a change in intracellular Ca2+ These findings provide a new understanding of how the retina responds to changing illumination.
Collapse
Affiliation(s)
- Khris G Griffis
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095
| | - Katherine E Fehlhaber
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195
| | - Alapakkam P Sampath
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| |
Collapse
|
4
|
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
|
5
|
Agosto MA, Adeosun AAR, Kumar N, Wensel TG. The mGluR6 ligand-binding domain, but not the C-terminal domain, is required for synaptic localization in retinal ON-bipolar cells. J Biol Chem 2021; 297:101418. [PMID: 34793838 PMCID: PMC8671642 DOI: 10.1016/j.jbc.2021.101418] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 11/26/2022] Open
Abstract
Signals from retinal photoreceptors are processed in two parallel channels-the ON channel responds to light increments, while the OFF channel responds to light decrements. The ON pathway is mediated by ON type bipolar cells (BCs), which receive glutamatergic synaptic input from photoreceptors via a G-protein-coupled receptor signaling cascade. The metabotropic glutamate receptor mGluR6 is located at the dendritic tips of all ON-BCs and is required for synaptic transmission. Thus, it is critically important for delivery of information from photoreceptors into the ON pathway. In addition to detecting glutamate, mGluR6 participates in interactions with other postsynaptic proteins, as well as trans-synaptic interactions with presynaptic ELFN proteins. Mechanisms of mGluR6 synaptic targeting and functional interaction with other synaptic proteins are unknown. Here, we show that multiple regions in the mGluR6 ligand-binding domain are necessary for both synaptic localization in BCs and ELFN1 binding in vitro. However, these regions were not required for plasma membrane localization in heterologous cells, indicating that secretory trafficking and synaptic localization are controlled by different mechanisms. In contrast, the mGluR6 C-terminus was dispensable for synaptic localization. In mGluR6 null mice, localization of the postsynaptic channel protein TRPM1 was compromised. Introducing WT mGluR6 rescued TRPM1 localization, while a C-terminal deletion mutant had significantly reduced rescue ability. We propose a model in which trans-synaptic ELFN1 binding is necessary for mGluR6 postsynaptic localization, whereas the C-terminus has a role in mediating TRPM1 trafficking. These findings reveal different sequence determinants of the multifunctional roles of mGluR6 in ON-BCs.
Collapse
Affiliation(s)
- Melina A Agosto
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA.
| | - Abiodun Adefola R Adeosun
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA; Pharmacology and Chemical Biology Graduate Program, Baylor College of Medicine, Houston, Texas, USA
| | - Nitin Kumar
- 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; Pharmacology and Chemical Biology Graduate Program, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
6
|
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
|
7
|
Burger CA, Jiang D, Mackin RD, Samuel MA. Development and maintenance of vision's first synapse. Dev Biol 2021; 476:218-239. [PMID: 33848537 DOI: 10.1016/j.ydbio.2021.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 12/21/2022]
Abstract
Synapses in the outer retina are the first information relay points in vision. Here, photoreceptors form synapses onto two types of interneurons, bipolar cells and horizontal cells. Because outer retina synapses are particularly large and highly ordered, they have been a useful system for the discovery of mechanisms underlying synapse specificity and maintenance. Understanding these processes is critical to efforts aimed at restoring visual function through repairing or replacing neurons and promoting their connectivity. We review outer retina neuron synapse architecture, neural migration modes, and the cellular and molecular pathways that play key roles in the development and maintenance of these connections. We further discuss how these mechanisms may impact connectivity in the retina.
Collapse
Affiliation(s)
- Courtney A Burger
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Danye Jiang
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Robert D Mackin
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Melanie A Samuel
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Murenu E, Pavlou M, Richter L, Rapti K, Just S, Cehajic-Kapetanovic J, Tafrishi N, Hayes A, Scholey R, Lucas R, Büning H, Grimm D, Michalakis S. A universal protocol for isolating retinal ON bipolar cells across species via fluorescence-activated cell sorting. Mol Ther Methods Clin Dev 2021; 20:587-600. [PMID: 33665228 PMCID: PMC7895692 DOI: 10.1016/j.omtm.2021.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/19/2021] [Indexed: 11/17/2022]
Abstract
Inherited retinal dystrophies (IRDs) are characterized by progressive degeneration and loss of light-sensing photoreceptors. The most promising therapeutic approach for IRDs is gene supplementation therapy using viral vectors, which requires the presence of viable photoreceptors at the time of intervention. At later disease stages, photoreceptors are lost and can no longer be rescued with this approach. For these patients, conferring light-sensing abilities to the remaining interneurons of the ON circuit (i.e., ON bipolar cells) using optogenetic tools poses an alternative treatment strategy. Such treatments, however, are hampered by the lack of efficient gene delivery tools targeting ON bipolar cells, which in turn rely on the effective isolation of these cells to facilitate tool development. Herein, we describe a method to selectively isolate ON bipolar cells via fluorescence-activated cell sorting (FACS), based on the expression of two intracellular markers. We show that the method is compatible with highly sensitive downstream analyses and suitable for the isolation of ON bipolar cells from healthy as well as degenerated mouse retinas. Moreover, we demonstrate that this approach works effectively using non-human primate (NHP) retinal tissue, thereby offering a reliable pipeline for universal screening strategies that do not require inter-species adaptations or transgenic animals.
Collapse
Affiliation(s)
- Elisa Murenu
- Department of Ophthalmology, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- Department of Pharmacy, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Marina Pavlou
- Department of Ophthalmology, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- Department of Pharmacy, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Lisa Richter
- Core Facility Flow Cytometry, Biomedical Center, Ludwig-Maximilians-University Munich, 82152 Planegg-Martinsried, Germany
| | - Kleopatra Rapti
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany
- BioQuant Center, University of Heidelberg, 69120 Heidelberg, Germany
| | - Sabrina Just
- Laboratory for Infection Biology and Gene Transfer, Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Jasmina Cehajic-Kapetanovic
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, Oxford University and Oxford University Hospitals, Oxford OX3 9DU, UK
| | - Neda Tafrishi
- Core Facility Flow Cytometry, Gene Center, BioSysM, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Andrew Hayes
- Center for Biological Timing & School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Rachel Scholey
- Center for Biological Timing & School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Robert Lucas
- Center for Biological Timing & School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Hildegard Büning
- Laboratory for Infection Biology and Gene Transfer, Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- REBIRTH Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
- German Center for Infection Research (DZIF), partner site, Hannover, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany
- BioQuant Center, University of Heidelberg, 69120 Heidelberg, Germany
- German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), partner site, Heidelberg, Germany
| | - Stylianos Michalakis
- Department of Ophthalmology, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- Department of Pharmacy, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| |
Collapse
|
9
|
Srivastava D, Yadav RP, Inamdar SM, Huang Z, Sokolov M, Boyd K, Artemyev NO. Transducin Partners Outside the Phototransduction Pathway. Front Cell Neurosci 2020; 14:589494. [PMID: 33173469 PMCID: PMC7591391 DOI: 10.3389/fncel.2020.589494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/10/2020] [Indexed: 11/13/2022] Open
Abstract
Transducin mediates signal transduction in a classical G protein-coupled receptor (GPCR) phototransduction cascade. Interactions of transducin with the receptor and the effector molecules had been extensively investigated and are currently defined at the atomic level. However, partners and functions of rod transducin α (Gαt 1) and βγ (Gβ1γ1) outside the visual pathway are not well-understood. In particular, light-induced redistribution of rod transducin from the outer segment to the inner segment and synaptic terminal (IS/ST) allows Gαt1 and/or Gβ1γ1 to modulate synaptic transmission from rods to rod bipolar cells (RBCs). Protein-protein interactions underlying this modulation are largely unknown. We discuss known interactors of transducin in the rod IS/ST compartment and potential pathways leading to the synaptic effects of light-dispersed Gαt1 and Gβ1γ1. Furthermore, we show that a prominent non-GPCR guanine nucleotide exchange factor (GEF) and a chaperone of Gα subunits, resistance to inhibitors of cholinesterase 8A (Ric-8A) protein, is expressed throughout the retina including photoreceptor cells. Recent structures of Ric-8A alone and in complexes with Gα subunits have illuminated the structural underpinnings of the Ric-8A activities. We generated a mouse model with conditional knockout of Ric-8A in rods in order to begin defining the functional roles of the protein in rod photoreceptors and the retina. Our analysis suggests that Ric-8A is not an obligate chaperone of Gαt1. Further research is needed to investigate probable roles of Ric-8A as a GEF, trafficking chaperone, or a mediator of the synaptic effects of Gαt1.
Collapse
Affiliation(s)
- Dhiraj Srivastava
- Department of Molecular Physiology and Biophysics, The University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Ravi P Yadav
- Department of Molecular Physiology and Biophysics, The University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Shivangi M Inamdar
- Department of Molecular Physiology and Biophysics, The University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Zhen Huang
- Department of Neurology and Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
| | - Maxim Sokolov
- Department of Ophthalmology, Biochemistry and Neuroscience, West Virginia University, Morgantown, WV, United States
| | - Kimberly Boyd
- Department of Molecular Physiology and Biophysics, The University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Nikolai O Artemyev
- Department of Molecular Physiology and Biophysics, The University of Iowa Carver College of Medicine, Iowa City, IA, United States.,Department of Ophthalmology and Visual Sciences, Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, United States
| |
Collapse
|
10
|
Palazzo E, Boccella S, Marabese I, Pierretti G, Guida F, Maione S. The Cold Case of Metabotropic Glutamate Receptor 6: Unjust Detention in the Retina? Curr Neuropharmacol 2020; 18:120-125. [PMID: 31573889 PMCID: PMC7324884 DOI: 10.2174/1570159x17666191001141849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/20/2019] [Accepted: 09/29/2019] [Indexed: 02/03/2023] Open
Abstract
It is a common opinion that metabotropic glutamate receptor subtype 6 (mGluR6) is expressed exclusively in the retina, and in particular in the dendrites of ON-bipolar cells. Glutamate released in darkness from photoreceptors activates mGluR6, which is negatively associated with a membrane non-selective cation channel, the transient receptor potential melanoma-related 1, TRPM1, resulting in cell hyperpolarization. The evidence that mGluR6 is expressed not only in the retina but also in other tissues and cell populations has accumulated over time. The expression of mGluR6 has been identified in microglia, bone marrow stromal and prostate cancer cells, B lymphocytes, melanocytes and keratinocytes and non-neural tissues such as testis, kidney, cornea, conjunctiva, and eyelid. The receptor also appears to be expressed in brain areas, such as the hypothalamus, cortex, hippocampus, nucleus of tractus solitarius, superior colliculus, axons of the corpus callosum and accessory olfactory bulb. The pharmacological activation of mGluR6 in the hippocampus produced an anxiolytic-like effect and in the periaqueductal gray analgesic potential. This review aims to collect all the evidence on the expression and functioning of mGluR6 outside the retina that has been accumulated over the years for a broader view of the potential of the receptor whose retinal confinement appears understimated.
Collapse
Affiliation(s)
- E Palazzo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Naples, Italy
| | - S Boccella
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Naples, Italy
| | - I Marabese
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Naples, Italy
| | - G Pierretti
- Department of Plastic Surgery, University of Campania "L. Vanvitelli", Naples, Italy
| | - F Guida
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Naples, Italy
| | - S Maione
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", Naples, Italy
| |
Collapse
|
11
|
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
|
12
|
Hack YL, Crabtree EE, Avila F, Sutton RB, Grahn R, Oh A, Gilger B, Bellone RR. Whole-genome sequencing identifies missense mutation in GRM6 as the likely cause of congenital stationary night blindness in a Tennessee Walking Horse. Equine Vet J 2020; 53:316-323. [PMID: 32654228 DOI: 10.1111/evj.13318] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 06/01/2020] [Accepted: 06/25/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND The only known genetic cause of congenital stationary night blindness (CSNB) in horses is a 1378 bp insertion in TRPM1. However, an affected Tennessee Walking Horse was found to have no copies of this variant. OBJECTIVES To identify the genetic cause for CSNB in an affected Tennessee Walking Horse. STUDY DESIGN Case report detailing a whole-genome sequencing (WGS) approach to identify a causal variant. METHODS A complete ophthalmic exam, including an electroretinogram (ERG), was performed on suspected CSNB-affected horse. WGS data were generated from the case and compared with data from seven other breeds (n = 29). One hundred candidate genes were evaluated for coding variants homozygous in the case and absent in all other horses. Protein modelling was used to assess the functional effects of the identified variant. A random cohort of 90 unrelated Tennessee Walking Horses and 273 horses from additional breeds were screened to estimate allele frequency of the GRM6 variant. RESULTS ERG results were consistent with CSNB. WGS analysis identified a missense mutation in metabotropic glutamate receptor 6 (GRM6) (c.533C>T p.Thr178Met). This single nucleotide polymorphism (SNP) is predicted to be deleterious and protein modelling supports impaired binding of the neurotransmitter glutamate. This variant was not detected in 273 horses from three additional breeds. The estimated allele frequency in Tennessee Walking Horses is 10%. MAIN LIMITATIONS Limited phenotype information for controls and no additional cases with which to replicate this finding. CONCLUSIONS We identified a likely causal recessive missense variant in GRM6. Based on protein modelling, this variant alters GRM6 binding, and thus signalling from the retinal rod cell to the ON-bipolar cell, impairing vision in low light conditions. Given the 10% population allele frequency, it is likely that additional affected horses exist in this breed and further work is needed to identify and examine these animals.
Collapse
Affiliation(s)
- Yael L Hack
- Veterinary Genetics Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Elizabeth E Crabtree
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Felipe Avila
- Veterinary Genetics Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Roger B Sutton
- Cell Physiology and Molecular Biophysics, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Robert Grahn
- Veterinary Genetics Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA
| | - Annie Oh
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Brian Gilger
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Rebecca R Bellone
- Veterinary Genetics Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA.,Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, California, USA
| |
Collapse
|
13
|
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
|
14
|
Chan K, Hoon M, Pattnaik BR, Ver Hoeve JN, Wahlgren B, Gloe S, Williams J, Wetherbee B, Kiland JA, Vogel KR, Jansen E, Salomons G, Walters D, Roullet JB, Gibson K M, McLellan GJ. Vigabatrin-Induced Retinal Functional Alterations and Second-Order Neuron Plasticity in C57BL/6J Mice. Invest Ophthalmol Vis Sci 2020; 61:17. [PMID: 32053727 PMCID: PMC7326505 DOI: 10.1167/iovs.61.2.17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose Vigabatrin (VGB) is an effective antiepileptic that increases concentrations of inhibitory γ-aminobutyric acid (GABA) by inhibiting GABA transaminase. Reports of VGB-associated visual field loss limit its clinical usefulness, and retinal toxicity studies in laboratory animals have yielded conflicting results. Methods We examined the functional and morphologic effects of VGB in C57BL/6J mice that received either VGB or saline IP from 10 to 18 weeks of age. Retinal structure and function were assessed in vivo by optical coherence tomography (OCT), ERG, and optomotor response. After euthanasia, retinas were processed for immunohistochemistry, and retinal GABA, and VGB quantified by mass spectrometry. Results No significant differences in visual acuity or total retinal thickness were identified between groups by optomotor response or optical coherence tomography, respectively. After 4 weeks of VGB treatment, ERG b-wave amplitude was enhanced, and amplitudes of oscillatory potentials were reduced. Dramatic rod and cone bipolar and horizontal cell remodeling, with extension of dendrites into the outer nuclear layer, was observed in retinas of VGB-treated mice. VGB treatment resulted in a mean 3.3-fold increase in retinal GABA concentration relative to controls and retinal VGB concentrations that were 20-fold greater than brain. Conclusions No evidence of significant retinal thinning or ERG a- or b-wave deficits were apparent, although we describe significant alterations in ERG b-wave and oscillatory potentials and in retinal cell morphology in VGB-treated C57BL/6J mice. The dramatic concentration of VGB in retina relative to the target tissue (brain), with a corresponding increase in retinal GABA, offers insight into the pathophysiology of VGB-associated visual field loss.
Collapse
Affiliation(s)
- Kore Chan
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, Madison, Wisconsin, United States
| | - Mrinalini Hoon
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, Madison, Wisconsin, United States
| | - Bikash R. Pattnaik
- McPherson Eye Research Institute, Madison, Wisconsin, United States
- Pediatrics Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - James N. Ver Hoeve
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, Madison, Wisconsin, United States
| | - Brad Wahlgren
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Shawna Gloe
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Jeremy Williams
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Brenna Wetherbee
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Julie A. Kiland
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
| | - Kara R. Vogel
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, Madison, Wisconsin, United States
| | - Erwin Jansen
- Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Gajja Salomons
- Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Dana Walters
- Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, Washington, United States
| | - Jean-Baptiste Roullet
- Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, Washington, United States
| | - K Michael Gibson
- Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, Washington, United States
| | - Gillian J. McLellan
- Department of Ophthalmology & Visual Science, University of Wisconsin–Madison, Madison, Wisconsin, United States
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States
- McPherson Eye Research Institute, Madison, Wisconsin, United States
| |
Collapse
|
15
|
Kinoshita J, Hasan N, Bell BA, Peachey NS. Reduced expression of the nob8 gene does not normalize the distribution or function of mGluR6 in the mouse retina. Mol Vis 2019; 25:890-901. [PMID: 32025181 PMCID: PMC6982428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 12/30/2019] [Indexed: 11/21/2022] Open
Abstract
Purpose The Grm6nob8 mouse carries a missense mutation in the Grm6 gene (p.Met66Leu), and exhibits a reduced b-wave of the electroretinogram (ERG), abnormal localization of metabotropic glutamate receptor 6 (mGluR6) to the depolarizing bipolar cell (DBC) soma, and a reduced level of mGluR6 at the DBC dendritic tips. Although the underlying mechanism remains unknown, one possible explanation is that DBCs cannot efficiently traffic the mutant mGluR6. In that scenario, reducing the total amount of mutant mGluR6 protein might normalize localization, and thus, improve the ERG phenotype as well. The second purpose of this study was to determine whether the abnormal cellular distribution of mutant mGluR6 in Grm6nob8 retinas might induce late onset DBC degeneration. Methods We crossed Grm6nob8 animals with Grm6nob3 mice, which carry a null mutation in Grm6, to generate Grm6nob3/nob8 compound heterozygotes. We used western blotting to measure the total mGluR6 content, and immunohistochemistry to document mGluR6 localization within DBCs. In addition, we examined outer retinal function with ERG and retinal architecture in vivo with spectral domain optical coherence tomography (SD-OCT). Results The retinal content of mGluR6 was reduced in the retinas of the Grm6nob3/nob8 compound heterozygotes compared to the Grm6nob8 homozygotes. The cellular distribution of mGluR6 in the Grm6nob3/nob8 compound heterozygotes matched that of the Grm6nob8 homozygotes, with extensive expression throughout the DBC cell body and limited expression at the DBC dendritic tips. The dark-adapted ERG b-waves of the Grm6nob3/nob8 mice were reduced in comparison to those of the Grm6nob8 homozygotes at postnatal day 21 and 28. The overall ERG waveforms obtained from 4- through 68-week old Grm6nob8 mice were in general agreement for dark- and light-adapted conditions. The maximum response and sensitivity of the dark-adapted ERG b-wave did not change statistically significantly with age. SD-OCT revealed the maintained laminar structure of the retina, including a clear inner nuclear layer (INL) at each age examined (from 11 to 57 weeks old), although the INL in the mice older than 39 weeks of age was somewhat thinner than that seen at 11 weeks. Conclusions Mislocalization of mutant mGluR6 is not normalized by reducing the total mGluR6. Mislocalized mutant mGluR6 does not trigger substantial loss of DBCs.
Collapse
Affiliation(s)
| | - Nazarul Hasan
- Department of Biochemistry & Molecular Genetics, University of Louisville, Louisville, KY
| | | | - Neal S. Peachey
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH
- Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| |
Collapse
|
16
|
Qin B, Humberg TH, Kim A, Kim HS, Short J, Diao F, White BH, Sprecher SG, Yuan Q. Muscarinic acetylcholine receptor signaling generates OFF selectivity in a simple visual circuit. Nat Commun 2019; 10:4093. [PMID: 31501438 PMCID: PMC6733798 DOI: 10.1038/s41467-019-12104-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 08/21/2019] [Indexed: 11/30/2022] Open
Abstract
ON and OFF selectivity in visual processing is encoded by parallel pathways that respond to either light increments or decrements. Despite lacking the anatomical features to support split channels, Drosophila larvae effectively perform visually-guided behaviors. To understand principles guiding visual computation in this simple circuit, we focus on investigating the physiological properties and behavioral relevance of larval visual interneurons. We find that the ON vs. OFF discrimination in the larval visual circuit emerges through light-elicited cholinergic signaling that depolarizes a cholinergic interneuron (cha-lOLP) and hyperpolarizes a glutamatergic interneuron (glu-lOLP). Genetic studies further indicate that muscarinic acetylcholine receptor (mAchR)/Gαo signaling produces the sign-inversion required for OFF detection in glu-lOLP, the disruption of which strongly impacts both physiological responses of downstream projection neurons and dark-induced pausing behavior. Together, our studies identify the molecular and circuit mechanisms underlying ON vs. OFF discrimination in the Drosophila larval visual system. Drosophila larvae are able to perform visually-guided behaviours yet the molecular and circuit mechanisms for discriminating changes in light intensity are not known. Here, the authors report that ON versus OFF discrimination results from opposing cholinergic and glutamatergic mechanisms.
Collapse
Affiliation(s)
- Bo Qin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Anna Kim
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hyong S Kim
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jacob Short
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Fengqiu Diao
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Benjamin H White
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Simon G Sprecher
- Department of Biology, University of Fribourg, 1700, Fribourg, Switzerland
| | - Quan Yuan
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
17
|
León-Navarro DA, Albasanz JL, Martín M. Functional Cross-Talk between Adenosine and Metabotropic Glutamate Receptors. Curr Neuropharmacol 2019; 17:422-437. [PMID: 29663888 PMCID: PMC6520591 DOI: 10.2174/1570159x16666180416093717] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/19/2018] [Accepted: 04/13/2018] [Indexed: 12/14/2022] Open
Abstract
Abstract: G-protein coupled receptors are transmembrane proteins widely expressed in cells and their transduction pathways are mediated by controlling second messenger levels through different G-protein interactions. Many of these receptors have been described as involved in the physiopathology of neurodegenerative diseases and even considered as potential targets for the design of novel therapeutic strategies. Endogenous and synthetic allosteric and orthosteric selective ligands are able to modulate GPCRs at both gene and protein expression levels and can also modify their physiological function. GPCRs that coexist in the same cells can homo- and heteromerize, therefore, modulating their function. Adenosine receptors are GPCRs which stimulate or inhibit adenylyl cyclase activity through Gi/Gs protein and are involved in the control of neurotransmitter release as glutamate. In turn, metabotropic glutamate receptors are also GPCRs which inhibit adenylyl cyclase or stimulate phospholipase C activities through Gi or Gq proteins, respectively. In recent years, evidence of crosstalk mechanisms be-tween different GPCRs have been described. The aim of the present review was to summarize the described mechanisms of interaction and crosstalking between adenosine and metabotropic glutamate receptors, mainly of group I, in both in vitro and in vivo systems, and their possible use for the design of novel ligands for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- David Agustín León-Navarro
- Departamento de Quimica Inorganica, Organica y Bioquimica. CRIB, Universidad de Castilla-La Mancha, Spain.,Facultad de Ciencias y Tecnologías Químicas, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain
| | - José Luis Albasanz
- Departamento de Quimica Inorganica, Organica y Bioquimica. CRIB, Universidad de Castilla-La Mancha, Spain.,Facultad de Ciencias y Tecnologías Químicas, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain.,Facultad de Medicina de Ciudad Real, Camino Moledores s/n. 13071 Ciudad Real, Spain
| | - Mairena Martín
- Departamento de Quimica Inorganica, Organica y Bioquimica. CRIB, Universidad de Castilla-La Mancha, Spain.,Facultad de Ciencias y Tecnologías Químicas, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain.,Facultad de Medicina de Ciudad Real, Camino Moledores s/n. 13071 Ciudad Real, Spain
| |
Collapse
|
18
|
Grassmeyer JJ, Cahill AL, Hays CL, Barta C, Quadros RM, Gurumurthy CB, Thoreson WB. Ca 2+ sensor synaptotagmin-1 mediates exocytosis in mammalian photoreceptors. eLife 2019; 8:e45946. [PMID: 31172949 PMCID: PMC6588344 DOI: 10.7554/elife.45946] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 06/06/2019] [Indexed: 11/24/2022] Open
Abstract
To encode light-dependent changes in membrane potential, rod and cone photoreceptors utilize synaptic ribbons to sustain continuous exocytosis while making rapid, fine adjustments to release rate. Release kinetics are shaped by vesicle delivery down ribbons and by properties of exocytotic Ca2+ sensors. We tested the role for synaptotagmin-1 (Syt1) in photoreceptor exocytosis by using novel mouse lines in which Syt1 was conditionally removed from rods or cones. Photoreceptors lacking Syt1 exhibited marked reductions in exocytosis as measured by electroretinography and single-cell recordings. Syt1 mediated all evoked release in cones, whereas rods appeared capable of some slow Syt1-independent release. Spontaneous release frequency was unchanged in cones but increased in rods lacking Syt1. Loss of Syt1 did not alter synaptic anatomy or reduce Ca2+ currents. These results suggest that Syt1 mediates both phasic and tonic release at photoreceptor synapses, revealing unexpected flexibility in the ability of Syt1 to regulate Ca2+-dependent synaptic transmission.
Collapse
Affiliation(s)
- Justin J Grassmeyer
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaUnited States
| | - Asia L Cahill
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
| | - Cassandra L Hays
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
- Department of Cellular and Integrative PhysiologyUniversity of Nebraska Medical CenterOmahaUnited States
| | - Cody Barta
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
| | - Rolen M Quadros
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research OfficeUniversity of Nebraska Medical CenterOmahaUnited States
| | - Channabasavaiah B Gurumurthy
- Mouse Genome Engineering Core Facility, Vice Chancellor for Research OfficeUniversity of Nebraska Medical CenterOmahaUnited States
- Developmental Neuroscience, Munroe Meyer Institute for Genetics and RehabilitationUniversity of Nebraska Medical CenterOmahaUnited States
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaUnited States
- Department of Pharmacology and Experimental NeuroscienceUniversity of Nebraska Medical CenterOmahaUnited States
| |
Collapse
|
19
|
Abstract
The transient receptor potential channel TRPM1 is required for synaptic transmission between photoreceptors and the ON subtype of bipolar cells (ON-BPC), mediating depolarization in response to light. TRPM1 is present in the somas and postsynaptic dendritic tips of ON-BPCs. Monoclonal antibodies generated against full-length TRPM1 were found to have differential labeling patterns when used to immunostain the mouse retina, with some yielding reduced labeling of dendritic tips relative to the labeling of cell bodies. Epitope mapping revealed that those antibodies that poorly label the dendritic tips share a binding site (N2d) in the N-terminal arm near the transmembrane domain. A major splice variant of TRPM1 lacking exon 19 does not contain the N2d binding site, but quantitative immunoblotting revealed no enrichment of this variant in synaptsomes. One explanation of the differential labeling is masking of the N2d epitope by formation of a synapse-specific multiprotein complex. Identifying the binding partners that are specific for the fraction of TRPM1 present at the synapses is an ongoing challenge for understanding TRPM1 function.
Collapse
|
20
|
Field GD, Sampath AP. Behavioural and physiological limits to vision in mammals. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0072. [PMID: 28193817 DOI: 10.1098/rstb.2016.0072] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2016] [Indexed: 01/22/2023] Open
Abstract
Human vision is exquisitely sensitive-a dark-adapted observer is capable of reliably detecting the absorption of a few quanta of light. Such sensitivity requires that the sensory receptors of the retina, rod photoreceptors, generate a reliable signal when single photons are absorbed. In addition, the retina must be able to extract this information and relay it to higher visual centres under conditions where very few rods signal single-photon responses while the majority generate only noise. Critical to signal transmission are mechanistic optimizations within rods and their dedicated retinal circuits that enhance the discriminability of single-photon responses by mitigating photoreceptor and synaptic noise. We describe behavioural experiments over the past century that have led to the appreciation of high sensitivity near absolute visual threshold. We further consider mechanisms within rod photoreceptors and dedicated rod circuits that act to extract single-photon responses from cellular noise. We highlight how these studies have shaped our understanding of brain function and point out several unresolved questions in the processing of light near the visual threshold.This article is part of the themed issue 'Vision in dim light'.
Collapse
Affiliation(s)
- Greg D Field
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Alapakkam P Sampath
- Stein Eye Institute, Department of Ophthalmology, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
21
|
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
|
22
|
Hendrickson A, Zhang C. Development of cone photoreceptors and their synapses in the human and monkey fovea. J Comp Neurol 2017; 527:38-51. [PMID: 28074469 DOI: 10.1002/cne.24170] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 11/05/2022]
Abstract
During retinal development, ribbon synapse assembly in the photoreceptors is a crucial step involving numerous molecules. While the developmental sequence of plexiform layers in human retina has been characterized, the molecular steps of synaptogenesis remain largely unknown. In the present study, we focused on the central rod-free region of primate retina, the fovea, to specifically investigate the development of cone photoreceptor ribbon synapses. Immunocytochemistry and electron microscopy were utilized to track the expression of photoreceptor transduction proteins and ribbon and synaptic markers in fetal human and Macaca retina. Although the inner plexiform layer appears earlier than the outer plexiform layer, synaptic proteins, and ribbons are first reliably recognized in cone pedicles. Markers first appear at fetal week 9. Both short (S) and medium/long (M/L) wavelength-selective cones express synaptic markers in the same temporal sequence; this is independent of opsin expression which takes place in S cones a month before M/L cones. The majority of ribbon markers, presynaptic vesicular release and postsynaptic neurotransduction-related machinery is present in both plexiform layers by fetal week 13. By contrast, two crucial components for cone to bipolar cell glutamatergic transmission, the metabotropic glutamate receptor 6 and voltage-dependent calcium channel α1.4, are not detected until fetal week 22 when bipolar cell invagination is present in the cone pedicle. These results suggest an intrinsically programmed but nonsynchronous expression of molecules in cone synaptic development. Moreover, functional ribbon synapses and active neurotransmission at foveal cone pedicles are possibly present as early as mid-gestation in human retina.
Collapse
Affiliation(s)
- Anita Hendrickson
- Department of Ophthalmology, University of Washington, Seattle, Washington.,Department of Biological Structure, University of Washington, Seattle, Washington
| | - Chi Zhang
- Department of Biological Structure, University of Washington, Seattle, Washington
| |
Collapse
|
23
|
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
|
24
|
Brüggen B, Kremser C, Bickert A, Ebel P, Vom Dorp K, Schultz K, Dörmann P, Willecke K, Dedek K. Defective ceramide synthases in mice cause reduced amplitudes in electroretinograms and altered sphingolipid composition in retina and cornea. Eur J Neurosci 2016; 44:1700-13. [PMID: 27086873 DOI: 10.1111/ejn.13260] [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] [Received: 01/12/2016] [Accepted: 04/08/2016] [Indexed: 01/24/2023]
Abstract
Complex sphingolipids are strongly expressed in neuronal tissue and contain ceramides in their backbone. Ceramides are synthesized by six ceramide synthases (CerS1-6). Although it is known that each tissue has a unique profile of ceramide synthase expression and ceramide synthases are implicated in several neurodegenerative disorders, the expression of ceramide synthase isoforms has not been investigated in the retina. Here we demonstrate CerS1, CerS2 and CerS4 expression in mouse retina and cornea, with CerS4 ubiquitously expressed in all retinal neurons and Müller cells. To test whether ceramide synthase deficiency affects retinal function, we compared electroretinograms and retina morphology between wild-type and CerS1-, CerS2- and CerS4-deficient mice. Electroretinograms were strongly reduced in amplitude in ceramide synthase-deficient mice, suggesting that signalling in the outer retina is affected. However, the number of photoreceptors and cone outer segment length were unaltered and no changes in retinal layer thickness or synaptic structures were found. Mass spectrometric analyses of ceramides, hexosyl-ceramides and sphingomyelins showed that C20 to C24 acyl-containing species were decreased whereas C16-containing species were increased in the retina of ceramide synthase-deficient mice. Similar but smaller changes were also found in the cornea. Thus, we hypothesize that the replacement of very long-chain fatty acyl residues by shorter C16 residues may affect the electrical properties of retina and cornea, and alter receptor-mediated signal transduction, vesicle-mediated synaptic transmission or corneal light transmission. Future studies need to identify the molecular targets of ceramides or derived sphingolipids in light signal transduction and transmission in the eye.
Collapse
Affiliation(s)
- Bianca Brüggen
- Neurobiology, University of Oldenburg, 26111, Oldenburg, Germany
| | | | - Andreas Bickert
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Philipp Ebel
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Katharina Vom Dorp
- Institute of Molecular Physiology and Biotechnology of Plants, University of Bonn, Bonn, Germany
| | - Konrad Schultz
- Neurobiology, University of Oldenburg, 26111, Oldenburg, Germany
| | - Peter Dörmann
- Institute of Molecular Physiology and Biotechnology of Plants, University of Bonn, Bonn, Germany
| | - Klaus Willecke
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Karin Dedek
- Neurobiology, University of Oldenburg, 26111, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Vincent A, Audo I, Tavares E, Maynes J, Tumber A, Wright T, Li S, Michiels C, Condroyer C, MacDonald H, Verdet R, Sahel JA, Hamel CP, Zeitz C, Héon E, Banin E, Bocquet B, De Baere E, Casteels I, Defoort-Dhellemmes S, Drumare I, Friedburg C, Gottlob I, Jacobson S, Kellner U, Koenekoop R, Kohl S, Leroy B, Lorenz B, McLean R, Meire F, Meunier I, Munier F, de Ravel T, Reiff C, Mohand-Saïd S, Sharon D, Schorderet D, Schwartz S, Zanlonghi X. Biallelic Mutations in GNB3 Cause a Unique Form of Autosomal-Recessive Congenital Stationary Night Blindness. Am J Hum Genet 2016; 98:1011-1019. [PMID: 27063057 DOI: 10.1016/j.ajhg.2016.03.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/18/2016] [Indexed: 01/13/2023] Open
Abstract
Congenital stationary night blindness (CSNB) is a heterogeneous group of non-progressive inherited retinal disorders with characteristic electroretinogram (ERG) abnormalities. Riggs and Schubert-Bornschein are subtypes of CSNB and demonstrate distinct ERG features. Riggs CSNB demonstrates selective rod photoreceptor dysfunction and occurs due to mutations in genes encoding proteins involved in rod phototransduction cascade; night blindness is the only symptom and eye examination is otherwise normal. Schubert-Bornschein CSNB is a consequence of impaired signal transmission between the photoreceptors and bipolar cells. Schubert-Bornschein CSNB is subdivided into complete CSNB with an ON bipolar signaling defect and incomplete CSNB with both ON and OFF pathway involvement. Both subtypes are associated with variable degrees of night blindness or photophobia, reduced visual acuity, high myopia, and nystagmus. Whole-exome sequencing of a family screened negative for mutations in genes associated with CSNB identified biallelic mutations in the guanine nucleotide-binding protein subunit beta-3 gene (GNB3). Two siblings were compound heterozygous for a deletion (c.170_172delAGA [p.Lys57del]) and a nonsense mutation (c.1017G>A [p.Trp339(∗)]). The maternal aunt was homozygous for the nonsense mutation (c.1017G>A [p.Trp339(∗)]). Mutational analysis of GNB3 in a cohort of 58 subjects with CSNB identified a sporadic case individual with a homozygous GNB3 mutation (c.200C>T [p.Ser67Phe]). GNB3 encodes the β subunit of G protein heterotrimer (Gαβγ) and is known to modulate ON bipolar cell signaling and cone transducin function in mice. Affected human subjects showed an unusual CSNB phenotype with variable degrees of ON bipolar dysfunction and reduced cone sensitivity. This unique retinal disorder with dual anomaly in visual processing expands our knowledge about retinal signaling.
Collapse
|
28
|
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
|
29
|
Full-field electroretinogram in autism spectrum disorder. Doc Ophthalmol 2016; 132:83-99. [DOI: 10.1007/s10633-016-9529-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 02/02/2016] [Indexed: 11/25/2022]
|
30
|
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
|
31
|
Scalabrino ML, Boye SL, Fransen KMH, Noel JM, Dyka FM, Min SH, Ruan Q, De Leeuw CN, Simpson EM, Gregg RG, McCall MA, Peachey NS, Boye SE. Intravitreal delivery of a novel AAV vector targets ON bipolar cells and restores visual function in a mouse model of complete congenital stationary night blindness. Hum Mol Genet 2015; 24:6229-39. [PMID: 26310623 DOI: 10.1093/hmg/ddv341] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/17/2015] [Indexed: 11/13/2022] Open
Abstract
Adeno-associated virus (AAV) effectively targets therapeutic genes to photoreceptors, pigment epithelia, Müller glia and ganglion cells of the retina. To date, no one has shown the ability to correct, with gene replacement, an inherent defect in bipolar cells (BCs), the excitatory interneurons of the retina. Targeting BCs with gene replacement has been difficult primarily due to the relative inaccessibility of BCs to standard AAV vectors. This approach would be useful for restoration of vision in patients with complete congenital stationary night blindness (CSNB1), where signaling through the ON BCs is eliminated due to mutations in their G-protein-coupled cascade genes. For example, the majority of CSNB1 patients carry a mutation in nyctalopin (NYX), which encodes a protein essential for proper localization of the TRPM1 cation channel required for ON BC light-evoked depolarization. As a group, CSNB1 patients have a normal electroretinogram (ERG) a-wave, indicative of photoreceptor function, but lack a b-wave due to defects in ON BC signaling. Despite retinal dysfunction, the retinas of CSNB1 patients do not degenerate. The Nyx(nob) mouse model of CSNB1 faithfully mimics this phenotype. Here, we show that intravitreally injected, rationally designed AAV2(quadY-F+T-V) containing a novel 'Ple155' promoter drives either GFP or YFP_Nyx in postnatal Nyx(nob) mice. In treated Nyx(nob) retina, robust and targeted Nyx transgene expression in ON BCs partially restored the ERG b-wave and, at the cellular level, signaling in ON BCs. Our results support the potential for gene delivery to BCs and gene replacement therapy in human CSNB1.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Charles N De Leeuw
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada V5Z 4H4, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | - Elizabeth M Simpson
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada V5Z 4H4, Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | - Ronald G Gregg
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY 40202, USA
| | - Maureen A McCall
- Department of Ophthalmology and Visual Sciences, Department of Anatomical Sciences and Neurobiology and
| | - Neal S Peachey
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA, Cole Eye Institute, Cleveland Clinic, Cleveland, OH 44195, USA and Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Shannon E Boye
- Department of Ophthalmology and, Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32610, USA,
| |
Collapse
|
32
|
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
|
33
|
Ou J, Vijayasarathy C, Ziccardi L, Chen S, Zeng Y, Marangoni D, Pope JG, Bush RA, Wu Z, Li W, Sieving PA. Synaptic pathology and therapeutic repair in adult retinoschisis mouse by AAV-RS1 transfer. J Clin Invest 2015; 125:2891-903. [PMID: 26098217 DOI: 10.1172/jci81380] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/30/2015] [Indexed: 01/24/2023] Open
Abstract
Strategies aimed at invoking synaptic plasticity have therapeutic potential for several neurological conditions. The human retinal synaptic disease X-linked retinoschisis (XLRS) is characterized by impaired visual signal transmission through the retina and progressive visual acuity loss, and mice lacking retinoschisin (RS1) recapitulate human disease. Here, we demonstrate that restoration of RS1 via retina-specific delivery of adeno-associated virus type 8-RS1 (AAV8-RS1) vector rescues molecular pathology at the photoreceptor-depolarizing bipolar cell (photoreceptor-DBC) synapse and restores function in adult Rs1-KO animals. Initial development of the photoreceptor-DBC synapse was normal in the Rs1-KO retina; however, the metabotropic glutamate receptor 6/transient receptor potential melastatin subfamily M member 1-signaling (mGluR6/TRPM1-signaling) cascade was not properly maintained. Specifically, the TRPM1 channel and G proteins Gαo, Gβ5, and RGS11 were progressively lost from postsynaptic DBC dendritic tips, whereas the mGluR6 receptor and RGS7 maintained proper synaptic position. This postsynaptic disruption differed from other murine night-blindness models with an electronegative electroretinogram response, which is also characteristic of murine and human XLRS disease. Upon AAV8-RS1 gene transfer to the retina of adult XLRS mice, TRPM1 and the signaling molecules returned to their proper dendritic tip location, and the DBC resting membrane potential was restored. These findings provide insight into the molecular plasticity of a critical synapse in the visual system and demonstrate potential therapeutic avenues for some diseases involving synaptic pathology.
Collapse
|
34
|
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
|
35
|
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
|
36
|
Pérez de Sevilla Müller L, Sargoy A, Fernández-Sánchez L, Rodriguez A, Liu J, Cuenca N, Brecha N. Expression and cellular localization of the voltage-gated calcium channel α2δ3 in the rodent retina. J Comp Neurol 2015; 523:1443-60. [PMID: 25631988 DOI: 10.1002/cne.23751] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 01/21/2015] [Accepted: 01/24/2015] [Indexed: 12/11/2022]
Abstract
High-voltage-activated calcium channels are hetero-oligomeric protein complexes that mediate multiple cellular processes, including the influx of extracellular Ca(2+), neurotransmitter release, gene transcription, and synaptic plasticity. These channels consist of a primary α(1) pore-forming subunit, which is associated with an extracellular α(2)δ subunit and an intracellular β auxiliary subunit, which alter the gating properties and trafficking of the calcium channel. The cellular localization of the α(2)δ(3) subunit in the mouse and rat retina is unknown. In this study using RT-PCR, a single band at ∼ 305 bp corresponding to the predicted size of the α(2)δ(3) subunit fragment was found in mouse and rat retina and brain homogenates. Western blotting of rodent retina and brain homogenates showed a single 123-kDa band. Immunohistochemistry with an affinity-purified antibody to the α(2)δ(3) subunit revealed immunoreactive cell bodies in the ganglion cell layer and inner nuclear layer and immunoreactive processes in the inner plexiform layer and the outer plexiform layer. α(2)δ(3) immunoreactivity was localized to multiple cell types, including ganglion, amacrine, and bipolar cells and photoreceptors, but not horizontal cells. The expression of the α(2)δ(3) calcium channel subunit to multiple cell types suggests that this subunit participates widely in Ca-channel-mediated signaling in the retina.
Collapse
Affiliation(s)
- Luis Pérez de Sevilla Müller
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, 90095
| | - Allison Sargoy
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, 90095.,Department of Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, 90095.,Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, 90095
| | | | - Allen Rodriguez
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, 90095
| | - Janelle Liu
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, 90095
| | - Nicolás Cuenca
- Physiology, Genetics and Microbiology, University of Alicante, 03690, Alicante, Spain
| | - Nicholas Brecha
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, 90095.,Department of Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, 90095.,Jules Stein Eye Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, 90095.,CURE-Digestive Diseases Research Center, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, 90095.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California, 90073
| |
Collapse
|
37
|
Congenital stationary night blindness: An analysis and update of genotype–phenotype correlations and pathogenic mechanisms. Prog Retin Eye Res 2015; 45:58-110. [DOI: 10.1016/j.preteyeres.2014.09.001] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/25/2014] [Accepted: 09/30/2014] [Indexed: 01/18/2023]
|
38
|
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
|
39
|
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
|
40
|
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
|
41
|
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
|
42
|
Hoon M, Okawa H, Della Santina L, Wong ROL. Functional architecture of the retina: development and disease. Prog Retin Eye Res 2014; 42:44-84. [PMID: 24984227 DOI: 10.1016/j.preteyeres.2014.06.003] [Citation(s) in RCA: 388] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/08/2014] [Accepted: 06/22/2014] [Indexed: 12/22/2022]
Abstract
Structure and function are highly correlated in the vertebrate retina, a sensory tissue that is organized into cell layers with microcircuits working in parallel and together to encode visual information. All vertebrate retinas share a fundamental plan, comprising five major neuronal cell classes with cell body distributions and connectivity arranged in stereotypic patterns. Conserved features in retinal design have enabled detailed analysis and comparisons of structure, connectivity and function across species. Each species, however, can adopt structural and/or functional retinal specializations, implementing variations to the basic design in order to satisfy unique requirements in visual function. Recent advances in molecular tools, imaging and electrophysiological approaches have greatly facilitated identification of the cellular and molecular mechanisms that establish the fundamental organization of the retina and the specializations of its microcircuits during development. Here, we review advances in our understanding of how these mechanisms act to shape structure and function at the single cell level, to coordinate the assembly of cell populations, and to define their specific circuitry. We also highlight how structure is rearranged and function is disrupted in disease, and discuss current approaches to re-establish the intricate functional architecture of the retina.
Collapse
Affiliation(s)
- Mrinalini Hoon
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Haruhisa Okawa
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Luca Della Santina
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - Rachel O L Wong
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA.
| |
Collapse
|
43
|
Abstract
The retina is one of the major energy consuming tissues within the body. In this context, synaptic transmission between light-excited rod and cone photoreceptors and downstream ON-bipolar neurons is a highly demanding energy consuming process. Sirtuin 6 (SIRT6), a NAD-dependent deacylase, plays a key role in regulating glucose metabolism. In this study, we demonstrate that SIRT6 is highly expressed in the retina, controlling levels of histone H3K9 and H3K56 acetylation. Notably, despite apparent normal histology, SIRT6 deficiency caused major retinal transmission defects concomitant to changes in expression of glycolytic genes and glutamate receptors, as well as elevated levels of apoptosis in inner retina cells. Our results identify SIRT6 as a critical modulator of retinal function, likely through its effects on chromatin.
Collapse
|
44
|
Retinal neurodegenerative changes in the adult insulin receptor substrate-2 deficient mouse. Exp Eye Res 2014; 124:1-10. [PMID: 24792588 DOI: 10.1016/j.exer.2014.04.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 04/17/2014] [Accepted: 04/22/2014] [Indexed: 11/23/2022]
Abstract
Insulin receptor substrate-2 (Irs2) mediates peripheral insulin action and is essential for retinal health. Previous investigations have reported severe photoreceptor degeneration and abnormal visual function in Irs2-deficient mice. However, molecular changes in the Irs2(-)(/)(-) mouse retina have not been described. In this study, we examined retinal degenerative changes in neuronal and glial cells of adult (9- and 12-week old) Irs2(-)(/)(-) mice by immunohistochemistry. 9-week old Irs2(-)(/)(-) mice showed significant thinning of outer retinal layers, concomitant to Müller and microglial cell activation. Photoreceptor cells displayed different signs of degeneration, such as outer/inner segment atrophy, redistribution of rod- and cone-opsins and spatial disorganization of cone cells. This was accompanied by synaptic changes at the outer plexiform layer, including the retraction of rod-spherules, reduction of photoreceptor synaptic ribbons and synaptic remodeling in second order neurons (i.e. loss and sprouting of dendritic processes in rod bipolar and horizontal cells). By 12 weeks of age, the thickness of inner retinal layers was severely affected. Although inner plexiform layer stratification remained unchanged at this stage, rod bipolar cell axon terminals were significantly depleted. Significant loss of Brn3a(+) retinal ganglion cells occurred in 12-week old Irs2(-)(/)(-) mice, in contrast to younger ages. Adult Irs2(-)(/)(-) mice showed clear hallmarks of neurodegeneration and disruption of the inner retina with increasing age. Pharmacological stimulation of Irs2 signaling pathway may provide additional neuroprotection in certain degenerative retinopathies.
Collapse
|
45
|
Neuillé M, El Shamieh S, Orhan E, Michiels C, Antonio A, Lancelot ME, Condroyer C, Bujakowska K, Poch O, Sahel JA, Audo I, Zeitz C. Lrit3 deficient mouse (nob6): a novel model of complete congenital stationary night blindness (cCSNB). PLoS One 2014; 9:e90342. [PMID: 24598786 PMCID: PMC3943948 DOI: 10.1371/journal.pone.0090342] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 01/31/2014] [Indexed: 01/10/2023] Open
Abstract
Mutations in LRIT3, coding for a Leucine-Rich Repeat, immunoglobulin-like and transmembrane domains 3 protein lead to autosomal recessive complete congenital stationary night blindness (cCSNB). The role of the corresponding protein in the ON-bipolar cell signaling cascade remains to be elucidated. Here we genetically and functionally characterize a commercially available Lrit3 knock-out mouse, a model to study the function and the pathogenic mechanism of LRIT3. We confirm that the insertion of a Bgeo/Puro cassette in the knock-out allele introduces a premature stop codon, which presumably codes for a non-functional protein. The mouse line does not harbor other mutations present in common laboratory mouse strains or in other known cCSNB genes. Lrit3 mutant mice exhibit a so-called no b-wave (nob) phenotype with lacking or severely reduced b-wave amplitudes in the scotopic and photopic electroretinogram (ERG), respectively. Optomotor tests reveal strongly decreased optomotor responses in scotopic conditions. No obvious fundus auto-fluorescence or histological retinal structure abnormalities are observed. However, spectral domain optical coherence tomography (SD-OCT) reveals thinned inner nuclear layer and part of the retina containing inner plexiform layer, ganglion cell layer and nerve fiber layer in these mice. To our knowledge, this is the first time that SD-OCT technology is used to characterize an animal model for CSNB. This phenotype is noted at 6 weeks and at 6 months. The stationary nob phenotype of mice lacking Lrit3, which we named nob6, confirms the findings previously reported in patients carrying LRIT3 mutations and is similar to other cCSNB mouse models. This novel mouse model will be useful for investigating the pathogenic mechanism(s) associated with LRIT3 mutations and clarifying the role of LRIT3 in the ON-bipolar cell signaling cascade.
Collapse
Affiliation(s)
- Marion Neuillé
- INSERM, U968, Paris, France
- CNRS, UMR_7210, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
| | - Said El Shamieh
- INSERM, U968, Paris, France
- CNRS, UMR_7210, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
| | - Elise Orhan
- INSERM, U968, Paris, France
- CNRS, UMR_7210, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
| | - Christelle Michiels
- INSERM, U968, Paris, France
- CNRS, UMR_7210, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
| | - Aline Antonio
- INSERM, U968, Paris, France
- CNRS, UMR_7210, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France
| | - Marie-Elise Lancelot
- INSERM, U968, Paris, France
- CNRS, UMR_7210, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
| | - Christel Condroyer
- INSERM, U968, Paris, France
- CNRS, UMR_7210, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
| | - Kinga Bujakowska
- INSERM, U968, Paris, France
- CNRS, UMR_7210, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
- Massachusetts Eye and Ear Infirmary, Ocular Genomics Institute, Boston, Massachusetts, United States of America
| | - Olivier Poch
- Laboratoire de Bioinformatique Intégrative et Génomique, ICube, CNRS, UMR_7357, Strasbourg, France
| | - José-Alain Sahel
- INSERM, U968, Paris, France
- CNRS, UMR_7210, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France
- Institute of Ophthalmology, University College of London, London, United Kingdom
- Fondation Ophtalmologique Adolphe de Rothschild, Paris, France
- Académie des Sciences–Institut de France, Paris, France
| | - Isabelle Audo
- INSERM, U968, Paris, France
- CNRS, UMR_7210, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France
- Institute of Ophthalmology, University College of London, London, United Kingdom
| | - Christina Zeitz
- INSERM, U968, Paris, France
- CNRS, UMR_7210, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, Paris, France
- * E-mail:
| |
Collapse
|
46
|
De Sevilla Müller LP, Liu J, Solomon A, Rodriguez A, Brecha NC. Expression of voltage-gated calcium channel α(2)δ(4) subunits in the mouse and rat retina. J Comp Neurol 2014; 521:2486-501. [PMID: 23296739 DOI: 10.1002/cne.23294] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 12/12/2012] [Accepted: 12/27/2012] [Indexed: 01/18/2023]
Abstract
High-voltage activated Ca channels participate in multiple cellular functions, including transmitter release, excitation, and gene transcription. Ca channels are heteromeric proteins consisting of a pore-forming α(1) subunit and auxiliary α(2)δ and β subunits. Although there are reports of α(2)δ(4) subunit mRNA in the mouse retina and localization of the α(2)δ(4) subunit immunoreactivity to salamander photoreceptor terminals, there is a limited overall understanding of its expression and localization in the retina. α(2)δ(4) subunit expression and distribution in the mouse and rat retina were evaluated by using reverse transcriptase polymerase chain reaction, western blot, and immunohistochemistry with specific primers and a well-characterized antibody to the α(2)δ(4) subunit. α(2)δ(4) subunit mRNA and protein are present in mouse and rat retina, brain, and liver homogenates. Immunostaining for the α(2)δ(4) subunit is mainly localized to Müller cell processes and endfeet, photoreceptor terminals, and photoreceptor outer segments. This subunit is also expressed in a few displaced ganglion cells and bipolar cell dendrites. These findings suggest that the α(2)δ(4) subunit participates in the modulation of L-type Ca(2+) current regulating neurotransmitter release from photoreceptor terminals and Ca(2+)-dependent signaling pathways in bipolar and Müller cells.
Collapse
Affiliation(s)
- Luis Pérez De Sevilla Müller
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California 90095, USA.
| | | | | | | | | |
Collapse
|
47
|
|
48
|
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
|
49
|
Abstract
The transient receptor potential (TRP) channels play a wide variety of essential roles in the sensory systems of various species, both invertebrates and vertebrates. The TRP channel was first identified as a molecule required for proper light response in Drosophila melanogaster. We and another group recently revealed that TRPM1, the founding member of the melanoma-related transient receptor potential (TRPM) subfamily, is required for the photoresponse in mouse retinal ON-bipolar cells. We further demonstrated that Trpm1 is a component of the transduction cation channel negatively regulated by the metabotropic glutamate receptor 6 (mGulR6) cascade in ON-bipolar cells through a reconstitution experiment using CHO cells expressing Trpm1, mGluR6, and Goα. Furthermore, human TRPM1 mutations are associated with congenital stationary night blindness (CSNB), whose patients lack rod function and suffer from night blindness starting in early childhood. In addition to the function of transduction cation channel, TRPM1 is one of the retinal autoantigens in some paraneoplastic retinopathy (PR) associated with retinal ON-bipolar cell dysfunction. In this chapter, we describe physiological functions of the TRPM1 channel and its underlying biochemical mechanisms in retinal ON-bipolar cells in association with CSNB and PR.
Collapse
Affiliation(s)
- Shoichi Irie
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | | |
Collapse
|
50
|
Cain MD, Vo BQ, Kolesnikov AV, Kefalov VJ, Culican SM, Kerschensteiner D, Blumer KJ. An allosteric regulator of R7-RGS proteins influences light-evoked activity and glutamatergic waves in the inner retina. PLoS One 2013; 8:e82276. [PMID: 24349243 PMCID: PMC3857278 DOI: 10.1371/journal.pone.0082276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 10/31/2013] [Indexed: 11/23/2022] Open
Abstract
In the outer retina, G protein-coupled receptor (GPCR) signaling mediates phototransduction and synaptic transmission between photoreceptors and ON bipolar cells. In contrast, the functions of modulatory GPCR signaling networks in the inner retina are less well understood. We addressed this question by determining the consequences of augmenting modulatory Gi/o signaling driven by endogenous transmitters. This was done by analyzing the effects of genetically ablating the R7 RGS-binding protein (R7BP), a membrane-targeting protein and positive allosteric modulator of R7-RGS (regulator of the G protein signaling 7) family that deactivates Gi/oα subunits. We found that R7BP is expressed highly in starburst amacrine cells and retinal ganglion cells (RGCs). As indicated by electroretinography and multielectrode array recordings of adult retina, ablation of R7BP preserved outer retina function, but altered the firing rate and latency of ON RGCs driven by rods and cones but not rods alone. In developing retina, R7BP ablation increased the burst duration of glutamatergic waves whereas cholinergic waves were unaffected. This effect on glutamatergic waves did not result in impaired segregation of RGC projections to eye-specific domains of the dorsal lateral geniculate nucleus. R7BP knockout mice exhibited normal spatial contrast sensitivity and visual acuity as assessed by optomotor reflexes. Taken together these findings indicate that R7BP-dependent regulation of R7-RGS proteins shapes specific aspects of light-evoked and spontaneous activity of RGCs in mature and developing retina.
Collapse
Affiliation(s)
- Matthew D. Cain
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Bradly Q. Vo
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Alexander V. Kolesnikov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Vladimir J. Kefalov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Susan M. Culican
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kendall J. Blumer
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|