1
|
Tetenborg S, Shihabeddin E, Kumar EOAM, Sigulinsky CL, Dedek K, Lin YP, Echeverry FA, Hoff H, Pereda AE, Jones BW, Ribelayga CP, Ebnet K, Matsuura K, O'Brien J. Uncovering the electrical synapse proteome in retinal neurons via in vivo proximity labeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.26.625481. [PMID: 39651118 PMCID: PMC11623651 DOI: 10.1101/2024.11.26.625481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Electrical synapses containing Connexin 36 (Cx36) represent the main means for direct electrical communication among neurons in the mammalian nervous system. However, little is known about the protein complexes that constitute these synapses. In the present study, we applied different BioID strategies to screen the interactomes of Connexin 36 and its zebrafish orthologue Cx35b in retinal neurons. For in vivo proximity labeling in mice, we took advantage of the Cx36-EGFP strain and expressed a GFP-nanobody-TurboID fusion construct selectively in AII amacrine cells. For in vivo BioID in zebrafish, we generated a transgenic line expressing a Cx35b-TurboID fusion under control of the Cx35b promoter. Both strategies allowed us to capture a plethora of molecules that were associated with electrical synapses and showed a high degree of evolutionary conservation in the proteomes of both species. Besides known interactors of Cx36 such as ZO-1 and ZO-2 we have identified more than 50 new proteins, such as scaffold proteins, adhesion molecules and regulators of the cytoskeleton. Moreover, we determined the subcellular localization of these proteins in mouse retina and tested potential binding interactions with Cx36. Amongst these new interactors, we identified signal induced proliferation associated 1 like 3 (Sipa1l3), a protein that has been implicated in cell junction formation and cell polarity, as a new scaffold of electrical synapses. Interestingly, Sipa1l3 was able to interact with ZO-1, ZO-2 and Cx36, suggesting a pivotal role in electrical synapse function. In summary, our study provides the first detailed view of the electrical synapse proteome in retinal neurons, which is likely to apply to electrical synapses elsewhere.
Collapse
|
2
|
Bhoi JD, Goel M, Ribelayga CP, Mangel SC. Circadian clock organization in the retina: From clock components to rod and cone pathways and visual function. Prog Retin Eye Res 2023; 94:101119. [PMID: 36503722 PMCID: PMC10164718 DOI: 10.1016/j.preteyeres.2022.101119] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 12/13/2022]
Abstract
Circadian (24-h) clocks are cell-autonomous biological oscillators that orchestrate many aspects of our physiology on a daily basis. Numerous circadian rhythms in mammalian and non-mammalian retinas have been observed and the presence of an endogenous circadian clock has been demonstrated. However, how the clock and associated rhythms assemble into pathways that support and control retina function remains largely unknown. Our goal here is to review the current status of our knowledge and evaluate recent advances. We describe many previously-observed retinal rhythms, including circadian rhythms of morphology, biochemistry, physiology, and gene expression. We evaluate evidence concerning the location and molecular machinery of the retinal circadian clock, as well as consider findings that suggest the presence of multiple clocks. Our primary focus though is to describe in depth circadian rhythms in the light responses of retinal neurons with an emphasis on clock control of rod and cone pathways. We examine evidence that specific biochemical mechanisms produce these daily light response changes. We also discuss evidence for the presence of multiple circadian retinal pathways involving rhythms in neurotransmitter activity, transmitter receptors, metabolism, and pH. We focus on distinct actions of two dopamine receptor systems in the outer retina, a dopamine D4 receptor system that mediates circadian control of rod/cone gap junction coupling and a dopamine D1 receptor system that mediates non-circadian, light/dark adaptive regulation of gap junction coupling between horizontal cells. Finally, we evaluate the role of circadian rhythmicity in retinal degeneration and suggest future directions for the field of retinal circadian biology.
Collapse
Affiliation(s)
- Jacob D Bhoi
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA; Neuroscience Honors Research Program, William Marsh Rice University, Houston, TX, USA
| | - Manvi Goel
- Department of Neuroscience, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Christophe P Ribelayga
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA; Neuroscience Honors Research Program, William Marsh Rice University, Houston, TX, USA.
| | - Stuart C Mangel
- Department of Neuroscience, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
3
|
Brown-Panton CA, Sabour S, Zoidl GSO, Zoidl C, Tabatabaei N, Zoidl GR. Gap junction Delta-2b ( gjd2b/Cx35.1) depletion causes hyperopia and visual-motor deficiencies in the zebrafish. Front Cell Dev Biol 2023; 11:1150273. [PMID: 36936688 PMCID: PMC10017553 DOI: 10.3389/fcell.2023.1150273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
The zebrafish is a powerful model to investigate the developmental roles of electrical synapses because many signaling pathways that regulate the development of the nervous system are highly conserved from fish to humans. Here, we provide evidence linking the mammalian connexin-36 (Cx36) ortholog gjd2b/Cx35.1, a major component of electrical synapses in the zebrafish, with a refractive error in the context of morphological, molecular, and behavioral changes of zebrafish larvae. Two abnormalities were identified. The optical coherence tomography analysis of the adult retina confirmed changes to the refractive properties caused by eye axial length reduction, leading to hyperopic shifts. The gjd2b/Cx35.1 depletion was also correlated with morphological changes to the head and body ratios in larvae. The differential expression of Wnt/ß-catenin signaling genes, connexins, and dopamine receptors suggested a contribution to the observed phenotypic differences. The alteration of visual-motor behavioral responses to abrupt light transitions was aggravated in larvae, providing evidence that cone photoreceptor cell activity was enhanced when gjd2b/Cx35.1 was depleted. The visual disturbances were reversed under low light conditions in gjd2b -/- /Cx35.1-/- larvae. Since qRT-PCR data demonstrated that two rhodopsin genes were downregulated, we speculated that rod photoreceptor cells in gjd2b/Cx35.1-/- larvae were less sensitive to bright light transitions, thus providing additional evidence that a cone-mediated process caused the VMR light-ON hyperactivity after losing Cx35.1 expression. Together, this study provides evidence for the role of gjd2b/Cx35.1 in the development of the visual system and visually guided behaviors.
Collapse
Affiliation(s)
- Cherie A. Brown-Panton
- Department of Biology, York University, Toronto, ON, Canada
- Center for Vision Research, York University, Toronto, ON, Canada
- *Correspondence: Cherie A. Brown-Panton, ; Georg R. Zoidl,
| | - Shiva Sabour
- Department of Mechanical Engineering, York University, Toronto, ON, Canada
| | - Georg S. O. Zoidl
- Department of Biology, York University, Toronto, ON, Canada
- Center for Vision Research, York University, Toronto, ON, Canada
| | - Christiane Zoidl
- Department of Biology, York University, Toronto, ON, Canada
- Center for Vision Research, York University, Toronto, ON, Canada
| | - Nima Tabatabaei
- Center for Vision Research, York University, Toronto, ON, Canada
- Department of Mechanical Engineering, York University, Toronto, ON, Canada
| | - Georg R. Zoidl
- Department of Biology, York University, Toronto, ON, Canada
- Center for Vision Research, York University, Toronto, ON, Canada
- Department of Psychology, York University, Toronto, ON, Canada
- *Correspondence: Cherie A. Brown-Panton, ; Georg R. Zoidl,
| |
Collapse
|
4
|
Bolstad K, Novales Flamarique I. Chromatic organization of retinal photoreceptors during eye migration of Atlantic halibut (Hippoglossus hippoglossus). J Comp Neurol 2022; 531:256-280. [PMID: 36217253 DOI: 10.1002/cne.25423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 11/08/2022]
Abstract
The retinas of fishes often have single and double cone photoreceptors that are organized in lattice-like mosaics. In flatfishes experiencing eye migration (i.e., the metamorphic process whereby one eye migrates to the other side of the head), the hexagonal lattice of single cones present in the larva undergoes major restructuring resulting in a dominant square mosaic postmetamorphosis consisting of four double cones surrounding each single cone. The expression of different opsin types during eye migration has not been examined despite its importance in understanding photoreceptor plasticity and whether cell fate (in terms of spectral phenotype) could influence square mosaic formation. Here, we probed the retina of Atlantic halibut undergoing eye migration for opsin expression using two antibodies, AHblue and AB5407, that labeled short wavelength sensitive 2 (SWS2) opsin and longer wavelength (predominantly middle wavelength sensitive, RH2) opsins, respectively. Throughout the retina, double and triple cones labeled with AB5407 exclusively, whereas the vast majority of single cones labeled with AHblue. A minority (<5%) of single cones in the square mosaic of the centroventral retina labeled with AB5407. In regions of mosaic transition and near peripheral growth zones, some single cones co-expressed at least two opsins as they labeled with both antibodies. Short wavelength (SWS2 expressing, or S) cones formed a nonrandom mosaic gradient from central to dorsal retina in a region dominated by the larval single cone mosaic. Our results demonstrate the expression of at least two opsins throughout the postmetamorphic retina and suggest opsin switching as a mechanism to create new cone spectral phenotypes. In addition, the S cone gradient at the onset of eye migration may underlie a plastic, cell induction mechanism by which a cone's phenotype determines that of its neighbors and the formation of the square mosaic.
Collapse
Affiliation(s)
- Kennedy Bolstad
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Iñigo Novales Flamarique
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada.,Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| |
Collapse
|
5
|
van der Sande E, Haarman AEG, Quint WH, Tadema KCD, Meester-Smoor MA, Kamermans M, De Zeeuw CI, Klaver CCW, Winkelman BHJ, Iglesias AI. The Role of GJD2(Cx36) in Refractive Error Development. Invest Ophthalmol Vis Sci 2022; 63:5. [PMID: 35262731 PMCID: PMC8934558 DOI: 10.1167/iovs.63.3.5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
Refractive errors are common eye disorders characterized by a mismatch between the focal power of the eye and its axial length. An increased axial length is a common cause of the refractive error myopia (nearsightedness). The substantial increase in myopia prevalence over the last decades has raised public health concerns because myopia can lead to severe ocular complications later in life. Genomewide association studies (GWAS) have made considerable contributions to the understanding of the genetic architecture of refractive errors. Among the hundreds of genetic variants identified, common variants near the gap junction delta-2 (GJD2) gene have consistently been reported as one of the top hits. GJD2 encodes the connexin 36 (Cx36) protein, which forms gap junction channels and is highly expressed in the neural retina. In this review, we provide current evidence that links GJD2(Cx36) to the development of myopia. We summarize the gap junctional communication in the eye and the specific role of GJD2(Cx36) in retinal processing of visual signals. Finally, we discuss the pathways involving dopamine and gap junction phosphorylation and coupling as potential mechanisms that may explain the role of GJD2(Cx36) in refractive error development.
Collapse
Affiliation(s)
- Emilie van der Sande
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
| | - Annechien E. G. Haarman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wim H. Quint
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Kirke C. D. Tadema
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Magda A. Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Maarten Kamermans
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
- Department of Biomedical Physics and Biomedical Photonics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Chris I. De Zeeuw
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caroline C. W. Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Beerend H. J. Winkelman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Adriana I. Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
6
|
Quint WH, Tadema KCD, de Vrieze E, Lukowicz RM, Broekman S, Winkelman BHJ, Hoevenaars M, de Gruiter HM, van Wijk E, Schaeffel F, Meester-Smoor M, Miller AC, Willemsen R, Klaver CCW, Iglesias AI. Loss of Gap Junction Delta-2 (GJD2) gene orthologs leads to refractive error in zebrafish. Commun Biol 2021; 4:676. [PMID: 34083742 PMCID: PMC8175550 DOI: 10.1038/s42003-021-02185-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 05/04/2021] [Indexed: 12/20/2022] Open
Abstract
Myopia is the most common developmental disorder of juvenile eyes, and it has become an increasing cause of severe visual impairment. The GJD2 locus has been consistently associated with myopia in multiple independent genome-wide association studies. However, despite the strong genetic evidence, little is known about the functional role of GJD2 in refractive error development. Here, we find that depletion of gjd2a (Cx35.5) or gjd2b (Cx35.1) orthologs in zebrafish, cause changes in the biometry and refractive status of the eye. Our immunohistological and scRNA sequencing studies show that Cx35.5 (gjd2a) is a retinal connexin and its depletion leads to hyperopia and electrophysiological changes in the retina. These findings support a role for Cx35.5 (gjd2a) in the regulation of ocular biometry. Cx35.1 (gjd2b) has previously been identified in the retina, however, we found an additional lenticular role. Lack of Cx35.1 (gjd2b) led to a nuclear cataract that triggered axial elongation. Our results provide functional evidence of a link between gjd2 and refractive error.
Collapse
Affiliation(s)
- Wim H Quint
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Kirke C D Tadema
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Erik de Vrieze
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rachel M Lukowicz
- Institute of Neuroscience, University of Oregon, Eugene, United States
| | - Sanne Broekman
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Beerend H J Winkelman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Cerebellar Coordination and Cognition, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Melanie Hoevenaars
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Erwin van Wijk
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frank Schaeffel
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Magda Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Adam C Miller
- Institute of Neuroscience, University of Oregon, Eugene, United States
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Adriana I Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
7
|
Aseervatham J, Li X, Mitchell CK, Lin YP, Heidelberger R, O’Brien J. Calmodulin Binding to Connexin 35: Specializations to Function as an Electrical Synapse. Int J Mol Sci 2020; 21:E6346. [PMID: 32882943 PMCID: PMC7504508 DOI: 10.3390/ijms21176346] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 08/27/2020] [Accepted: 08/30/2020] [Indexed: 12/25/2022] Open
Abstract
Calmodulin binding is a nearly universal property of gap junction proteins, imparting a calcium-dependent uncoupling behavior that can serve in an emergency to decouple a stressed cell from its neighbors. However, gap junctions that function as electrical synapses within networks of neurons routinely encounter large fluctuations in local cytoplasmic calcium concentration; frequent uncoupling would be impractical and counterproductive. We have studied the properties and functional consequences of calmodulin binding to the electrical synapse protein Connexin 35 (Cx35 or gjd2b), homologous to mammalian Connexin 36 (Cx36 or gjd2). We find that specializations in Cx35 calmodulin binding sites make it relatively impervious to moderately high levels of cytoplasmic calcium. Calmodulin binding to a site in the C-terminus causes uncoupling when calcium reaches low micromolar concentrations, a behavior prevented by mutations that eliminate calmodulin binding. However, milder stimuli promote calcium/calmodulin-dependent protein kinase II activity that potentiates coupling without interference from calmodulin binding. A second calmodulin binding site in the end of the Cx35 cytoplasmic loop, homologous to a calmodulin binding site present in many connexins, binds calmodulin with very low affinity and stoichiometry. Together, the calmodulin binding sites cause Cx35 to uncouple only at extreme levels of intracellular calcium.
Collapse
Affiliation(s)
- Jaya Aseervatham
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (X.L.); (C.K.M.); (Y.-P.L.)
| | - Xiaofan Li
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (X.L.); (C.K.M.); (Y.-P.L.)
| | - Cheryl K. Mitchell
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (X.L.); (C.K.M.); (Y.-P.L.)
| | - Ya-Ping Lin
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (X.L.); (C.K.M.); (Y.-P.L.)
| | - Ruth Heidelberger
- Department of Neurobiology & Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA;
- The MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - John O’Brien
- Ruiz Department of Ophthalmology & Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.A.); (X.L.); (C.K.M.); (Y.-P.L.)
- The MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
8
|
Pfeiffer RL, Anderson JR, Dahal J, Garcia JC, Yang JH, Sigulinsky CL, Rapp K, Emrich DP, Watt CB, Johnstun HA, Houser AR, Marc RE, Jones BW. A pathoconnectome of early neurodegeneration: Network changes in retinal degeneration. Exp Eye Res 2020; 199:108196. [PMID: 32810483 DOI: 10.1016/j.exer.2020.108196] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/27/2020] [Accepted: 08/11/2020] [Indexed: 10/23/2022]
Abstract
Connectomics has demonstrated that synaptic networks and their topologies are precise and directly correlate with physiology and behavior. The next extension of connectomics is pathoconnectomics: to map neural network synaptology and circuit topologies corrupted by neurological disease in order to identify robust targets for therapeutics. In this report, we characterize a pathoconnectome of early retinal degeneration. This pathoconnectome was generated using serial section transmission electron microscopy to achieve an ultrastructural connectome with 2.18nm/px resolution for accurate identification of all chemical and gap junctional synapses. We observe aberrant connectivity in the rod-network pathway and novel synaptic connections deriving from neurite sprouting. These observations reveal principles of neuron responses to the loss of network components and can be extended to other neurodegenerative diseases.
Collapse
Affiliation(s)
- Rebecca L Pfeiffer
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA.
| | - James R Anderson
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA
| | - Jeebika Dahal
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA
| | - Jessica C Garcia
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA
| | - Jia-Hui Yang
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA
| | | | - Kevin Rapp
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA
| | - Daniel P Emrich
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA
| | - Carl B Watt
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA
| | - Hope Ab Johnstun
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA
| | - Alexis R Houser
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA
| | - Robert E Marc
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA; Signature Immunologics, Torrey, UT, USA
| | - Bryan W Jones
- John Moran Eye Center at the University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
9
|
Moore KB, Mitchell CK, Lin YP, Lee YH, Shihabeddin E, O'Brien J. Localized Calcium Signaling and the Control of Coupling at Cx36 Gap Junctions. eNeuro 2020; 7:ENEURO.0445-19.2020. [PMID: 32179580 PMCID: PMC7168262 DOI: 10.1523/eneuro.0445-19.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/29/2020] [Accepted: 02/21/2020] [Indexed: 01/10/2023] Open
Abstract
A variety of electrical synapses are capable of activity-dependent plasticity, including both activity-dependent potentiation and activity-dependent depression. In several types of neurons, activity-dependent electrical synapse plasticity depends on changes in the local Ca2+ environment. To enable study of local Ca2+ signaling that regulates plasticity, we developed a GCaMP Ca2+ biosensor fused to the electrical synapse protein Connexin 36 (Cx36). Cx36-GCaMP transfected into mammalian cell cultures formed gap junctions at cell-cell boundaries and supported Neurobiotin tracer coupling that was regulated by protein kinase A signaling in the same way as Cx36. Cx36-GCaMP gap junctions robustly reported local Ca2+ increases in response to addition of a Ca2+ ionophore with increases in fluorescence that recovered during washout. Recovery was strongly dependent on Na+-Ca2+ exchange activity. In cells transfected with NMDA receptor subunits, Cx36-GCaMP revealed transient and concentration-dependent increases in local Ca2+ on brief application of glutamate. In HeLa cells, glutamate application increased Cx36-GCaMP tracer coupling through a mechanism that depended in part on Ca2+, calmodulin-dependent protein kinase II (CaMKII) activity. This potentiation of coupling did not require exogenous expression of glutamate receptors, but could be accomplished by endogenously expressed glutamate receptors with pharmacological characteristics reminiscent of NMDA and kainate receptors. Analysis of RNA Sequencing data from HeLa cells confirmed expression of NMDA receptor subunits NR1, NR2C, and NR3B. In summary, Cx36-GCaMP is an effective tool to measure changes in the Ca2+ microenvironment around Cx36 gap junctions. Furthermore, HeLa cells can serve as a model system to study glutamate receptor-driven potentiation of electrical synapses.
Collapse
Affiliation(s)
- Keith B Moore
- Richard S. Ruiz, M.D. Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Cheryl K Mitchell
- Richard S. Ruiz, M.D. Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Ya-Ping Lin
- Richard S. Ruiz, M.D. Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Yuan-Hao Lee
- Richard S. Ruiz, M.D. Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Eyad Shihabeddin
- Richard S. Ruiz, M.D. Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
- The MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030
| | - John O'Brien
- Richard S. Ruiz, M.D. Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
- The MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030
| |
Collapse
|
10
|
Martin EA, Lasseigne AM, Miller AC. Understanding the Molecular and Cell Biological Mechanisms of Electrical Synapse Formation. Front Neuroanat 2020; 14:12. [PMID: 32372919 PMCID: PMC7179694 DOI: 10.3389/fnana.2020.00012] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
In this review article, we will describe the recent advances made towards understanding the molecular and cell biological mechanisms of electrical synapse formation. New evidence indicates that electrical synapses, which are gap junctions between neurons, can have complex molecular compositions including protein asymmetries across joined cells, diverse morphological arrangements, and overlooked similarities with other junctions, all of which indicate new potential roles in neurodevelopmental disease. Aquatic organisms, and in particular the vertebrate zebrafish, have proven to be excellent models for elucidating the molecular mechanisms of electrical synapse formation. Zebrafish will serve as our main exemplar throughout this review and will be compared with other model organisms. We highlight the known cell biological processes that build neuronal gap junctions and compare these with the assemblies of adherens junctions, tight junctions, non-neuronal gap junctions, and chemical synapses to explore the unknown frontiers remaining in our understanding of the critical and ubiquitous electrical synapse.
Collapse
Affiliation(s)
| | | | - Adam C. Miller
- Department of Biology, Institute of Neuroscience, University of Oregon, Eugene, OR, United States
| |
Collapse
|
11
|
Abstract
The complete description of the expression of gap junction proteins in the nervous system of the worm reveals a great complexity of their distribution amongst different neuronal classes, opening an unprecedented opportunity to expose the functional diversity of electrical synapses.
Collapse
Affiliation(s)
- Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
12
|
Telkes I, Kóbor P, Orbán J, Kovács-Öller T, Völgyi B, Buzás P. Connexin-36 distribution and layer-specific topography in the cat retina. Brain Struct Funct 2019; 224:2183-2197. [PMID: 31172263 PMCID: PMC6591202 DOI: 10.1007/s00429-019-01876-y] [Citation(s) in RCA: 5] [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: 10/01/2018] [Accepted: 04/11/2019] [Indexed: 11/29/2022]
Abstract
Connexin-36 (Cx36) is the major constituent of mammalian retinal gap junctions positioned in key signal pathways. Here, we examined the laminar and large-scale topographical distribution of Cx36 punctate immunolabels in the retina of the cat, a classical model of the mammalian visual system. Calretinin-immunoreactive (CaR-IR) cell populations served to outline the nuclear and plexiform layers and to stain specific neuronal populations. CaR-IR cells included horizontal cells in the outer retina, numerous amacrine cells, and scattered cells in the ganglion cell layer. Cx36-IR plaques were found among horizontal cell dendrites albeit without systematic colocalization of the two labels. Diffuse Cx36 immunoreactivity was found in the cytoplasm of AII amacrine cells, but no colocalization of Cx36 plaques was observed with either the perikarya or the long varicose dendrites of the CaR-IR non-AII amacrine cells. Cx36 puncta were seen throughout the entire inner plexiform layer showing their highest density in the ON sublamina. The densities of AII amacrine cell bodies and Cx36 plaques in the ON sublamina were strongly correlated across a wide range of eccentricities suggesting their anatomical association. However, the high number of plaques per AII cell suggests that a considerable fraction of Cx36 gap junctions in the ON sublamina is formed by other cell types than AII amacrine cells drawing attention to extensive but less studied electrically coupled networks.
Collapse
Affiliation(s)
- Ildikó Telkes
- Institute of Physiology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary
| | - Péter Kóbor
- Institute of Physiology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary
| | - József Orbán
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary
- Department of Biophysics, Medical School, University of Pécs, Pécs, 7624, Hungary
| | - Tamás Kovács-Öller
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, 7624, Hungary
- Retinal Electrical Synapses Research Group, MTA-PTE NAP-2, University of Pécs, Pécs, 7624, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, 7624, Hungary
- Retinal Electrical Synapses Research Group, MTA-PTE NAP-2, University of Pécs, Pécs, 7624, Hungary
- Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary
| | - Péter Buzás
- Institute of Physiology, Medical School, University of Pécs, Szigeti út 12, Pécs, 7624, Hungary.
- Szentágothai Research Centre, University of Pécs, Pécs, 7624, Hungary.
- Centre for Neuroscience, University of Pécs, Pécs, 7624, Hungary.
| |
Collapse
|
13
|
O'Brien J, Bloomfield SA. Plasticity of Retinal Gap Junctions: Roles in Synaptic Physiology and Disease. Annu Rev Vis Sci 2018; 4:79-100. [DOI: 10.1146/annurev-vision-091517-034133] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Electrical synaptic transmission via gap junctions underlies direct and rapid neuronal communication in the central nervous system. The diversity of functional roles played by electrical synapses is perhaps best exemplified in the vertebrate retina, in which gap junctions are expressed by each of the five major neuronal types. These junctions are highly plastic; they are dynamically regulated by ambient illumination and circadian rhythms acting through light-activated neuromodulators. The networks formed by electrically coupled neurons provide plastic, reconfigurable circuits positioned to play key and diverse roles in the transmission and processing of visual information at every retinal level. Recent work indicates gap junctions also play a role in the progressive cell death and aberrant activity seen in various pathological conditions of the retina. Gap junctions thus form potential targets for novel neuroprotective therapies in the treatment of neurodegenerative retinal diseases such as glaucoma and ischemic retinopathies.
Collapse
Affiliation(s)
- John O'Brien
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Stewart A. Bloomfield
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY 10036, USA
| |
Collapse
|
14
|
Pang JJ, Yang Z, Jacoby RA, Wu SM. Cone synapses in mammalian retinal rod bipolar cells. J Comp Neurol 2018; 526:1896-1909. [PMID: 29667170 PMCID: PMC6031453 DOI: 10.1002/cne.24456] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 01/25/2023]
Abstract
Some mammalian rod bipolar cells (RBCs) can receive excitatory chemical synaptic inputs from both rods and cones (DBCR2 ), but anatomical evidence for mammalian cone-RBC contacts has been sparse. We examined anatomical cone-RBC contacts using neurobiotin (NB) to visualize individual mouse cones and standard immuno-markers to identify RBCs, cone pedicles and synapses in mouse and baboon retinas. Peanut agglutinin (PNA) stained the basal membrane of all cone pedicles, and mouse cones were positive for red/green (R/G)-opsin, whereas baboon cones were positive for calbindin D-28k. All synapses in the outer plexiform layer were labeled for synaptic vesicle protein 2 (SV2) and PSD (postsynaptic density)-95, and those that coincided with PNA resided closest to bipolar cell somas. Cone-RBC synaptic contacts were identified by: (a) RBC dendrites deeply invaginating into the center of cone pedicles (invaginating synapses), (b) RBC dendritic spines intruding into the surface of cone pedicles (superficial synapses), and (c) PKCα immunoreactivity coinciding with synaptic marker SV2, PSD-95, mGluR6, G protein beta 5 or PNA at cone pedicles. One RBC could form 0-1 invaginating and 1-3 superficial contacts with cones. 20.7% and 38.9% of mouse RBCs contacted cones in the peripheral and central retina (p < .05, n = 14 samples), respectively, while 34.4% (peripheral) and 48.5% (central) of cones contacted RBCs (p > .05). In baboon retinas (n = 4 samples), cone-RBC contacts involved 12.2% of RBCs (n = 416 cells) and 22.5% of cones (n = 225 cells). This suggests that rod and cone signals in the ON pathway are integrated in some RBCs before reaching AII amacrine cells.
Collapse
Affiliation(s)
- Ji-Jie Pang
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, 77030
| | - Zhuo Yang
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, 77030
| | - Roy A Jacoby
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, 77030
| | - Samuel M Wu
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, 77030
| |
Collapse
|
15
|
Cerebrospinal Fluid-Contacting Neurons Sense pH Changes and Motion in the Hypothalamus. J Neurosci 2018; 38:7713-7724. [PMID: 30037834 DOI: 10.1523/jneurosci.3359-17.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 07/04/2018] [Accepted: 07/15/2018] [Indexed: 01/20/2023] Open
Abstract
CSF-contacting (CSF-c) cells are present in the walls of the brain ventricles and the central canal of the spinal cord and found throughout the vertebrate phylum. We recently identified ciliated somatostatin-/GABA-expressing CSF-c neurons in the lamprey spinal cord that act as pH sensors as well as mechanoreceptors. In the same neuron, acidic and alkaline responses are mediated through ASIC3-like and PKD2L1 channels, respectively. Here, we investigate the functional properties of the ciliated somatostatin-/GABA-positive CSF-c neurons in the hypothalamus by performing whole-cell recordings in hypothalamic slices. Depolarizing current pulses readily evoked action potentials, but hypothalamic CSF-c neurons had no or a very low level of spontaneous activity at pH 7.4. They responded, however, with membrane potential depolarization and trains of action potentials to small deviations in pH in both the acidic and alkaline direction. Like in spinal CSF-c neurons, the acidic response in hypothalamic cells is mediated via ASIC3-like channels. In contrast, the alkaline response appears to depend on connexin hemichannels, not on PKD2L1 channels. We also show that hypothalamic CSF-c neurons respond to mechanical stimulation induced by fluid movements along the wall of the third ventricle, a response mediated via ASIC3-like channels. The hypothalamic CSF-c neurons extend their processes dorsally, ventrally, and laterally, but as yet, the effects exerted on hypothalamic circuits are unknown. With similar neurons being present in rodents, the pH- and mechanosensing ability of hypothalamic CSF-c neurons is most likely conserved throughout vertebrate phylogeny.SIGNIFICANCE STATEMENT CSF-contacting neurons are present in all vertebrates and are located mainly in the hypothalamic area and the spinal cord. Here, we report that the somatostatin-/GABA-expressing CSF-c neurons in the lamprey hypothalamus sense bidirectional deviations in the extracellular pH and do so via different molecular mechanisms. They also serve as mechanoreceptors. The hypothalamic CSF-c neurons have extensive axonal ramifications and may decrease the level of motor activity via release of somatostatin. In conclusion, hypothalamic somatostatin-/GABA-expressing CSF-c neurons, as well as their spinal counterpart, represent a novel homeostatic mechanism designed to sense any deviation from physiological pH and thus constitute a feedback regulatory system intrinsic to the CNS, possibly serving a protective role from damage caused by changes in pH.
Collapse
|
16
|
Hoshi H, Sato F. The morphological characterization of orientation-biased displaced large-field ganglion cells in the central part of goldfish retina. J Comp Neurol 2018; 526:243-261. [PMID: 28921532 DOI: 10.1002/cne.24331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 11/10/2022]
Abstract
The vertebrate retina has about 30 subtypes of ganglion cells. Each ganglion cell receives synaptic inputs from specific types of bipolar and amacrine cells ramifying at the same depth of the inner plexiform layer (IPL), each of which is thought to process a specific aspect of visual information. Here, we identified one type of displaced ganglion cell in the goldfish retina which had a large and elongated dendritic field. As a population, all of these ganglion cells were oriented in the horizontal axis and perpendicular to the dorsal-ventral axis of the goldfish eye in the central part of retina. This ganglion cell has previously been classified as Type 1.2. However, the circuit elements which synapse with this ganglion cell are not yet characterized. We found that this displaced ganglion cell was directly tracer-coupled only with homologous ganglion cells at sites containing Cx35/36 puncta. We further illustrated that the processes of dopaminergic neurons often terminated next to intersections between processes of ganglion cells, close to where dopamine D1 receptors were localized. Finally, we showed that Mb1 ON bipolar cells had ribbon synapses in the axonal processes passing through the IPL and made ectopic synapses with this displaced ganglion cell that stratified into stratum 1 of the IPL. These results suggest that the displaced ganglion cell may synapse with both Mb1 cells using ectopic ribbon synapses and OFF cone bipolar cells with regular ribbon synapses in the IPL to function in both scotopic and photopic light conditions.
Collapse
Affiliation(s)
- Hideo Hoshi
- Department of Anatomy, School of Medicine, Toho University, Tokyo, Japan
| | - Fumi Sato
- Department of Anatomy, School of Medicine, Toho University, Tokyo, Japan
| |
Collapse
|
17
|
Nagy JI, Pereda AE, Rash JE. Electrical synapses in mammalian CNS: Past eras, present focus and future directions. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2018; 1860:102-123. [PMID: 28577972 PMCID: PMC5705454 DOI: 10.1016/j.bbamem.2017.05.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/26/2017] [Accepted: 05/27/2017] [Indexed: 12/19/2022]
Abstract
Gap junctions provide the basis for electrical synapses between neurons. Early studies in well-defined circuits in lower vertebrates laid the foundation for understanding various properties conferred by electrical synaptic transmission. Knowledge surrounding electrical synapses in mammalian systems unfolded first with evidence indicating the presence of gap junctions between neurons in various brain regions, but with little appreciation of their functional roles. Beginning at about the turn of this century, new approaches were applied to scrutinize electrical synapses, revealing the prevalence of neuronal gap junctions, the connexin protein composition of many of those junctions, and the myriad diverse neural systems in which they occur in the mammalian CNS. Subsequent progress indicated that electrical synapses constitute key elements in synaptic circuitry, govern the collective activity of ensembles of electrically coupled neurons, and in part orchestrate the synchronized neuronal network activity and rhythmic oscillations that underlie fundamental integrative processes. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- James I Nagy
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - John E Rash
- Department of Biomedical Sciences, and Program in Molecular, Cellular and Integrative Neurosciences, Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
18
|
Noel NCL, Allison WT. Connectivity of cone photoreceptor telodendria in the zebrafish retina. J Comp Neurol 2017; 526:609-625. [PMID: 29127712 DOI: 10.1002/cne.24354] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 01/29/2023]
Abstract
The connectivity amongst photoreceptors is critical to their function, as it underpins lateral inhibition and effective translation of stimuli into neural signals. Despite much work characterizing second-order interneurons in the outer retina, the synapses directly connecting photoreceptors have often been overlooked. Telodendria are fine processes that connect photoreceptor pedicles. They have been observed in diverse vertebrate groups, yet their roles in vision remain speculative. Here, we visualize telodendria via fluorescent protein expression in photoreceptor subtypes. We characterized short wavelength cone telodendria in adult and larval zebrafish retina. Additionally, in the larval retina, we investigated rod telodendria and UV cone telodendria in mutant and transgenic retinas with altered complements of cone types. In the adult retina, telodendria are twice as abundant and branch almost twice as often on blue cones compared to UV cones. Pedicles of neighboring UV and blue cones typically converge into contiguous pairs, despite the regular spacing of their cell bodies. In contrast to adults, larval UV cone telodendria are more numerous (1.3 times) than blue cone telodendria. UV cone telodendria are not detectably affected by ablation of blue cones, and are reduced twofold in mutant larval retina with few UV cones. We thus saw no evidence that telodendria increase in number in the absence of their typical cellular neighbors. We also found that larval rod telodendria are less abundant than short wavelength cone telodendria. In summary, we describe the development and morphology of zebrafish photoreceptor synaptic connectivity toward appreciating the function of telodendria in visual signal processing.
Collapse
Affiliation(s)
- Nicole C L Noel
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - W Ted Allison
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
- Centre for Prions & Protein Folding Disease, University of Alberta, Edmonton, Alberta, Canada
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
19
|
Nagashima M, Hadidjojo J, Barthel LK, Lubensky DK, Raymond PA. Anisotropic Müller glial scaffolding supports a multiplex lattice mosaic of photoreceptors in zebrafish retina. Neural Dev 2017; 12:20. [PMID: 29141686 PMCID: PMC5688757 DOI: 10.1186/s13064-017-0096-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/19/2017] [Indexed: 11/21/2022] Open
Abstract
Background The multiplex, lattice mosaic of cone photoreceptors in the adult fish retina is a compelling example of a highly ordered epithelial cell pattern, with single cell width rows and columns of cones and precisely defined neighbor relationships among different cone types. Cellular mechanisms patterning this multiplex mosaic are not understood. Physical models can provide new insights into fundamental mechanisms of biological patterning. In earlier work, we developed a mathematical model of photoreceptor cell packing in the zebrafish retina, which predicted that anisotropic mechanical tension in the retinal epithelium orients planar polarized adhesive interfaces to align the columns as cone photoreceptors are generated at the retinal margin during post-embryonic growth. Methods With cell-specific fluorescent reporters and in vivo imaging of the growing retinal margin in transparent juvenile zebrafish we provide the first view of how cell packing, spatial arrangement, and cell identity are coordinated to build the lattice mosaic. With targeted laser ablation we probed the tissue mechanics of the retinal epithelium. Results Within the lattice mosaic, planar polarized Crumbs adhesion proteins pack cones into a single cell width column; between columns, N-cadherin-mediated adherens junctions stabilize Müller glial apical processes. The concentration of activated pMyosin II at these punctate adherens junctions suggests that these glial bands are under tension, forming a physical barrier between cone columns and contributing to mechanical stress anisotropies in the epithelial sheet. Unexpectedly, we discovered that the appearance of such parallel bands of Müller glial apical processes precedes the packing of cones into single cell width columns, hinting at a possible role for glia in the initial organization of the lattice mosaic. Targeted laser ablation of Müller glia directly demonstrates that these glial processes support anisotropic mechanical tension in the planar dimension of the retinal epithelium. Conclusions These findings uncovered a novel structural feature of Müller glia associated with alignment of photoreceptors into a lattice mosaic in the zebrafish retina. This is the first demonstration, to our knowledge, of planar, anisotropic mechanical forces mediated by glial cells. Electronic supplementary material The online version of this article (10.1186/s13064-017-0096-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mikiko Nagashima
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 830 North University Avenue, Ann Arbor, MI, 48109-1048, USA
| | - Jeremy Hadidjojo
- Department of Physics, University of Michigan, 450 Church Street, Ann Arbor, MI, 48109-1040, USA
| | - Linda K Barthel
- Microscopy and Image Analysis Laboratory, University of Michigan, Ann Arbor, MI, USA
| | - David K Lubensky
- Department of Physics, University of Michigan, 450 Church Street, Ann Arbor, MI, 48109-1040, USA.
| | - Pamela A Raymond
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 830 North University Avenue, Ann Arbor, MI, 48109-1048, USA.
| |
Collapse
|
20
|
Miller AC, Whitebirch AC, Shah AN, Marsden KC, Granato M, O'Brien J, Moens CB. A genetic basis for molecular asymmetry at vertebrate electrical synapses. eLife 2017; 6. [PMID: 28530549 PMCID: PMC5462537 DOI: 10.7554/elife.25364] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/20/2017] [Indexed: 01/18/2023] Open
Abstract
Neural network function is based upon the patterns and types of connections made between neurons. Neuronal synapses are adhesions specialized for communication and they come in two types, chemical and electrical. Communication at chemical synapses occurs via neurotransmitter release whereas electrical synapses utilize gap junctions for direct ionic and metabolic coupling. Electrical synapses are often viewed as symmetrical structures, with the same components making both sides of the gap junction. By contrast, we show that a broad set of electrical synapses in zebrafish, Danio rerio, require two gap-junction-forming Connexins for formation and function. We find that one Connexin functions presynaptically while the other functions postsynaptically in forming the channels. We also show that these synapses are required for the speed and coordination of escape responses. Our data identify a genetic basis for molecular asymmetry at vertebrate electrical synapses and show they are required for appropriate behavioral performance. DOI:http://dx.doi.org/10.7554/eLife.25364.001
Collapse
Affiliation(s)
- Adam C Miller
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Alex C Whitebirch
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Arish N Shah
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Kurt C Marsden
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, United States
| | - Michael Granato
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, United States
| | - John O'Brien
- Department of Ophthalmology and Visual Science, McGovern Medical School, University of Texas Health Sciences Center at Houston, Houston, United States
| | - Cecilia B Moens
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| |
Collapse
|
21
|
Yoshikawa S, Vila A, Segelken J, Lin YP, Mitchell CK, Nguyen D, O'Brien J. Zebrafish connexin 79.8 (Gja8a): A lens connexin used as an electrical synapse in some neurons. Dev Neurobiol 2016; 77:548-561. [PMID: 27402207 DOI: 10.1002/dneu.22418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/23/2016] [Accepted: 07/05/2016] [Indexed: 11/07/2022]
Abstract
In the mammalian central nervous system, a remarkably small number of connexins is used in electrical synapses, with the majority formed from Cx36. A larger number has been detected in teleosts, with some seeming to serve restricted roles. Here, we report the discovery of a new connexin expressed in the zebrafish lens and a limited set of neurons. Zebrafish cx79.8 (gja8a), previously annotated incorrectly as cx50.5 based on a partial cDNA sequence, is a homologue of mammalian Cx50 (Gja8). We examined its expression through transgenic promoter-reporter constructs, in situ hybridization, and immunolabeling, and examined regulation of coupling in transfected HeLa cells. cx79.8 was expressed most strongly in the lens, but expression was also found in several groups of neurons in the cerebellum and related areas at the midbrain-hindbrain boundary, in cone photoreceptors, and in neurons in the retinal inner nuclear and ganglion cell layers. Labeling in the retina with antibodies against two C-terminal epitopes revealed numerous small punctate spots in the inner plexiform layer and along the somata of cones. Abundant gap junctions were labeled in the outer 1/3 of the lens, but were absent from the center, suggesting that the epitopes or the entire protein was absent from the center. Cx79.8 tracer coupling was strongly regulated by phosphorylation, and was extremely low in control conditions in HeLa cells due to protein phosphatase 2A activity. These properties allow coupling to be strongly restricted in situ, a frequently observed property for electrical synapses. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 548-561, 2017.
Collapse
Affiliation(s)
- Shunichi Yoshikawa
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas
| | - Alejandro Vila
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas.,University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Jasmin Segelken
- Visual Neuroscience, Faculty of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Ya-Ping Lin
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas
| | - Cheryl K Mitchell
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas
| | - Duc Nguyen
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas
| | - John O'Brien
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas.,University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
22
|
Wang HY, Lin YP, Mitchell CK, Ram S, O'Brien J. Two-color fluorescent analysis of connexin 36 turnover: relationship to functional plasticity. J Cell Sci 2015; 128:3888-97. [PMID: 26359298 PMCID: PMC4647165 DOI: 10.1242/jcs.162586] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 09/04/2015] [Indexed: 01/14/2023] Open
Abstract
Gap junctions formed of connexin 36 (Cx36, also known as Gjd2) show tremendous functional plasticity on several time scales. Changes in connexin phosphorylation modify coupling in minutes through an order of magnitude, but recent studies also imply involvement of connexin turnover in regulating cell-cell communication. We utilized Cx36 with an internal HaloTag to study Cx36 turnover and trafficking in cultured cells. Irreversible, covalent pulse-chase labeling with fluorescent HaloTag ligands allowed clear discrimination of newly formed and pre-existing Cx36. Cx36 in junctional plaques turned over with a half-life of 3.1 h, and the turnover rate was unchanged by manipulations of protein kinase A (PKA) activity. In contrast, changes in PKA activity altered coupling within 20 min. New Cx36 in cargo vesicles was added directly to existing gap junctions and newly made Cx36 was not confined to points of addition, but diffused throughout existing gap junctions. Existing connexins also diffused into photobleached areas with a half-time of less than 2 s. In conclusion, studies of Cx36-HaloTag revealed novel features of connexin trafficking and demonstrated that phosphorylation-based changes in coupling occur on a different time scale than turnover.
Collapse
Affiliation(s)
- Helen Yanran Wang
- Ruiz Department of Ophthalmology & Visual Science, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ya-Ping Lin
- Ruiz Department of Ophthalmology & Visual Science, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Cheryl K Mitchell
- Ruiz Department of Ophthalmology & Visual Science, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sripad Ram
- Carl Zeiss Microscopy LLC, Thornwood, NY 10594, USA
| | - John O'Brien
- Ruiz Department of Ophthalmology & Visual Science, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
23
|
Ivanova E, Yee CW, Sagdullaev BT. Increased phosphorylation of Cx36 gap junctions in the AII amacrine cells of RD retina. Front Cell Neurosci 2015; 9:390. [PMID: 26483638 PMCID: PMC4589668 DOI: 10.3389/fncel.2015.00390] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 09/18/2015] [Indexed: 11/24/2022] Open
Abstract
Retinal degeneration (RD) encompasses a family of diseases that lead to photoreceptor death and visual impairment. Visual decline due to photoreceptor cell loss is further compromised by emerging spontaneous hyperactivity in inner retinal cells. This aberrant activity acts as a barrier to signals from the remaining photoreceptors, hindering therapeutic strategies to restore light sensitivity in RD. Gap junctions, particularly those expressed in AII amacrine cells, have been shown to be integral to the generation of aberrant activity. It is unclear whether gap junction expression and coupling are altered in RD. To test this, we evaluated the expression and phosphorylation state of connexin36 (Cx36), the gap junction subunit predominantly expressed in AII amacrine cells, in two mouse models of RD, rd10 (slow degeneration) and rd1 (fast degeneration). Using Ser293-P antibody, which recognizes a phosphorylated form of connexin36, we found that phosphorylation of connexin36 in both slow and fast RD models was significantly greater than in wildtype controls. This elevated phosphorylation may underlie the increased gap junction coupling of AII amacrine cells exhibited by RD retina.
Collapse
Affiliation(s)
- Elena Ivanova
- Departments of Ophthalmology and Neurology, Burke Medical Research Institute, Weill Medical College of Cornell University White Plains, NY, USA
| | - Christopher W Yee
- Departments of Ophthalmology and Neurology, Burke Medical Research Institute, Weill Medical College of Cornell University White Plains, NY, USA
| | - Botir T Sagdullaev
- Departments of Ophthalmology and Neurology, Burke Medical Research Institute, Weill Medical College of Cornell University White Plains, NY, USA
| |
Collapse
|
24
|
Heterotypic gap junctions at glutamatergic mixed synapses are abundant in goldfish brain. Neuroscience 2014; 285:166-93. [PMID: 25451276 DOI: 10.1016/j.neuroscience.2014.10.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/28/2014] [Accepted: 10/29/2014] [Indexed: 11/22/2022]
Abstract
Gap junctions provide for direct intercellular electrical and metabolic coupling. The abundance of gap junctions at "large myelinated club ending (LMCE)" synapses on Mauthner cells (M-cells) of the teleost brain provided a convenient model to correlate anatomical and physiological properties of electrical synapses. There, presynaptic action potentials were found to evoke short-latency electrical "pre-potentials" immediately preceding their accompanying glutamate-induced depolarizations, making these the first unambiguously identified "mixed" (i.e., chemical plus electrical) synapses in the vertebrate CNS. We recently showed that gap junctions at these synapses exhibit asymmetric electrical resistance (i.e., electrical rectification), which we correlated with total molecular asymmetry of connexin composition in their apposing gap junction hemiplaques, with connexin35 (Cx35) restricted to axon terminal hemiplaques and connexin34.7 (Cx34.7) restricted to apposing M-cell plasma membranes. We now show that similarly heterotypic neuronal gap junctions are abundant throughout goldfish brain, with labeling exclusively for Cx35 in presynaptic hemiplaques and exclusively for Cx34.7 in postsynaptic hemiplaques. Moreover, the vast majority of these asymmetric gap junctions occur at glutamatergic axon terminals. The widespread distribution of heterotypic gap junctions at glutamatergic mixed synapses throughout goldfish brain and spinal cord implies that pre- vs. postsynaptic asymmetry at electrical synapses evolved early in the chordate lineage. We propose that the advantages of the molecular and functional asymmetry of connexins at electrical synapses that are so prominently expressed in the teleost CNS are unlikely to have been abandoned in higher vertebrates. However, to create asymmetric coupling in mammals, where most gap junctions are composed of connexin36 (Cx36) on both sides, would require some other mechanism, such as differential phosphorylation of connexins on opposite sides of the same gap junction or on asymmetric differences in the complement of their scaffolding and regulatory proteins.
Collapse
|
25
|
Emran F, Dowling JE. Circadian Rhythms and Vision in Zebrafish. THE RETINA AND CIRCADIAN RHYTHMS 2014:171-193. [DOI: 10.1007/978-1-4614-9613-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
26
|
Li H, Zhang Z, Blackburn MR, Wang SW, Ribelayga CP, O'Brien J. Adenosine and dopamine receptors coregulate photoreceptor coupling via gap junction phosphorylation in mouse retina. J Neurosci 2013; 33:3135-50. [PMID: 23407968 PMCID: PMC3711184 DOI: 10.1523/jneurosci.2807-12.2013] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 12/20/2012] [Accepted: 12/24/2012] [Indexed: 11/21/2022] Open
Abstract
Gap junctions in retinal photoreceptors suppress voltage noise and facilitate input of rod signals into the cone pathway during mesopic vision. These synapses are highly plastic and regulated by light and circadian clocks. Recent studies have revealed an important role for connexin36 (Cx36) phosphorylation by protein kinase A (PKA) in regulating cell-cell coupling. Dopamine is a light-adaptive signal in the retina, causing uncoupling of photoreceptors via D4 receptors (D4R), which inhibit adenylyl cyclase (AC) and reduce PKA activity. We hypothesized that adenosine, with its extracellular levels increasing in darkness, may serve as a dark signal to coregulate photoreceptor coupling through modulation of gap junction phosphorylation. Both D4R and A2a receptor (A2aR) mRNAs were present in photoreceptors, inner nuclear layer neurons, and ganglion cells in C57BL/6 mouse retina, and showed cyclic expression with partially overlapping rhythms. Pharmacologically activating A2aR or inhibiting D4R in light-adapted daytime retina increased photoreceptor coupling. Cx36 among photoreceptor terminals, representing predominantly rod-cone gap junctions but possibly including some rod-rod and cone-cone gap junctions, was phosphorylated in a PKA-dependent manner by the same treatments. Conversely, inhibiting A2aR or activating D4R in daytime dark-adapted retina decreased Cx36 phosphorylation with similar PKA dependence. A2a-deficient mouse retina showed defective regulation of photoreceptor gap junction phosphorylation, fairly regular dopamine release, and moderately downregulated expression of D4R and AC type 1 mRNA. We conclude that adenosine and dopamine coregulate photoreceptor coupling through opposite action on the PKA pathway and Cx36 phosphorylation. In addition, loss of the A2aR hampered D4R gene expression and function.
Collapse
MESH Headings
- Adenylyl Cyclases/metabolism
- Animals
- Chromatography, High Pressure Liquid
- Connexins/metabolism
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Dark Adaptation/physiology
- Gap Junctions/metabolism
- Gap Junctions/physiology
- Gene Expression/physiology
- Image Processing, Computer-Assisted
- Immunohistochemistry
- In Situ Hybridization
- In Vitro Techniques
- Mice
- Mice, Inbred C57BL
- Phosphorylation
- Real-Time Polymerase Chain Reaction
- Receptors, Adenosine A2/genetics
- Receptors, Adenosine A2/physiology
- Receptors, Dopamine/genetics
- Receptors, Dopamine/physiology
- Receptors, Dopamine D4/biosynthesis
- Receptors, Dopamine D4/genetics
- Receptors, Purinergic P1/genetics
- Receptors, Purinergic P1/physiology
- Retinal Cone Photoreceptor Cells/physiology
- Retinal Rod Photoreceptor Cells/physiology
- Gap Junction delta-2 Protein
Collapse
Affiliation(s)
- Hongyan Li
- Richard S. Ruiz, MD, Department of Ophthalmology and Visual Science, The University of Texas Medical School and
| | - Zhijing Zhang
- Richard S. Ruiz, MD, Department of Ophthalmology and Visual Science, The University of Texas Medical School and
| | - Michael R. Blackburn
- Department of Biochemistry and Molecular Biology, The University of Texas Medical School, Houston, Texas 77030; and
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas 77030
| | - Steven W. Wang
- Richard S. Ruiz, MD, Department of Ophthalmology and Visual Science, The University of Texas Medical School and
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas 77030
| | - Christophe P. Ribelayga
- Richard S. Ruiz, MD, Department of Ophthalmology and Visual Science, The University of Texas Medical School and
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas 77030
| | - John O'Brien
- Richard S. Ruiz, MD, Department of Ophthalmology and Visual Science, The University of Texas Medical School and
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas 77030
| |
Collapse
|
27
|
Völgyi B, Kovács-Oller T, Atlasz T, Wilhelm M, Gábriel R. Gap junctional coupling in the vertebrate retina: variations on one theme? Prog Retin Eye Res 2013; 34:1-18. [PMID: 23313713 DOI: 10.1016/j.preteyeres.2012.12.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 12/18/2012] [Accepted: 12/28/2012] [Indexed: 10/27/2022]
Abstract
Gap junctions connect cells in the bodies of all multicellular organisms, forming either homologous or heterologous (i.e. established between identical or different cell types, respectively) cell-to-cell contacts by utilizing identical (homotypic) or different (heterotypic) connexin protein subunits. Gap junctions in the nervous system serve electrical signaling between neurons, thus they are also called electrical synapses. Such electrical synapses are particularly abundant in the vertebrate retina where they are specialized to form links between neurons as well as glial cells. In this article, we summarize recent findings on retinal cell-to-cell coupling in different vertebrates and identify general features in the light of the evergrowing body of data. In particular, we describe and discuss tracer coupling patterns, connexin proteins, junctional conductances and modulatory processes. This multispecies comparison serves to point out that most features are remarkably conserved across the vertebrate classes, including (i) the cell types connected via electrical synapses; (ii) the connexin makeup and the conductance of each cell-to-cell contact; (iii) the probable function of each gap junction in retinal circuitry; (iv) the fact that gap junctions underlie both electrical and/or tracer coupling between glial cells. These pan-vertebrate features thus demonstrate that retinal gap junctions have changed little during the over 500 million years of vertebrate evolution. Therefore, the fundamental architecture of electrically coupled retinal circuits seems as old as the retina itself, indicating that gap junctions deeply incorporated in retinal wiring from the very beginning of the eye formation of vertebrates. In addition to hard wiring provided by fast synaptic transmitter-releasing neurons and soft wiring contributed by peptidergic, aminergic and purinergic systems, electrical coupling may serve as the 'skeleton' of lateral processing, enabling important functions such as signal averaging and synchronization.
Collapse
Affiliation(s)
- Béla Völgyi
- Department of Ophthalmology, School of Medicine, New York University, 550 First Avenue, MSB 149, New York, NY 10016, USA.
| | | | | | | | | |
Collapse
|
28
|
|
29
|
Vila A, Satoh H, Rangel C, Mills SL, Hoshi H, O'Brien J, Marshak DR, Macleish PR, Marshak DW. Histamine receptors of cones and horizontal cells in Old World monkey retinas. J Comp Neurol 2012; 520:528-43. [PMID: 21800315 DOI: 10.1002/cne.22731] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In primates the retina receives input from histaminergic neurons in the posterior hypothalamus that are active during the day. In order to understand how this input contributes to information processing in Old World monkey retinas, we have been localizing histamine receptors (HR) and studying the effects of histamine on the neurons that express them. Previously, we localized HR3 to the tips of ON bipolar cell dendrites and showed that histamine hyperpolarizes the cells via this receptor. We raised antisera against synthetic peptides corresponding to an extracellular domain of HR1 between the 4th and 5th transmembrane domains and to an intracellular domain near the carboxyl terminus of HR2. Using these, we localized HR1 to horizontal cells and a small number of amacrine cells and localized HR2 to puncta closely associated with synaptic ribbons inside cone pedicles. Consistent with this, HR1 mRNA was detected in horizontal cell perikarya and primary dendrites and HR2 mRNA was found in cone inner segments. We studied the effect of 5 μM exogenous histamine on primate cones in macaque retinal slices. Histamine reduced I(h) at moderately hyperpolarized potentials, but not the maximal current. This would be expected to increase the operating range of cones and conserve ATP in bright, ambient light. Thus, all three major targets of histamine are in the outer plexiform layer, but the retinopetal axons containing histamine terminate in the inner plexiform layer. Taken together, the findings in these three studies suggest that histamine acts primarily via volume transmission in primate retina.
Collapse
Affiliation(s)
- Alejandro Vila
- Department of Neurobiology and Anatomy, Medical School, University of Texas at Health Science Center at Houston, Houston, Texas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Connexin composition in apposed gap junction hemiplaques revealed by matched double-replica freeze-fracture replica immunogold labeling. J Membr Biol 2012; 245:333-44. [PMID: 22760604 PMCID: PMC3401501 DOI: 10.1007/s00232-012-9454-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 06/08/2012] [Indexed: 10/28/2022]
Abstract
Despite the combination of light-microscopic immunocytochemistry, histochemical mRNA detection techniques and protein reporter systems, progress in identifying the protein composition of neuronal versus glial gap junctions, determination of the differential localization of their constituent connexin proteins in two apposing membranes and understanding human neurological diseases caused by connexin mutations has been problematic due to ambiguities introduced in the cellular and subcellular assignment of connexins. Misassignments occurred primarily because membranes and their constituent proteins are below the limit of resolution of light microscopic imaging techniques. Currently, only serial thin-section transmission electron microscopy and freeze-fracture replica immunogold labeling have sufficient resolution to assign connexin proteins to either or both sides of gap junction plaques. However, freeze-fracture replica immunogold labeling has been limited because conventional freeze fracturing allows retrieval of only one of the two membrane fracture faces within a gap junction, making it difficult to identify connexin coupling partners in hemiplaques removed by fracturing. We now summarize progress in ascertaining the connexin composition of two coupled hemiplaques using matched double-replicas that are labeled simultaneously for multiple connexins. This approach allows unambiguous identification of connexins and determination of the membrane "sidedness" and the identities of connexin coupling partners in homotypic and heterotypic gap junctions of vertebrate neurons.
Collapse
|
31
|
Tarboush R, Chapman GB, Connaughton VP. Ultrastructure of the distal retina of the adult zebrafish, Danio rerio. Tissue Cell 2012; 44:264-79. [PMID: 22608306 DOI: 10.1016/j.tice.2012.04.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 02/07/2012] [Accepted: 04/20/2012] [Indexed: 01/01/2023]
Abstract
The organization, morphological characteristics, and synaptic structure of photoreceptors in the adult zebrafish retina were studied using light and electron microscopy. Adult photoreceptors show a typical ordered tier arrangement with rods easily distinguished from cones based on outer segment (OS) morphology. Both rods and cones contain mitochondria within the inner segments (IS), including the large, electron-dense megamitochondria previously described (Kim et al.) Four major ultrastructural differences were observed between zebrafish rods and cones: (1) the membranes of cone lamellar disks showed a wider variety of relationships to the plasma membrane than those of rods, (2) cone pedicles typically had multiple synaptic ribbons, while rod spherules had 1-2 ribbons, (3) synaptic ribbons in rod spherules were ∼2 times longer than ribbons in cone pedicles, and (4) rod spherules had a more electron-dense cytoplasm than cone pedicles. Examination of photoreceptor terminals identified four synaptic relationships at cone pedicles: (1) invaginating contacts postsynaptic to cone ribbons forming dyad, triad, and quadrad synapses, (2) presumed gap junctions connecting adjacent postsynaptic processes invaginating into cone terminals, (3) basal junctions away from synaptic ribbons, and (4) gap junctions between adjacent photoreceptor terminals. More vitread and slightly farther removed from photoreceptor terminals, extracellular microtubule-like structures were identified in association with presumed horizontal cell processes in the OPL. These findings, the first to document the ultrastructure of the distal retina in adult zebrafish, indicate that zebrafish photoreceptors have many characteristics similar to other species, further supporting the use of zebrafish as a model for the vertebrate visual system.
Collapse
Affiliation(s)
- R Tarboush
- Department of Biology, American University, Washington, DC 20016, USA.
| | | | | |
Collapse
|
32
|
Hamzei-Sichani F, Davidson KGV, Yasumura T, Janssen WGM, Wearne SL, Hof PR, Traub RD, Gutiérrez R, Ottersen OP, Rash JE. Mixed Electrical-Chemical Synapses in Adult Rat Hippocampus are Primarily Glutamatergic and Coupled by Connexin-36. Front Neuroanat 2012; 6:13. [PMID: 22615687 PMCID: PMC3351785 DOI: 10.3389/fnana.2012.00013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Accepted: 04/19/2012] [Indexed: 01/08/2023] Open
Abstract
Dendrodendritic electrical signaling via gap junctions is now an accepted feature of neuronal communication in mammalian brain, whereas axodendritic and axosomatic gap junctions have rarely been described. We present ultrastructural, immunocytochemical, and dye-coupling evidence for “mixed” (electrical/chemical) synapses on both principal cells and interneurons in adult rat hippocampus. Thin-section electron microscopic images of small gap junction-like appositions were found at mossy fiber (MF) terminals on thorny excrescences of CA3 pyramidal neurons (CA3pyr), apparently forming glutamatergic mixed synapses. Lucifer Yellow injected into weakly fixed CA3pyr was detected in MF axons that contacted four injected CA3pyr, supporting gap junction-mediated coupling between those two types of principal cells. Freeze-fracture replica immunogold labeling revealed diverse sizes and morphologies of connexin-36-containing gap junctions throughout hippocampus. Of 20 immunogold-labeled gap junctions, seven were large (328–1140 connexons), three of which were consistent with electrical synapses between interneurons; but nine were at axon terminal synapses, three of which were immediately adjacent to distinctive glutamate receptor-containing postsynaptic densities, forming mixed glutamatergic synapses. Four others were adjacent to small clusters of immunogold-labeled 10-nm E-face intramembrane particles, apparently representing extrasynaptic glutamate receptor particles. Gap junctions also were on spines in stratum lucidum, stratum oriens, dentate gyrus, and hilus, on both interneurons and unidentified neurons. In addition, one putative GABAergic mixed synapse was found in thin-section images of a CA3pyr, but none were found by immunogold labeling, suggesting the rarity of GABAergic mixed synapses. Cx36-containing gap junctions throughout hippocampus suggest the possibility of reciprocal modulation of electrical and chemical signals in diverse hippocampal neurons.
Collapse
|
33
|
O'Brien JJ, Chen X, MacLeish PR, O'Brien J, Massey SC. Photoreceptor coupling mediated by connexin36 in the primate retina. J Neurosci 2012; 32:4675-87. [PMID: 22457514 PMCID: PMC3335500 DOI: 10.1523/jneurosci.4749-11.2012] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 02/15/2012] [Accepted: 02/17/2012] [Indexed: 11/21/2022] Open
Abstract
Photoreceptors are coupled via gap junctions in many mammalian species. Cone-to-cone coupling is thought to improve sensitivity and signal-to-noise ratio, while rod-to-cone coupling provides an alternative rod pathway active under twilight or mesopic conditions (Smith et al., 1986; DeVries et al., 2002; Hornstein et al., 2005). Gap junctions are composed of connexins, and connexin36 (Cx36), the dominant neuronal connexin, is expressed in the outer plexiform layer. Primate (Macaca mulatta) cone pedicles, labeled with an antibody against cone arrestin (7G6) were connected by a network of fine processes called telodendria and, in double-labeled material, Cx36 plaques were located precisely at telodendrial contacts between cones, suggesting strongly they are Cx36 gap junctions. Each red/green cone made nonselective connections with neighboring red/green cones. In contrast, blue cone pedicles were smaller with relatively few short telodendria and they made only rare or equivocal Cx36 contacts with adjacent cones. There were also many smaller Cx36 plaques around the periphery of every cone pedicle and along a series of very fine telodendria that were too short to reach adjacent members of the cone pedicle mosaic. These small Cx36 plaques were closely aligned with nearly every rod spherule and may identify sites of rod-to-cone coupling, even though the identity of the rod connexin has not been established. We conclude that the matrix of cone telodendria is the substrate for photoreceptor coupling. Red/green cones were coupled indiscriminately but blue cones were rarely connected with other cones. All cone types, including blue cones, made gap junctions with surrounding rod spherules.
Collapse
Affiliation(s)
- Jennifer J. O'Brien
- Department of Ophthalmology and Visual Science, University of Texas Medical School at Houston, Houston, Texas 77030, and
| | - Xiaoming Chen
- Neuroscience Institute, Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - Peter R. MacLeish
- Neuroscience Institute, Department of Neurobiology, Morehouse School of Medicine, Atlanta, Georgia 30310
| | - John O'Brien
- Department of Ophthalmology and Visual Science, University of Texas Medical School at Houston, Houston, Texas 77030, and
| | - Stephen C. Massey
- Department of Ophthalmology and Visual Science, University of Texas Medical School at Houston, Houston, Texas 77030, and
| |
Collapse
|
34
|
Rivera-Rivera NL, Martinez-Rivera N, Torres-Vazquez I, Serrano-Velez JL, Lauder GV, Rosa-Molinar E. A male poecillid's sexually dimorphic body plan, behavior, and nervous system. Integr Comp Biol 2010; 50:1081-90. [PMID: 21082070 DOI: 10.1093/icb/icq147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Here we review the literature of a male poecillid's sexually dimorphic body plan, behavior, and nervous system, including work dating from the mid 1800s to the mid 1990s as well as work in press or in preparation for publication. Rosa-Molinar described the remodeling of the sexually dimorphic anal fin appendicular support, confirmed earlier claims about the development of the male and female secondary sex characteristics in the Western Mosquitofish, Gambusia affinis and provided for the first time direct embryonic evidence suggesting that remodeling of the sexually dimorphic anal fin appendicular support is biphasic. The first process begins in embryos and proceeds similarly in immature males and females; the second process occurs only in males and results in the anterior transposition of the anal fin and its appendicular support to the level of vertebra 11 [Rosa-Molinar E, Hendricks SE, Rodriguez-Sierra JF, Fritzsch B. 1994. Development of the anal fin appendicular support in the western mosquitofish, Gambusia affinis (Baird and Girard, 1854): a reinvestigation and reinterpretation. Acta Anat 151:20-35.] and the formation of a gonopodium used for internal fertilization. Studies using high-speed video cameras confirmed and extended Peden's and others' observations of copulatory behavior. The cameras showed that circumduction is a complex movement combining in a very fast sequence abduction, extension and pronation, S-start-type fast-start (defined as torque-thrust), and adduction movements. Recent work on the nervous system demonstrated dye-coupling between motor neurons and interneurons via gap junctions, suggesting an attractive substrate for the rapid motions involved in poecillid copulatory reflexes.
Collapse
Affiliation(s)
- Nydia L Rivera-Rivera
- Biological Imaging Group, University of Puerto Rico-Rio Piedras, San Juan, Puerto Rico
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
Gap junctions are aggregates of intercellular channels that permit direct cell-cell transfer of ions and small molecules. Initially described as low-resistance ion pathways joining excitable cells (nerve and muscle), gap junctions are found joining virtually all cells in solid tissues. Their long evolutionary history has permitted adaptation of gap-junctional intercellular communication to a variety of functions, with multiple regulatory mechanisms. Gap-junctional channels are composed of hexamers of medium-sized families of integral proteins: connexins in chordates and innexins in precordates. The functions of gap junctions have been explored by studying mutations in flies, worms, and humans, and targeted gene disruption in mice. These studies have revealed a wide diversity of function in tissue and organ biology.
Collapse
|
36
|
Photoreceptor coupling is controlled by connexin 35 phosphorylation in zebrafish retina. J Neurosci 2009; 29:15178-86. [PMID: 19955370 DOI: 10.1523/jneurosci.3517-09.2009] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Electrical coupling of neurons is widespread throughout the CNS and is observed among retinal photoreceptors from essentially all vertebrates. Coupling dampens voltage noise in photoreceptors and rod-cone coupling provides a means for rod signals to enter the cone pathway, extending the dynamic range of rod-mediated vision. This coupling is dynamically regulated by a circadian rhythm and light adaptation. We examined the molecular mechanism that controls photoreceptor coupling in zebrafish retina. Connexin 35 (homologous to Cx36 of mammals) was found at both cone-cone and rod-cone gap junctions. Photoreceptors showed strong Neurobiotin tracer coupling at night, extensively labeling the network of cones. Tracer coupling was significantly reduced in the daytime, showing a 20-fold lower diffusion coefficient for Neurobiotin transfer. The phosphorylation state of Cx35 at two regulatory phosphorylation sites, Ser110 and Ser276, was directly related to tracer coupling. Phosphorylation was high at night and low during the day. Protein kinase A (PKA) activity directly controlled both phosphorylation state and tracer coupling. Both were significantly increased in the day by pharmacological activation of PKA and significantly reduced at night by inhibition of PKA. The data are consistent with direct phosphorylation of Cx35 by PKA. We conclude that the magnitude of photoreceptor coupling is controlled by the dynamic phosphorylation and dephosphorylation of Cx35. Furthermore, the nighttime state is characterized by extensive coupling that results in a well connected cone network.
Collapse
|
37
|
Expression of connexin 35/36 in retinal horizontal and bipolar cells of carp. Neuroscience 2009; 164:1161-9. [DOI: 10.1016/j.neuroscience.2009.09.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2008] [Revised: 09/16/2009] [Accepted: 09/16/2009] [Indexed: 11/23/2022]
|
38
|
Kothmann WW, Massey SC, O'Brien J. Dopamine-stimulated dephosphorylation of connexin 36 mediates AII amacrine cell uncoupling. J Neurosci 2009; 29:14903-11. [PMID: 19940186 PMCID: PMC2839935 DOI: 10.1523/jneurosci.3436-09.2009] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 09/24/2009] [Accepted: 10/16/2009] [Indexed: 11/21/2022] Open
Abstract
Gap junction proteins form the substrate for electrical coupling between neurons. These electrical synapses are widespread in the CNS and serve a variety of important functions. In the retina, connexin 36 (Cx36) gap junctions couple AII amacrine cells and are a requisite component of the high-sensitivity rod photoreceptor pathway. AII amacrine cell coupling strength is dynamically regulated by background light intensity, and uncoupling is thought to be mediated by dopamine signaling via D(1)-like receptors. One proposed mechanism for this uncoupling involves dopamine-stimulated phosphorylation of Cx36 at regulatory sites, mediated by protein kinase A. Here we provide evidence against this hypothesis and demonstrate a direct relationship between Cx36 phosphorylation and AII amacrine cell coupling strength. Dopamine receptor-driven uncoupling of the AII network results from protein kinase A activation of protein phosphatase 2A and subsequent dephosphorylation of Cx36. Protein phosphatase 1 activity negatively regulates this pathway. We also find that Cx36 gap junctions can exist in widely different phosphorylation states within a single neuron, implying that coupling is controlled at the level of individual gap junctions by locally assembled signaling complexes. This kind of synapse-by-synapse plasticity allows for precise control of neuronal coupling, as well as cell-type-specific responses dependent on the identity of the signaling complexes assembled.
Collapse
Affiliation(s)
- W Wade Kothmann
- Richard S. Ruiz Department of Ophthalmology, University of Texas Medical School at Houston, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
39
|
HOSHI HIDEO, MILLS STEPHENL. Components and properties of the G3 ganglion cell circuit in the rabbit retina. J Comp Neurol 2009; 513:69-82. [PMID: 19107780 PMCID: PMC2834241 DOI: 10.1002/cne.21941] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Each point on the retina is sampled by about 15 types of ganglion cell, each of which is an element in a circuit also containing specific types of bipolar cell and amacrine cell. Only a few of these circuits are well characterized. We found that intracellular injection of Neurobiotin into a specific ganglion cell type targeted by fluorescent markers also stained an asymmetrically branching ganglion cell. It was also tracer-coupled to an unusual type of amacrine cell whose dendrites were strongly asymmetric, coursing in a narrow bundle from the soma in the dorsal direction only. The dendritic field of the ganglion cell stratifies initially in sublamina b (the ON layers), but with few specializations and branches, and then more extensively in sublamina a (the OFF layers) at the level of the processes of the coupled amacrine cell. Intersections of the ganglion and amacrine cell processes contain puncta immunopositive for Cx36. Additionally, we found that the dopaminergic amacrine cell makes contact with both the ganglion cell and the amacrine cell, and that a bipolar cell immunopositive for calbindin synapses onto the sublamina b processes of the ganglion cell. Dopamine D(1) receptor activation reduced tracer flow to the amacrine cells. We have thus targeted and characterized two poorly understood retinal cell types and placed them with two other cell types in a substantial portion of a new retinal circuit. This unique circuit comprised of pronounced asymmetries in the ganglion cell and amacrine cell dendritic fields may result in a substantial orientation bias.
Collapse
Affiliation(s)
- HIDEO HOSHI
- Department of Ophthalmology and Visual Science, University of Texas at Houston, Houston, Texas 77030
| | - STEPHEN L. MILLS
- Department of Ophthalmology and Visual Science, University of Texas at Houston, Houston, Texas 77030
| |
Collapse
|
40
|
Ramsden SD, Anderson L, Mussi M, Kamermans M, Hawryshyn CW. Retinal processing and opponent mechanisms mediating ultraviolet polarization sensitivity in rainbow trout (Oncorhynchus mykiss). ACTA ACUST UNITED AC 2008; 211:1376-85. [PMID: 18424671 DOI: 10.1242/jeb.015941] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A number of teleost fishes have photoreceptor mechanisms to detect linearly polarized light. We studied the neuronal mechanism underlying this ability. It was found that a polarized signal could be detected in rainbow trout (Oncorhynchus mykiss) both in the electroretinogram (ERG) and in the compound action potential (CAP) measured in the optic nerve, indicating a strong retinal contribution to the processing of polarized light. The CAP recordings showed a W-shaped sensitivity curve, with a peak at 0 degrees , 90 degrees and 180 degrees , consistent with processes for both vertical and horizontal orientation. By contrast, the ERG recordings reveal a more complex pattern. In addition to the peaks at 0 degrees , 90 degrees and 180 degrees , two additional peaks appeared at 45 degrees and 135 degrees . This result suggests a specialized contribution of the outer retina in the processing of polarized light. The spectral sensitivity of the mechanisms responsible for these intermediate peaks was studied using chromatic adaptation. Here we show that long wavelength-sensitive (LWS) cone mechanism adaptation shifted the intermediate peaks towards 90 degrees , whereas ultraviolet-sensitive (UVS) cone mechanism adaptation shifted the peaks away from 90 degrees towards either 0 degrees or 180 degrees . These results provide further confirmation that the 90 degrees peak is dominated by the LWS cone mechanism and the 0 degrees and 180 degrees peaks are dominated by the UVS cone mechanism. In addition, a pharmacological approach was used to examine the retinal neural mechanisms underlying polarization sensitivity. The effect of blocking negative feedback from horizontal cells to cones on the ERG was studied by making intraocular injections of low doses of cobalt, known to block this feedback pathway. It was found that the intermediate peaks seen in the ERG polarization sensitivity curves were eliminated after application of cobalt, suggesting that these peaks are due to outer retinal inhibition derived from feedback of horizontal cells onto cones. A simple computational model was developed to evaluate these results. The model consists of opponent and non-opponent processing elements for the two polarization detectors. This model provides a first approximation analysis suggesting that opponent processing occurs in the outer retina for polarization vision. Although it seems that polarization vision uses a slightly more complicated coding scheme than colour vision, the results presented in this paper suggest that opponent and non-opponent channels process polarization information.
Collapse
Affiliation(s)
- Samuel D Ramsden
- Department of Biology, University of Victoria, PO Box 3020 STN CSC, Victoria, British Columbia, V8W 3N5, Canada
| | | | | | | | | |
Collapse
|
41
|
Innocenti B, Heidelberger R. Mechanisms contributing to tonic release at the cone photoreceptor ribbon synapse. J Neurophysiol 2007; 99:25-36. [PMID: 17989244 DOI: 10.1152/jn.00737.2007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Time-resolved capacitance measurements in combination with fluorescence measurements of internal calcium suggested three kinetic components of release in acutely isolated cone photoreceptors of the tiger salamander. A 45-fF releasable pool, corresponding to about 1,000 vesicles, was identified. This pool could be depleted with a time constant of a few hundred milliseconds and its recovery from depletion was quite rapid (tau approximately 1 s). The fusion of vesicles in this pool was blocked by low-millimolar EGTA. Endocytosis was sufficiently slow that it is likely that refilling of the releasable pool occurred from preformed vesicles. A second, slower component of release (tau(depletion) approximately 3 s) was identified that was approximately twice the size of the releasable pool. This pool may serve as a first reserve pool that replenishes the releasable pool. Computer simulations indicate that the properties of the releasable and first reserve pools are sufficient to maintain synaptic signaling for several seconds in the face of near-maximal stimulations and in the absence of other sources of vesicles. Along with lower rates of depletion, additional mechanisms, such as replenishment from distal reserve pools and the fast recycling of vesicles, may further contribute to the maintenance of graded, tonic release from cone photoreceptors.
Collapse
Affiliation(s)
- Barbara Innocenti
- The W. M. Keck Center for the Neurobiology of Learning and Memory, University of Texas Medical School at Houston, 6431 Fannin Street, Houston, TX 77025, USA.
| | | |
Collapse
|
42
|
Pan F, Mills SL, Massey SC. Screening of gap junction antagonists on dye coupling in the rabbit retina. Vis Neurosci 2007; 24:609-18. [PMID: 17711600 PMCID: PMC2213422 DOI: 10.1017/s0952523807070472] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Accepted: 05/11/2007] [Indexed: 11/07/2022]
Abstract
Many cell types in the retina are coupled via gap junctions and so there is a pressing need for a potent and reversible gap junction antagonist. We screened a series of potential gap junction antagonists by evaluating their effects on dye coupling in the network of A-type horizontal cells. We evaluated the following compounds: meclofenamic acid (MFA), mefloquine, 2-aminoethyldiphenyl borate (2-APB), 18-alpha-glycyrrhetinic acid, 18-beta-glycyrrhetinic acid (18-beta-GA), retinoic acid, flufenamic acid, niflumic acid, and carbenoxolone. The efficacy of each drug was determined by measuring the diffusion coefficient for Neurobiotin (Mills & Massey, 1998). MFA, 18-beta-GA, 2-APB and mefloquine were the most effective antagonists, completely eliminating A-type horizontal cell coupling at a concentration of 200 muM. Niflumic acid, flufenamic acid, and carbenoxolone were less potent. Additionally, carbenoxolone was difficult to wash out and also may be harmful, as the retina became opaque and swollen. MFA, 18-beta-GA, 2-APB and mefloquine also blocked coupling in B-type horizontal cells and AII amacrine cells. Because these cell types express different connexins, this suggests that the antagonists were relatively non-selective across several different types of gap junction. It should be emphasized that MFA was water-soluble and its effects on dye coupling were easily reversible. In contrast, the other gap junction antagonists, except carbenoxolone, required DMSO to make stock solutions and were difficult to wash out of the preparation at the doses required to block coupling in A-type HCs. The combination of potency, water solubility and reversibility suggest that MFA may be a useful compound to manipulate gap junction coupling.
Collapse
Affiliation(s)
- Feng Pan
- Department of Ophthalmology and Visual Science, University of Texas Medical School at Houston, Houston, Texas 77030, USA
| | | | | |
Collapse
|
43
|
Kothmann WW, Li X, Burr GS, O’Brien J. Connexin 35/36 is phosphorylated at regulatory sites in the retina. Vis Neurosci 2007; 24:363-75. [PMID: 17640446 PMCID: PMC2170900 DOI: 10.1017/s095252380707037x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Accepted: 04/11/2007] [Indexed: 11/06/2022]
Abstract
Connexin 35/36 is the most widespread neuronal gap junction protein in the retina and central nervous system. Electrical and/or tracer coupling in a number of neuronal circuits that express this connexin are regulated by light adaptation. In many cases, the regulation of coupling depends on signaling pathways that activate protein kinases such as PKA, and Cx35 has been shown to be regulated by PKA phosphorylation in cell culture systems. To examine whether phosphorylation might regulate Cx35/36 in the retina we developed phospho-specific polyclonal antibodies against the two regulatory phosphorylation sites of Cx35 and examined the phosphorylation state of this connexin in the retina. Western blot analysis with hybrid bass retinal membrane preparations showed Cx35 to be phosphorylated at both the Ser110 and Ser276 sites, and this labeling was eliminated by alkaline phosphatase digestion. The homologous sites of mouse and rabbit Cx36 were also phosphorylated in retinal membrane preparations. Quantitative confocal immunofluorescence analysis showed gap junctions identified with a monoclonal anti-Cx35 antibody to have variable levels of phosphorylation at both the Ser110 and Ser276 sites. Unusual gap junctions that could be identified by their large size (up to 32 microm2) and location in the IPL showed a prominent shift in phosphorylation state from heavily phosphorylated in nighttime, dark-adapted retina to weakly phosphorylated in daytime, light-adapted retina. Both Ser110 and Ser276 sites showed significant changes in this manner. Under both lighting conditions, other gap junctions varied from non-phosphorylated to heavily phosphorylated. We predict that changes in the phosphorylation states of these sites correlate with changes in the degree of coupling through Cx35/36 gap junctions. This leads to the conclusion that connexin phosphorylation mediates changes in coupling in some retinal networks. However, these changes are not global and likely occur in a cell type-specific or possibly a gap junction-specific manner.
Collapse
Affiliation(s)
- W. Wade Kothmann
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston
| | - Xiaofan Li
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
| | - Gary S. Burr
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
| | - John O’Brien
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston
| |
Collapse
|
44
|
Urschel S, Höher T, Schubert T, Alev C, Söhl G, Wörsdörfer P, Asahara T, Dermietzel R, Weiler R, Willecke K. Protein kinase A-mediated phosphorylation of connexin36 in mouse retina results in decreased gap junctional communication between AII amacrine cells. J Biol Chem 2006; 281:33163-71. [PMID: 16956882 DOI: 10.1074/jbc.m606396200] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Gap junctions in AII amacrine cells of mammalian retina participate in the coordination of the rod and cone signaling pathway involved in visual adaptation. Upon stimulation by light, released dopamine binds to D(1) receptors on AII amacrine cells leading to increased intracellular cAMP (cyclic adenosine monophosphate) levels. AII amacrine cells express the gap junctional protein connexin36 (Cx36). Phosphorylation of Cx36 has been hypothesized to regulate gap junctional activity of AII amacrine cells. However, until now in vivo phosphorylation of Cx36 has not been reported. Indeed, it had been concluded that Cx36 in bovine retina is not phosphorylated, but in vitro phosphorylation for Cx35, the bass ortholog of Cx36, had been shown. To clarify this experimental discrepancy, we examined protein kinase A (PKA)-induced phosphorylation of Cx36 in mouse retina as a possible mechanism to modulate the extent of gap junctional coupling. The cytoplasmic domains of Cx36 and the total Cx36 protein were phosphorylated in vitro by PKA. Mass spectroscopy revealed that all four possible PKA consensus motifs were phosphorylated; however, domains point mutated at the sites in question showed a prevalent usage of Ser-110 and Ser-293. Additionally, we demonstrated that Cx36 was phosphorylated in cultured mouse retina. Furthermore, activation of PKA increased the level of phosphorylation of Cx36. cAMP-stimulated, PKA-mediated phosphorylation of Cx36 protein was accompanied by a decrease of tracer coupling between AII amacrine cells. Our results link increased phosphorylation of Cx36 to down-regulation of permeability through gap junction channels mediating light adaptation in the retina.
Collapse
|
45
|
Patel LS, Mitchell CK, Dubinsky WP, O’Brien J. Regulation of gap junction coupling through the neuronal connexin Cx35 by nitric oxide and cGMP. ACTA ACUST UNITED AC 2006; 13:41-54. [PMID: 16613779 PMCID: PMC2189984 DOI: 10.1080/15419060600631474] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Gap-junctional coupling among neurons is subject to regulation by a number of neurotransmitters including nitric oxide. We studied the mechanisms by which NO regulates coupling in cells expressing Cx35, a connexin expressed in neurons throughout the central nervous system. NO donors caused potent uncoupling of HeLa cells stably transfected with Cx35. This effect was mimicked by Bay 21-4272, an activator of guanylyl cyclase. A pharmacological analysis indicated that NO-induced uncoupling involved both PKG-dependent and PKG-independent pathways. PKA was involved in both pathways, suggesting that PKG-dependent uncoupling may be indirect. In vitro, PKG phosphorylated Cx35 at three sites: Ser110, Ser276, and Ser289. A mutational analysis indicated that phosphorylation on Ser110 and Ser276, sites previously shown also to be phosphorylated by PKA, had a significant influence on regulation. Ser289 phosphorylation had very limited effects. We conclude that NO can regulate coupling through Cx35 and that regulation is indirect in HeLa cells.
Collapse
Affiliation(s)
- Leena S. Patel
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
| | - Cheryl K. Mitchell
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
| | - William P. Dubinsky
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston
| | - John O’Brien
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston
- *Corresponding author: John O’Brien, Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, 6431 Fannin St., MSB 7.024, Houston, Texas 77030, Phone: (713) 500-5983, FAX: (713) 500-0682, e-mail:
| |
Collapse
|
46
|
Hoshi H, O'Brien J, Mills SL. A novel fluorescent tracer for visualizing coupled cells in neural circuits of living tissue. J Histochem Cytochem 2006; 54:1169-76. [PMID: 16864895 PMCID: PMC1851887 DOI: 10.1369/jhc.6a6935.2006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Gap junctions have diverse roles in a wide variety of tissues and have recently become a subject of intense investigation in neural circuits where synchrony and oscillations may play an important part. In circuits where gap junctions are present, the possibility arises of identifying intercommunicating cells via introduction of tracer into one cell and observing its spread into its coupled neighbors. Staining the coupled cells by this means opens the door to many vital techniques including paired-cell electrophysiology, RT-PCR, and morphological characterization of previously unknown coupled cells. Tracers commonly used at the present time are not generally suitable for these purposes in many tissues, including neurons. This paper describes how a fluorescent nuclear tracer, Po-pro-1, can be used to visualize coupled cells in several types of retinal neurons thought to be comprised of different connexin proteins including Cx36, Cx45, Cx50, and Cx57.
Collapse
Affiliation(s)
- Hideo Hoshi
- Department of Ophthalmology and Visual Science, University of Texas at Houston-Health Science Center, Houston, Texas
| | - John O'Brien
- Department of Ophthalmology and Visual Science, University of Texas at Houston-Health Science Center, Houston, Texas
| | - Stephen L. Mills
- Department of Ophthalmology and Visual Science, University of Texas at Houston-Health Science Center, Houston, Texas
- Correspondence to: Stephen Mills, Department of Ophthalmology and Visual Science, University of Texas at Houston-Health Science Center, 6431 Fannin, Rm. 7.024, Houston, TX 77030. E-mail:
| |
Collapse
|
47
|
Burr GS, Mitchell CK, Keflemariam YJ, Heidelberger R, O’Brien J. Calcium-dependent binding of calmodulin to neuronal gap junction proteins. Biochem Biophys Res Commun 2005; 335:1191-8. [PMID: 16112650 PMCID: PMC2222552 DOI: 10.1016/j.bbrc.2005.08.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2005] [Accepted: 08/01/2005] [Indexed: 11/29/2022]
Abstract
We examined the interactions of calmodulin with neuronal gap junction proteins connexin35 (Cx35) from perch, its mouse homologue Cx36, and the related perch Cx34.7 using surface plasmon resonance. Calmodulin bound to the C-terminal domains of all three connexins with rapid kinetics in a concentration- and Ca2+-dependent manner. Dissociation was also very rapid. K(d)'s for calmodulin binding at a high-affinity site ranged from 11 to 72 nM, and K(1/2)'s for Ca2+ were between 3 and 5 microM. No binding to the intracellular loops was observed. Binding competition experiments with synthetic peptides mapped the calmodulin binding site to a 10-30 amino acid segment at the beginning of the C-terminal domain of Cx36. The micromolar K(1/2)'s and rapid on and off rates suggest that this interaction may change dynamically in neurons, and may occur transiently when Ca2+ is elevated to a level that would occur in the near vicinity of an activated synapse.
Collapse
Affiliation(s)
- Gary S. Burr
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
| | - Cheryl K. Mitchell
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
| | - Yenabi J. Keflemariam
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
| | - Ruth Heidelberger
- Department of Neurobiology and Anatomy, University of Texas Health Science Center at Houston
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston
| | - John O’Brien
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston
| |
Collapse
|
48
|
Ouyang X, Winbow VM, Patel LS, Burr GS, Mitchell CK, O’Brien J. Protein kinase A mediates regulation of gap junctions containing connexin35 through a complex pathway. ACTA ACUST UNITED AC 2005; 135:1-11. [PMID: 15857663 PMCID: PMC2212611 DOI: 10.1016/j.molbrainres.2004.10.045] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2004] [Revised: 10/14/2004] [Accepted: 10/18/2004] [Indexed: 10/25/2022]
Abstract
Connexin 35 (Cx35) is a major component of electrical synapses in the central nervous system. Many gap junctions containing Cx35 are regulated by dopamine receptor pathways that involve protein kinase A (PKA). To study the mechanism of PKA regulation, we analyzed direct phosphorylation of Cx35 by PKA in vitro and studied the regulation of neurobiotin tracer coupling in HeLa cells expressing Cx35 or Cx35 mutants that lack phosphorylation sites. In Cx35-transfected cells, application of the PKA activator Sp-8-cpt-cAMPS caused a significant decline in coupling, while a PKA inhibitor, Rp-8-cpt-cAMPS, significantly increased tracer coupling. In vitro phosphorylation and mutagenic analysis showed that PKA phosphorylates Cx35 directly at two major sites, Ser110 in the intracellular loop and Ser276 in the carboxyl terminus. In addition, a minor phosphorylation site in the C-terminus was identified by truncation of the last 7 amino acids at Ser298. The mutations Ser110Ala or Ser276Ala significantly reduced regulation of coupling by the PKA activator while a combination of the two eliminated regulation. Truncation at Ser298 reversed the regulation such that the PKA activator significantly increased and the PKA inhibitor significantly decreased coupling. The activation was eliminated in the S110A, S276A, S298ter triple mutant. We conclude that PKA regulates Cx35 coupling in a complex manner that requires both major phosphorylation sites. Furthermore, the tip of the C-terminus acts as a "switch" that determines whether phosphorylation will inhibit or enhance coupling. Reliance on the combined states of three sites provides fine control over the degree of coupling through Cx35 gap junctions.
Collapse
Affiliation(s)
- Xiaosen Ouyang
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston
| | - Virginia M. Winbow
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston
- University of Houston, College of Optometry
| | - Leena S. Patel
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston
| | - Gary S. Burr
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston
| | - Cheryl K. Mitchell
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston
| | - John O’Brien
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston
- *Corresponding author: John O’Brien, Department of Ophthalmology and Visual Science, University of Texas, Houston Health Science Center, 6431 Fannin St., MSB 7.024, Houston, Texas 77030, Phone: (713) 500-5983, FAX: (713) 500-0682, e-mail:
| |
Collapse
|
49
|
Zhang J, Wu SM. Physiological properties of rod photoreceptor electrical coupling in the tiger salamander retina. J Physiol 2005; 564:849-62. [PMID: 15746168 PMCID: PMC1464472 DOI: 10.1113/jphysiol.2005.082859] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/08/2005] [Accepted: 02/25/2005] [Indexed: 11/08/2022] Open
Abstract
Using dual whole-cell voltage and current clamp recording techniques, we investigated the gap junctional conductance and the coupling coefficient between neighbouring rods in live salamander retinal slices. The application of sinusoidal stimuli over a wide range of temporal frequencies allowed us to characterize the band-pass filtering properties of the rod network. We found that the electrical coupling of all neighbouring rods exhibited reciprocal and symmetrical conductivities. On average, the junctional conductance between paired rods was 500 pS and the coupling coefficient (the ratio of voltage responses of the follower cell to those of the driver cell), or K-value, was 0.07. Our experimental results also demonstrated that the rod network behaved like a band-pass filter with a peak frequency of about 2-5 Hz. However, the gap junctions between adjacent rods exhibited linearity and voltage independency within the physiological range of rods. These gap junctions did not contribute to the filtering mechanisms of the rod network. Combined with the computational modelling, our data suggest that the filtering of higher frequency rod signals by the network is largely mediated by the passive resistive and capacitive (RC) properties of rod plasma membranes. Furthermore, we found several attributes of rod electrical coupling resembling the physiological properties of gene-encoded Cx35/36 gap junctions examined in other in vitro studies. This indicates that the previously found Cx35/36 expression in the salamander rod network may be functionally involved in rod-rod electrical coupling.
Collapse
Affiliation(s)
- Jian Zhang
- Cullen Eye Institute, Baylor College of Medicine, One Baylor Plaza, NC-205, Houston, TX 77030, USA.
| | | |
Collapse
|
50
|
Hidaka S, Akahori Y, Kurosawa Y. Dendrodendritic electrical synapses between mammalian retinal ganglion cells. J Neurosci 2004; 24:10553-67. [PMID: 15548670 PMCID: PMC6730298 DOI: 10.1523/jneurosci.3319-04.2004] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2004] [Revised: 09/24/2004] [Accepted: 09/26/2004] [Indexed: 11/21/2022] Open
Abstract
Electrical synapses between alpha-type ganglion cells were detected using combined techniques of dual patch-clamp recordings, intracellular labeling, electron microscopy, and channel subunit connexin immunocytochemistry in the albino rat retina. After intracellular injection of Neurobiotin into alpha-cells of inner (ON-center) and outer (OFF-center) ramifying types, measurement of tracer coupling resulted in a preferentially homologous occurrence among cells of the same morphological type (n = 19 of 24). In high-voltage as well as conventional electron microscopic analysis, direct dendrodendritic gap junctions (average size, 0.86 mum long) were present in contact sites between tracer-coupled alpha-cells. In simultaneous dual whole-cell recordings from pairs of neighboring alpha-cells, these cells generated TTX-sensitive sustained spiking against extrinsic current injection, and bidirectional electrical synapses (maximum coupling coefficient, 0.32) with symmetrical junction conductance (average, 1.35 nS) were observed in pairs with cells of the same morphological type. Precise temporal synchronization of spike activity (average time delay, 2.7 msec) was detected when depolarizing currents were simultaneously injected into the pairs. To address whether physiologically identified electrical synapses constitute gap junctional connectivity between cell pairs, identified neuronal connexin36 immunoreactivity was undertaken in Lucifer yellow-labeled cell pairs after patch-clamp recordings. All alpha-cells expressed connexin36, and confocal laser-scanning imaging demonstrated that connexin36 is primarily located at dendritic crossings between electrically coupled cells (seven sites in a pair, on average). These results give conclusive evidence for electrical synapses via dendrodendritic gap junctions involving connexin36 in alpha retinal ganglion cells of the same physiological type.
Collapse
Affiliation(s)
- Soh Hidaka
- Department of Physiology, School of Medicine, Institute for Comprehensive Medical Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | | | | |
Collapse
|