1
|
Majeed M, Han H, Zhang K, Cao WX, Liao CP, Hobert O, Lu H. Toolkits for detailed and high-throughput interrogation of synapses in C. elegans. eLife 2024; 12:RP91775. [PMID: 38224479 PMCID: PMC10945580 DOI: 10.7554/elife.91775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
Visualizing synaptic connectivity has traditionally relied on time-consuming electron microscopy-based imaging approaches. To scale the analysis of synaptic connectivity, fluorescent protein-based techniques have been established, ranging from the labeling of specific pre- or post-synaptic components of chemical or electrical synapses to transsynaptic proximity labeling technology such as GRASP and iBLINC. In this paper, we describe WormPsyQi, a generalizable image analysis pipeline that automatically quantifies synaptically localized fluorescent signals in a high-throughput and robust manner, with reduced human bias. We also present a resource of 30 transgenic strains that label chemical or electrical synapses throughout the nervous system of the nematode Caenorhabditis elegans, using CLA-1, RAB-3, GRASP (chemical synapses), or innexin (electrical synapse) reporters. We show that WormPsyQi captures synaptic structures in spite of substantial heterogeneity in neurite morphology, fluorescence signal, and imaging parameters. We use these toolkits to quantify multiple obvious and subtle features of synapses - such as number, size, intensity, and spatial distribution of synapses - in datasets spanning various regions of the nervous system, developmental stages, and sexes. Although the pipeline is described in the context of synapses, it may be utilized for other 'punctate' signals, such as fluorescently tagged neurotransmitter receptors and cell adhesion molecules, as well as proteins in other subcellular contexts. By overcoming constraints on time, sample size, cell morphology, and phenotypic space, this work represents a powerful resource for further analysis of synapse biology in C. elegans.
Collapse
Affiliation(s)
- Maryam Majeed
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Haejun Han
- School of Electrical and Computer Engineering, Georgia Institute of TechnologyAtlantaUnited States
- The Parker H Petit Institute of Bioengineering and Bioscience, Georgia Institute of TechnologyAtlantaUnited States
| | - Keren Zhang
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaUnited States
| | - Wen Xi Cao
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Chien-Po Liao
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia UniversityNew YorkUnited States
| | - Hang Lu
- The Parker H Petit Institute of Bioengineering and Bioscience, Georgia Institute of TechnologyAtlantaUnited States
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaUnited States
| |
Collapse
|
2
|
Ribelayga CP, O’Brien J. When microscopy and electrophysiology meet connectomics-Steve Massey's contribution to unraveling the structure and function of the rod/cone gap junction. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1305131. [PMID: 38983007 PMCID: PMC11182179 DOI: 10.3389/fopht.2023.1305131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 10/31/2023] [Indexed: 07/11/2024]
Abstract
Electrical synapses, formed of gap junctions, are ubiquitous components of the central nervous system (CNS) that shape neuronal circuit connectivity and dynamics. In the retina, electrical synapses can create a circuit, control the signal-to-noise ratio in individual neurons, and support the coordinated neuronal firing of ganglion cells, hence, regulating signal processing at the network, single-cell, and dendritic level. We, the authors, and Steve Massey have had a long interest in gap junctions in retinal circuits, in general, and in the network of photoreceptors, in particular. Our combined efforts, based on a wide array of techniques of molecular biology, microscopy, and electrophysiology, have provided fundamental insights into the molecular structure and properties of the rod/cone gap junction. Yet, a full understanding of how rod/cone coupling controls circuit dynamics necessitates knowing its operating range. It is well established that rod/cone coupling can be greatly reduced or eliminated by bright-light adaptation or pharmacological treatment; however, the upper end of its dynamic range has long remained elusive. This held true until Steve Massey's recent interest for connectomics led to the development of a new strategy to assess this issue. The effort proved effective in establishing, with precision, the connectivity rules between rods and cones and estimating the theoretical upper limit of rod/cone electrical coupling. Comparing electrophysiological measurements and morphological data indicates that under pharmacological manipulation, rod/cone coupling can reach the theoretical maximum of its operating range, implying that, under these conditions, all the gap junction channels present at the junctions are open. As such, channel open probability is likely the main determinant of rod/cone coupling that can change momentarily in a time-of-day- and light-dependent manner. In this article we briefly review our current knowledge of the molecular structure of the rod/cone gap junction and of the mechanisms behind its modulation, and we highlight the recent work led by Steve Massey. Steve's contribution has been critical toward asserting the modulation depth of rod/cone coupling as well as elevating the rod/cone gap junction as one of the most suitable models to examine the role of electrical synapses and their plasticity in neural processing.
Collapse
Affiliation(s)
- Christophe P. Ribelayga
- Department of Vision Sciences, University of Houston College of Optometry, Houston, TX, United States
| | | |
Collapse
|
3
|
Lee EJ, Diaz-Aguilar MS, Min H, Choi J, Valdez Duran DA, Grandjean JM, Wiseman RL, Kroeger H, Lin JH. Mitochondria and Endoplasmic Reticulum Stress in Retinal Organoids from Patients with Vision Loss. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1721-1739. [PMID: 36535406 PMCID: PMC10616714 DOI: 10.1016/j.ajpath.2022.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
Abstract
Activating transcription factor 6 (ATF6), a key regulator of the unfolded protein response, plays a key role in endoplasmic reticulum function and protein homeostasis. Variants of ATF6 that abrogate transcriptional activity cause morphologic and molecular defects in cones, clinically manifesting as the human vision loss disease achromatopsia (ACHM). ATF6 is expressed in all retinal cells. However, the effect of disease-associated ATF6 variants on other retinal cell types remains unclear. Herein, this was investigated by analyzing bulk RNA-sequencing transcriptomes from retinal organoids generated from patients with ACHM, carrying homozygous loss-of-function ATF6 variants. Marked dysregulation in mitochondrial respiratory complex gene expression and disrupted mitochondrial morphology in ACHM retinal organoids were identified. This indicated that loss of ATF6 leads to previously unappreciated mitochondrial defects in the retina. Next, gene expression from control and ACHM retinal organoids were compared with transcriptome profiles of seven major retinal cell types generated from recent single-cell transcriptomic maps of nondiseased human retina. This indicated pronounced down-regulation of cone genes and up-regulation in Müller glia genes, with no significant effects on other retinal cells. Overall, the current analysis of ACHM patient retinal organoids identified new cellular and molecular phenotypes in addition to cone dysfunction: activation of Müller cells, increased endoplasmic reticulum stress, disrupted mitochondrial structure, and elevated respiratory chain activity gene expression.
Collapse
Affiliation(s)
- Eun-Jin Lee
- Department of Ophthalmology, Stanford University, Stanford, California; Department of Pathology, VA Palo Alto Healthcare System, Palo Alto, California; Department of Pathology, Stanford University, Stanford, California
| | - Monica S Diaz-Aguilar
- Department of Ophthalmology, Stanford University, Stanford, California; Department of Pathology, VA Palo Alto Healthcare System, Palo Alto, California; Department of Pathology, Stanford University, Stanford, California; Department of Medicine, Rush University Medical College, Chicago, Illinois
| | - Hyejung Min
- Department of Pathology, VA Palo Alto Healthcare System, Palo Alto, California; Department of Pathology, Stanford University, Stanford, California
| | - Jihee Choi
- Department of Pathology, Stanford University, Stanford, California
| | | | - Julia M Grandjean
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Heike Kroeger
- Department of Cellular Biology, University of Georgia, Athens, Georgia
| | - Jonathan H Lin
- Department of Ophthalmology, Stanford University, Stanford, California; Department of Pathology, VA Palo Alto Healthcare System, Palo Alto, California; Department of Pathology, Stanford University, Stanford, California.
| |
Collapse
|
4
|
Kovács-Öller T, Szarka G, Hoffmann G, Péntek L, Valentin G, Ross L, Völgyi B. Extrinsic and Intrinsic Factors Determine Expression Levels of Gap Junction-Forming Connexins in the Mammalian Retina. Biomolecules 2023; 13:1119. [PMID: 37509155 PMCID: PMC10377540 DOI: 10.3390/biom13071119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Gap junctions (GJs) are not static bridges; instead, GJs as well as the molecular building block connexin (Cx) proteins undergo major expression changes in the degenerating retinal tissue. Various progressive diseases, including retinitis pigmentosa, glaucoma, age-related retinal degeneration, etc., affect neurons of the retina and thus their neuronal connections endure irreversible changes as well. Although Cx expression changes might be the hallmarks of tissue deterioration, GJs are not static bridges and as such they undergo adaptive changes even in healthy tissue to respond to the ever-changing environment. It is, therefore, imperative to determine these latter adaptive changes in GJ functionality as well as in their morphology and Cx makeup to identify and distinguish them from alterations following tissue deterioration. In this review, we summarize GJ alterations that take place in healthy retinal tissue and occur on three different time scales: throughout the entire lifespan, during daily changes and as a result of quick changes of light adaptation.
Collapse
Affiliation(s)
- Tamás Kovács-Öller
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
- NEURON-066 Rethealthsi Research Group, 7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Gergely Szarka
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
- NEURON-066 Rethealthsi Research Group, 7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Gyula Hoffmann
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
- NEURON-066 Rethealthsi Research Group, 7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Loretta Péntek
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
| | - Gréta Valentin
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
| | - Liliana Ross
- Faculty of Science, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary
- Department of Neurobiology, University of Pécs, 7624 Pécs, Hungary
- NEURON-066 Rethealthsi Research Group, 7624 Pécs, Hungary
- Center for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
5
|
Myles WE, McFadden SA. Analytical methods for assessing retinal cell coupling using cut-loading. PLoS One 2022; 17:e0271744. [PMID: 35853039 PMCID: PMC9295955 DOI: 10.1371/journal.pone.0271744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/06/2022] [Indexed: 11/25/2022] Open
Abstract
Electrical coupling between retinal neurons contributes to the functional complexity of visual circuits. “Cut-loading” methods allow simultaneous assessment of cell-coupling between multiple retinal cell-types, but existing analysis methods impede direct comparison with gold standard direct dye injection techniques. In the current study, we both improved an existing method and developed two new approaches to address observed limitations. Each method of analysis was applied to cut-loaded dark-adapted Guinea pig retinae (n = 29) to assess coupling strength in the axonless horizontal cell type (‘a-type’, aHCs). Method 1 was an improved version of the standard protocol and described the distance of dye-diffusion (space constant). Method 2 adjusted for the geometric path of dye-transfer through cut-loaded cells and extracted the rate of dye-transfer across gap-junctions in terms of the coupling coefficient (kj). Method 3 measured the diffusion coefficient (De) perpendicular to the cut-axis. Dye transfer was measured after one of five diffusion times (1–20 mins), or with a coupling inhibitor, meclofenamic acid (MFA) (50–500μM after 20 mins diffusion). The standard protocol fits an exponential decay function to the fluorescence profile of a specified retina layer but includes non-specific background fluorescence. This was improved by measuring the fluorescence of individual cell soma and excluding from the fit non-horizontal cells located at the cut-edge (p<0.001) (Method 1). The space constant (Method 1) increased with diffusion time (p<0.01), whereas Methods 2 (p = 0.54) and 3 (p = 0.63) produced consistent results across all diffusion times. Adjusting distance by the mean cell-cell spacing within each tissue reduced the incidence of outliers across all three methods. Method 1 was less sensitive to detecting changes induced by MFA than Methods 2 (p<0.01) and 3 (p<0.01). Although the standard protocol was easily improved (Method 1), Methods 2 and 3 proved more sensitive and generalisable; allowing for detailed assessment of the tracer kinetics between different populations of gap-junction linked cell networks and direct comparison to dye-injection techniques.
Collapse
Affiliation(s)
- William E. Myles
- College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
- * E-mail:
| | - Sally A. McFadden
- College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
6
|
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
|
7
|
Fusz K, Kovács-Öller T, Kóbor P, Szabó-Meleg E, Völgyi B, Buzás P, Telkes I. Regional Variation of Gap Junctional Connections in the Mammalian Inner Retina. Cells 2021; 10:2396. [PMID: 34572046 PMCID: PMC8466939 DOI: 10.3390/cells10092396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 11/24/2022] Open
Abstract
The retinas of many species show regional specialisations that are evident in the differences in the processing of visual input from different parts of the visual field. Regional specialisation is thought to reflect an adaptation to the natural visual environment, optical constraints, and lifestyle of the species. Yet, little is known about regional differences in synaptic circuitry. Here, we were interested in the topographical distribution of connexin-36 (Cx36), the major constituent of electrical synapses in the retina. We compared the retinas of mice, rats, and cats to include species with different patterns of regional specialisations in the analysis. First, we used the density of Prox1-immunoreactive amacrine cells as a marker of any regional specialisation, with higher cell density signifying more central regions. Double-labelling experiments showed that Prox1 is expressed in AII amacrine cells in all three species. Interestingly, large Cx36 plaques were attached to about 8-10% of Prox1-positive amacrine cell somata, suggesting the strong electrical coupling of pairs or small clusters of cell bodies. When analysing the regional changes in the volumetric density of Cx36-immunoreactive plaques, we found a tight correlation with the density of Prox1-expressing amacrine cells in the ON, but not in the OFF sublamina in all three species. The results suggest that the relative contribution of electrical synapses to the ON- and OFF-pathways of the retina changes with retinal location, which may contribute to functional ON/OFF asymmetries across the visual field.
Collapse
Affiliation(s)
- Katalin Fusz
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Kovács-Öller
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE NAP-2 Retinal Electrical Synapses Research Group, 7624 Pécs, Hungary
| | - Péter Kóbor
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Edina Szabó-Meleg
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Institute of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Béla Völgyi
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE NAP-2 Retinal Electrical Synapses Research Group, 7624 Pécs, Hungary
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary
| | - Péter Buzás
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| | - Ildikó Telkes
- Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary; (K.F.); (P.K.); (I.T.)
- Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary; (T.K.-Ö.); (E.S.-M.); (B.V.)
- Centre for Neuroscience, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
8
|
Zhi Z, Xiang J, Fu Q, Pei X, Zhou D, Cao Y, Xie L, Zhang S, Chen S, Qu J, Zhou X. The Role of Retinal Connexins Cx36 and Horizontal Cell Coupling in Emmetropization in Guinea Pigs. Invest Ophthalmol Vis Sci 2021; 62:27. [PMID: 34283211 PMCID: PMC8300059 DOI: 10.1167/iovs.62.9.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 06/24/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to determine whether retinal gap junctions (GJs) via connexin 36 (Cx36, mediating coupling of many retinal cell types) and horizontal cell (HC-HC) coupling, are involved in emmetropization. Methods Guinea pigs (3 weeks old) were monocularly form deprived (FD) or raised without FD (in normal visual [NV] environment) for 2 days or 4 weeks; alternatively, they wore a -4 D lens (hyperopic defocus [HD]) or 0 D lens for 2 days or 1 week. FD and NV eyes received daily subconjunctival injections of a nonspecific GJ-uncoupling agent, 18-β-Glycyrrhetinic Acid (18-β-GA). The amounts of total Cx36 and of phosphorylated Cx36 (P-Cx36; activated state that increases cell-cell coupling), in the inner and outer plexiform layers (IPLs and OPLs), were evaluated by quantitative immunofluorescence (IF), and HC-HC coupling was evaluated by cut-loading with neurobiotin. Results FD per se (excluding effect of light-attenuation) increased HC-HC coupling in OPL, whereas HD did not affect it. HD for 2 days or 1 week had no significant effect on retinal content of Cx36 or P-Cx36. FD for 4 weeks decreased the total amounts of Cx36 and P-Cx36, and the P-Cx36/Cx36 ratio, in the IPL. Subconjunctival 18-β-GA induced myopia in NV eyes and increased the myopic shifts in FD eyes, while reducing the amounts of Cx36 and P-Cx36 in both the IPL and OPL. Conclusions These results suggest that cell-cell coupling via GJs containing Cx36 (particularly those in the IPL) plays a role in emmetropization and form deprivation myopia (FDM) in mammals. Although both FD and 18-β-GA induced myopia, they had opposite effects on HC-HC coupling. These findings suggest that HC-HC coupling in the OPL might not play a significant role in emmetropization and myopia development.
Collapse
Affiliation(s)
- Zhina Zhi
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Jing Xiang
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Qian Fu
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Xiaomeng Pei
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Dengke Zhou
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Yuqing Cao
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Liqin Xie
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Sen Zhang
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Si Chen
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Jia Qu
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| | - Xiangtian Zhou
- School of Optometry and Ophthalmology, and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health People's Republic of China and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang, China
| |
Collapse
|
9
|
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
|
10
|
Cangiano L, Asteriti S. Interphotoreceptor coupling: an evolutionary perspective. Pflugers Arch 2021; 473:1539-1554. [PMID: 33988778 PMCID: PMC8370920 DOI: 10.1007/s00424-021-02572-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/13/2021] [Accepted: 04/23/2021] [Indexed: 12/16/2022]
Abstract
In the vertebrate retina, signals generated by cones of different spectral preference and by highly sensitive rod photoreceptors interact at various levels to extract salient visual information. The first opportunity for such interaction is offered by electrical coupling of the photoreceptors themselves, which is mediated by gap junctions located at the contact points of specialised cellular processes: synaptic terminals, telodendria and radial fins. Here, we examine the evolutionary pressures for and against interphotoreceptor coupling, which are likely to have shaped how coupling is deployed in different species. The impact of coupling on signal to noise ratio, spatial acuity, contrast sensitivity, absolute and increment threshold, retinal signal flow and colour discrimination is discussed while emphasising available data from a variety of vertebrate models spanning from lampreys to primates. We highlight the many gaps in our knowledge, persisting discrepancies in the literature, as well as some major unanswered questions on the actual extent and physiological role of cone-cone, rod-cone and rod-rod communication. Lastly, we point toward limited but intriguing evidence suggestive of the ancestral form of coupling among ciliary photoreceptors.
Collapse
Affiliation(s)
- Lorenzo Cangiano
- Dept. of Translational Research, University of Pisa, Via San Zeno 31, 56123, Pisa, Italy.
| | - Sabrina Asteriti
- Dept. of Translational Research, University of Pisa, Via San Zeno 31, 56123, Pisa, Italy
| |
Collapse
|
11
|
Burger CA, Jiang D, Mackin RD, Samuel MA. Development and maintenance of vision's first synapse. Dev Biol 2021; 476:218-239. [PMID: 33848537 DOI: 10.1016/j.ydbio.2021.04.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 12/21/2022]
Abstract
Synapses in the outer retina are the first information relay points in vision. Here, photoreceptors form synapses onto two types of interneurons, bipolar cells and horizontal cells. Because outer retina synapses are particularly large and highly ordered, they have been a useful system for the discovery of mechanisms underlying synapse specificity and maintenance. Understanding these processes is critical to efforts aimed at restoring visual function through repairing or replacing neurons and promoting their connectivity. We review outer retina neuron synapse architecture, neural migration modes, and the cellular and molecular pathways that play key roles in the development and maintenance of these connections. We further discuss how these mechanisms may impact connectivity in the retina.
Collapse
Affiliation(s)
- Courtney A Burger
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Danye Jiang
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Robert D Mackin
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Melanie A Samuel
- Huffington Center on Aging, Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
12
|
Function of cone and cone-related pathways in Ca V1.4 IT mice. Sci Rep 2021; 11:2732. [PMID: 33526839 PMCID: PMC7851161 DOI: 10.1038/s41598-021-82210-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 01/11/2021] [Indexed: 11/09/2022] Open
Abstract
CaV1.4 L-type calcium channels are predominantly expressed in photoreceptor terminals playing a crucial role for synaptic transmission and, consequently, for vision. Human mutations in the encoding gene are associated with congenital stationary night blindness type-2. Besides rod-driven scotopic vision also cone-driven photopic responses are severely affected in patients. The present study therefore examined functional and morphological changes in cones and cone-related pathways in mice carrying the CaV1.4 gain-of function mutation I756T (CaV1.4-IT) using multielectrode array, patch-clamp and immunohistochemical analyses. CaV1.4-IT ganglion cell responses to photopic stimuli were seen only in a small fraction of cells indicative of a major impairment in the cone pathway. Though cone photoreceptors underwent morphological rearrangements, they retained their ability to release glutamate. Our functional data suggested a postsynaptic cone bipolar cell defect, supported by the fact that the majority of cone bipolar cells showed sprouting, while horizontal cells maintained contacts with cones and cone-to-horizontal cell input was preserved. Furthermore a reduction of basal Ca2+ influx by a calcium channel blocker was not sufficient to rescue synaptic transmission deficits caused by the CaV1.4-IT mutation. Long term treatments with low-dose Ca2+ channel blockers might however be beneficial reducing Ca2+ toxicity without major effects on ganglion cells responses.
Collapse
|
13
|
Jin N, Zhang Z, Keung J, Youn SB, Ishibashi M, Tian LM, Marshak DW, Solessio E, Umino Y, Fahrenfort I, Kiyama T, Mao CA, You Y, Wei H, Wu J, Postma F, Paul DL, Massey SC, Ribelayga CP. Molecular and functional architecture of the mouse photoreceptor network. SCIENCE ADVANCES 2020; 6:eaba7232. [PMID: 32832605 PMCID: PMC7439306 DOI: 10.1126/sciadv.aba7232] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 04/21/2020] [Indexed: 06/11/2023]
Abstract
Mouse photoreceptors are electrically coupled via gap junctions, but the relative importance of rod/rod, cone/cone, or rod/cone coupling is unknown. Furthermore, while connexin36 (Cx36) is expressed by cones, the identity of the rod connexin has been controversial. We report that FACS-sorted rods and cones both express Cx36 but no other connexins. We created rod- and cone-specific Cx36 knockout mice to dissect the photoreceptor network. In the wild type, Cx36 plaques at rod/cone contacts accounted for more than 95% of photoreceptor labeling and paired recordings showed the transjunctional conductance between rods and cones was ~300 pS. When Cx36 was eliminated on one side of the gap junction, in either conditional knockout, Cx36 labeling and rod/cone coupling were almost abolished. We could not detect direct rod/rod coupling, and cone/cone coupling was minor. Rod/cone coupling is so prevalent that indirect rod/cone/rod coupling via the network may account for previous reports of rod coupling.
Collapse
Affiliation(s)
- Nange Jin
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhijing Zhang
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Joyce Keung
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sean B. Youn
- Summer Research Program, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Undergraduate Program, William Marsh Rice University, Houston, TX, USA
| | - Munenori Ishibashi
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lian-Ming Tian
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - David W. Marshak
- Department of Neurobiology and Anatomy, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Neuroscience Research Center, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eduardo Solessio
- Center for Vision Research and SUNY Eye Institute, Department of Ophthalmology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yumiko Umino
- Center for Vision Research and SUNY Eye Institute, Department of Ophthalmology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Iris Fahrenfort
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Takae Kiyama
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chai-An Mao
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Neuroscience Research Center, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yanan You
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, The University of Texas Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Haichao Wei
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, The University of Texas Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Jiaqian Wu
- Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, The University of Texas Brown Foundation Institute of Molecular Medicine, Houston, TX, USA
| | - Friso Postma
- Department of Neurobiology, Medical School, Harvard University, Boston, MA, USA
| | - David L. Paul
- Department of Neurobiology, Medical School, Harvard University, Boston, MA, USA
| | - Stephen C. Massey
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Summer Research Program, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Neuroscience Research Center, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Elizabeth Morford Distinguished Chair in Ophthalmology and Research Director, Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, 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
- Summer Research Program, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Neuroscience Research Center, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Neuroscience, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Program in Biochemistry and Cellular Biology, Graduate School of Biomedical Sciences, MD Anderson Cancer Center/UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
- Bernice Weingarten Chair in Ophthalmology, Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
14
|
Light adaptation in the chick retina: Dopamine, nitric oxide, and gap-junction coupling modulate spatiotemporal contrast sensitivity. Exp Eye Res 2020; 195:108026. [PMID: 32246982 DOI: 10.1016/j.exer.2020.108026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 11/20/2022]
Abstract
Adaptation to changes in ambient light intensity, in retinal cells and circuits, optimizes visual functions. In the retina, light-adaptation results in changes in light-sensitivity and spatiotemporal tuning of ganglion cells. Under light-adapted conditions, contrast sensitivity (CS) of ganglion cells is a bandpass function of spatial frequency; in contrast, dark-adaptation reduces CS, especially at higher spatial frequencies. In this work, we aimed to understand intrinsic neuromodulatory mechanisms that underlie retinal adaptation to changes in ambient light level. Specifically, we investigated how CS is affected by dopamine (DA), nitric oxide (NO), and modifiers of electrical coupling through gap junctions, under different conditions of adapting illumination. Using the optokinetic response as a behavioral readout of direction-selective ganglion cell activity, we characterized the spatial CS of chicks under high- and low-photopic conditions and how it was regulated by DA, NO, and gap-junction uncouplers. We observed that: (1) DA D2R-family agonists and a donor of NO increased CS tested in low-photopic illumination, as if observed in the high-photopic light; whereas (2) removing their effects using either DA antagonists or NO- synthase inhibitors mimicked low-photopic CS; (3) simulation of high-photopic CS by DA agonists was abolished by NO-synthase inhibitors; and (4) selectively blocking coupling via connexin 35/36-containing gap junctions, using a "designer" mimetic peptide, increased CS, as does strong illumination. We conclude that, in the chicken retina: (1) DA and NO induce changes in spatiotemporal processing, similar to those driven by increasing illumination, (2) DA possibly acts through stimulating NO synthesis, and (3) blockade of coupling via gap junctions containing connexin 35/36 also drives a change in retinal CS functions. As a noninvasive method, the optokinetic response can provide rapid, conditional, and reversible assessment of retinal functions when pharmacological reagents are injected into the vitreous humor. Finally, the chick's large eyes, and the many similarities between their adaptational circuit functions and those in mammals such as the mouse, make them a promising model for future retinal research.
Collapse
|
15
|
Sánchez OF, Rodríguez AV, Velasco-España JM, Murillo LC, Sutachan JJ, Albarracin SL. Role of Connexins 30, 36, and 43 in Brain Tumors, Neurodegenerative Diseases, and Neuroprotection. Cells 2020; 9:E846. [PMID: 32244528 PMCID: PMC7226843 DOI: 10.3390/cells9040846] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/15/2020] [Accepted: 02/24/2020] [Indexed: 02/07/2023] Open
Abstract
Gap junction (GJ) channels and their connexins (Cxs) are complex proteins that have essential functions in cell communication processes in the central nervous system (CNS). Neurons, astrocytes, oligodendrocytes, and microglial cells express an extraordinary repertory of Cxs that are important for cell to cell communication and diffusion of metabolites, ions, neurotransmitters, and gliotransmitters. GJs and Cxs not only contribute to the normal function of the CNS but also the pathological progress of several diseases, such as cancer and neurodegenerative diseases. Besides, they have important roles in mediating neuroprotection by internal or external molecules. However, regulation of Cx expression by epigenetic mechanisms has not been fully elucidated. In this review, we provide an overview of the known mechanisms that regulate the expression of the most abundant Cxs in the central nervous system, Cx30, Cx36, and Cx43, and their role in brain cancer, CNS disorders, and neuroprotection. Initially, we focus on describing the Cx gene structure and how this is regulated by epigenetic mechanisms. Then, the posttranslational modifications that mediate the activity and stability of Cxs are reviewed. Finally, the role of GJs and Cxs in glioblastoma, Alzheimer's, Parkinson's, and Huntington's diseases, and neuroprotection are analyzed with the aim of shedding light in the possibility of using Cx regulators as potential therapeutic molecules.
Collapse
Affiliation(s)
- Oscar F. Sánchez
- Department of Nutrition and Biochemistry, Pontificia Universidad Javeriana, 110911 Bogota, Colombia; (A.V.R.); (J.M.V.-E.); (L.C.M.); (J.-J.S.)
| | | | | | | | | | - Sonia-Luz Albarracin
- Department of Nutrition and Biochemistry, Pontificia Universidad Javeriana, 110911 Bogota, Colombia; (A.V.R.); (J.M.V.-E.); (L.C.M.); (J.-J.S.)
| |
Collapse
|
16
|
Yang GY, Liu FY, Li X, Zhu QR, Chen BJ, Liu LQ. Decreased expression of gap junction delta-2 (GJD2) messenger RNA and connexin 36 protein in form-deprivation myopia of guinea pigs. Chin Med J (Engl) 2019; 132:1700-1705. [PMID: 31283648 PMCID: PMC6759107 DOI: 10.1097/cm9.0000000000000319] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND More than ten genome-wide association studies have identified the significant association between the gap junction delta-2 (GJD2) gene and myopia. However, no functional studies have been performed to confirm that this gene is correlated with myopia. This study aimed to observe how this gene changed in mRNA and protein level in the form-deprivation myopia (FDM) animal model. METHODS Four-week-old guinea pigs were randomly divided into two groups: control and FDM groups (n = 12 for each group). The right eyes of the FDM group were covered with opaque hemispherical plastic lenses for 3 weeks. For all the animals, refractive status, axial length (AL), and corneal radius of curvature were measured at baseline and 3 weeks later by streak retinoscope, A-scan ultrasonography, and keratometer, respectively. Retinal GJD2 mRNA expression and connexin 36 (Cx36) levels in FDM and control groups were measured by quantitative real-time PCR and Western blot analyses, respectively. Those results were compared using independent t test, Mann-Whitney U test, or paired t test. A significance level of P < 0.05 was used. RESULTS Three weeks later, the FDM group (form-deprived eyes) showed about a myopic shift of approximately -6.75 (-7.94 to -6.31) D, while the control group remained hyperopic with only a shift of -0.50 (-0.75 to 0.25) D (Z = -3.38, P < 0.01). The AL increased by 0.74 (0.61-0.76) and 0.10 (0.05-0.21) mm in FDM and control groups, respectively (Z = -3.37, P < 0.01). The relative mRNA expression of GJD2 in the FDM group decreased 31.58% more than the control group (t = 11.44, P < 0.01). The relative protein expression of CX36 on the retina was lowered by 37.72% in form-deprivation eyes as compared to the controls (t = 17.74, P < 0.01). CONCLUSION Both the mRNA expression of GJD2 and Cx36 protein amount were significantly decreased in the retina of FDM guinea pigs. This indicates that Cx36 is involved in FDM development, providing compensating evidence for the results obtained from genome-wide association studies.
Collapse
Affiliation(s)
- Guo-Yuan Yang
- Department of Opthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Feng-Yang Liu
- Department of Opthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Ophthalmology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xia Li
- Department of Opthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiu-Rong Zhu
- Department of Opthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bing-Jie Chen
- Department of Opthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Long-Qian Liu
- Department of Opthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
17
|
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
|
18
|
Kántor O, Szarka G, Benkő Z, Somogyvári Z, Pálfi E, Baksa G, Rácz G, Nitschke R, Debertin G, Völgyi B. Strategic Positioning of Connexin36 Gap Junctions Across Human Retinal Ganglion Cell Dendritic Arbors. Front Cell Neurosci 2018; 12:409. [PMID: 30524239 PMCID: PMC6262005 DOI: 10.3389/fncel.2018.00409] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/22/2018] [Indexed: 11/18/2022] Open
Abstract
Connexin36 (Cx36) subunits form gap junctions (GJ) between neurons throughout the central nervous system. Such GJs of the mammalian retina serve the transmission, averaging and correlation of signals prior to conveying visual information to the brain. Retinal GJs have been exhaustively studied in various animal species, however, there is still a perplexing paucity of information regarding the presence and function of human retinal GJs. Particularly little is known about GJ formation of human retinal ganglion cells (hRGCs) due to the limited number of suitable experimental approaches. Compared to the neuronal coupling studies in animal models, where GJ permeable tracer injection is the gold standard method, the post-mortem nature of scarcely available human retinal samples leaves immunohistochemistry as a sole approach to obtain information on hRGC GJs. In this study Lucifer Yellow (LY) dye injections and Cx36 immunohistochemistry were performed in fixed short-post-mortem samples to stain hRGCs with complete dendritic arbors and locate dendritic Cx36 GJs. Subsequent neuronal reconstructions and morphometric analyses revealed that Cx36 plaques had a clear tendency to form clusters and particularly favored terminal dendritic segments.
Collapse
Affiliation(s)
- Orsolya Kántor
- Department of Neuroanatomy, Faculty of Medicine, Institute for Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary.,Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Gergely Szarka
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary.,Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary.,Center for Neuroscience, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Zsigmond Benkő
- Complex Systems and Computational Neuroscience Group, Wigner Research Center for Physics, Hungarian Academy of Sciences, Budapest, Hungary
| | - Zoltán Somogyvári
- Complex Systems and Computational Neuroscience Group, Wigner Research Center for Physics, Hungarian Academy of Sciences, Budapest, Hungary
| | - Emese Pálfi
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Gábor Baksa
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Gergely Rácz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Roland Nitschke
- Life Imaging Center, Center for Biological Systems Analysis, Albert-Ludwigs University, Freiburg, Germany.,BIOSS Center for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Gábor Debertin
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary.,Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary.,Center for Neuroscience, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Béla Völgyi
- MTA-PTE NAP 2 Retinal Electrical Synapses Research Group, Pécs, Hungary.,Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary.,Center for Neuroscience, University of Pécs, Pécs, Hungary.,János Szentágothai Research Center, University of Pécs, Pécs, Hungary
| |
Collapse
|
19
|
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
|
20
|
Nagy JI, Lynn BD, Senecal JMM, Stecina K. Connexin36 Expression in Primary Afferent Neurons in Relation to the Axon Reflex and Modality Coding of Somatic Sensation. Neuroscience 2018; 383:216-234. [PMID: 29746988 DOI: 10.1016/j.neuroscience.2018.04.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/02/2018] [Accepted: 04/26/2018] [Indexed: 01/25/2023]
Abstract
Electrical coupling mediated by connexin36-containing gap junctions that form electrical synapses is known to be prevalent in the central nervous system, but such coupling was long ago reported also to occur between cutaneous sensory fibers. Here, we provide evidence supporting the capability of primary afferent fibers to engage in electrical coupling. In transgenic mice with enhanced green fluorescent protein (eGFP) serving as a reporter for connexin36 expression, immunofluorescence labeling of eGFP was found in subpopulations of neurons in lumbar dorsal root and trigeminal sensory ganglia, and in fibers within peripheral nerves and tissues. Immunolabeling of connexin36 was robust in the sciatic nerve, weaker in sensory ganglia than in peripheral nerve, and absent in these tissues from Cx36 null mice. Connexin36 mRNA was detected in ganglia from wild-type mice, but not in those from Cx36 null mice. Labeling of eGFP was localized within a subpopulation of ganglion cells containing substance P and calcitonin gene-releasing peptide, and in peripheral fibers containing these peptides. Expression of eGFP was also found in various proportions of sensory ganglion neurons containing transient receptor potential (TRP) channels, including TRPV1 and TRPM8. Ganglion cells labeled for isolectin B4 and tyrosine hydroxylase displayed very little co-localization with eGFP. Our results suggest that previously observed electrical coupling between peripheral sensory fibers occurs via electrical synapses formed by Cx36-containing gap junctions, and that some degree of selectivity in the extent of electrical coupling may occur between fibers belonging to subpopulations of sensory neurons identified according to their sensory modality responsiveness.
Collapse
Affiliation(s)
- J I Nagy
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada.
| | - B D Lynn
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - J M M Senecal
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - K Stecina
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
21
|
Abstract
We have long known that rod and cone signals interact within the retina and can even contribute to color vision, but the extent of these influences has remained unclear. New results with more powerful methods of RNA expression profiling, specific cell labeling, and single-cell recording have provided greater clarity and are showing that rod and cone signals can mix at virtually every level of signal processing. These interactions influence the integration of retinal signals and make an important contribution to visual perception.
Collapse
Affiliation(s)
- Gordon Fain
- Department of Ophthalmology, Jules Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles, 100 Stein Plaza, Los Angeles, CA 90095-7000, USA.,Department of Integrative Biology and Physiology, University of California Los Angeles, Terasaki Life Sciences, 610 Charles E. Young Drive South, Los Angeles, CA 90095-7239, USA
| | - Alapakkam P Sampath
- Department of Ophthalmology, Jules Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles, 100 Stein Plaza, Los Angeles, CA 90095-7000, USA
| |
Collapse
|
22
|
Ma Y, Han X, de Castro RB, Zhang P, Zhang K, Hu Z, Qin L. Analysis of the bystander effect in cone photoreceptors via a guided neural network platform. SCIENCE ADVANCES 2018; 4:eaas9274. [PMID: 29750200 PMCID: PMC5942910 DOI: 10.1126/sciadv.aas9274] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/21/2018] [Indexed: 06/08/2023]
Abstract
The mammalian retina system consists of a complicated photoreceptor structure, which exhibits extensive random synaptic connections. To study retinal development and degeneration, various experimental models have been used previously, but these models are often uncontrollable, are difficult to manipulate, and do not provide sufficient similarity or precision. Therefore, the mechanisms in many retinal diseases remain unclear because of the limited capability in observing the progression and molecular driving forces. For example, photoreceptor degeneration can spread to surrounding healthy photoreceptors via a phenomenon known as the bystander effect; however, no in-depth observations can be made to decipher the molecular mechanisms or the pathways that contribute to the spreading. It is then necessary to build dissociated neural networks to investigate the communications with controllability of cells and their treatment. We developed a neural network chip (NN-Chip) to load single neurons into highly ordered microwells connected by microchannels for synapse formation to build the neural network. By observing the distribution of apoptosis spreading from light-induced apoptotic cones to the surrounding cones, we demonstrated convincing evidence of the existence of a cone-to-cone bystander killing effect. Combining the NN-Chip with microinjection technology, we also found that the gap junction protein connexin 36 (Cx36) is critical for apoptosis spreading and the bystander effect in cones. In addition, our unique NN-Chip platform provides a quantitative, high-throughput tool for investigating signaling mechanisms and behaviors in neurons and opens a new avenue for screening potential drug targets to cure retinal diseases.
Collapse
Affiliation(s)
- Yuan Ma
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- College of Materials Sciences and Optoelectronics, University of Chinese Academy of Sciences, Beijing 100049, China
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Xin Han
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ricardo Bessa de Castro
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
- College of Engineering, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| | - Pengchao Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kai Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Zhongbo Hu
- College of Materials Sciences and Optoelectronics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
23
|
Kántor O, Varga A, Nitschke R, Naumann A, Énzsöly A, Lukáts Á, Szabó A, Németh J, Völgyi B. Bipolar cell gap junctions serve major signaling pathways in the human retina. Brain Struct Funct 2017; 222:2603-2624. [PMID: 28070649 DOI: 10.1007/s00429-016-1360-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/22/2016] [Indexed: 11/26/2022]
Abstract
Connexin36 (Cx36) constituent gap junctions (GJ) throughout the brain connect neurons into functional syncytia. In the retina they underlie the transmission, averaging and correlation of signals prior conveying visual information to the brain. This is the first study that describes retinal bipolar cell (BC) GJs in the human inner retina, whose function is enigmatic even in the examined animal models. Furthermore, a number of unique features (e.g. fovea, trichromacy, midget system) necessitate a reexamination of the animal model results in the human retina. Well-preserved postmortem human samples of this study are allowed to identify Cx36 expressing BCs neurochemically. Results reveal that both rod and cone pathway interneurons display strong Cx36 expression. Rod BC inputs to AII amacrine cells (AC) appear in juxtaposition to AII GJs, thus suggesting a strategic AII cell targeting by rod BCs. Cone BCs serving midget, parasol or koniocellular signaling pathways display a wealth of Cx36 expression to form homologously coupled arrays. In addition, they also establish heterologous GJ contacts to serve an exchange of information between parallel signaling streams. Interestingly, a prominent Cx36 expression was exhibited by midget system BCs that appear to maintain intimate contacts with bistratified BCs serving other pathways. These findings suggest that BC GJs in parallel signaling streams serve both an intra- and inter-pathway exchange of signals in the human retina.
Collapse
Affiliation(s)
- Orsolya Kántor
- Department of Neuroanatomy, Faculty of Medicine, Institute for Anatomy and Cell Biology, University of Freiburg, 79104, Freiburg, Germany
- MTA-PTE NAP B Retinal Electrical Synapses Research Group, Pécs, 7624, Hungary
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Alexandra Varga
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Roland Nitschke
- Life Imaging Center, Center for Biological Systems Analysis, University of Freiburg, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Angela Naumann
- Life Imaging Center, Center for Biological Systems Analysis, University of Freiburg, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Anna Énzsöly
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
- Department of Ophthalmology, Semmelweis University, Budapest, 1085, Hungary
| | - Ákos Lukáts
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Arnold Szabó
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - János Németh
- Department of Ophthalmology, Semmelweis University, Budapest, 1085, Hungary
| | - Béla Völgyi
- MTA-PTE NAP B Retinal Electrical Synapses Research Group, Pécs, 7624, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, 7624, Hungary.
- János Szentágothai Research Center, University of Pécs, Ifjúság street 20, Pécs, 7624, Hungary.
- Department of Ophthalmology, New York University Langone Medical Center, New York, NY, 10016, USA.
| |
Collapse
|
24
|
Kovács-Öller T, Debertin G, Balogh M, Ganczer A, Orbán J, Nyitrai M, Balogh L, Kántor O, Völgyi B. Connexin36 Expression in the Mammalian Retina: A Multiple-Species Comparison. Front Cell Neurosci 2017; 11:65. [PMID: 28337128 PMCID: PMC5343066 DOI: 10.3389/fncel.2017.00065] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/23/2017] [Indexed: 11/23/2022] Open
Abstract
Much knowledge about interconnection of human retinal neurons is inferred from results on animal models. Likewise, there is a lack of information on human retinal electrical synapses/gap junctions (GJ). Connexin36 (Cx36) forms GJs in both the inner and outer plexiform layers (IPL and OPL) in most species including humans. However, a comparison of Cx36 GJ distribution in retinas of humans and popular animal models has not been presented. To this end a multiple-species comparison was performed in retinas of 12 mammals including humans to survey the Cx36 distribution. Areas of retinal specializations were avoided (e.g., fovea, visual streak, area centralis), thus observed Cx36 distribution differences were not attributed to these species-specific architecture of central retinal areas. Cx36 was expressed in both synaptic layers in all examined retinas. Cx36 plaques displayed an inhomogenous IPL distribution favoring the ON sublamina, however, this feature was more pronounced in the human, swine and guinea pig while it was less obvious in the rabbit, squirrel monkey, and ferret retinas. In contrast to the relative conservative Cx36 distribution in the IPL, the labels in the OPL varied considerably among mammals. In general, OPL plaques were rare and rather small in rod dominant carnivores and rodents, whereas the human and the cone rich guinea pig retinas displayed robust Cx36 labels. This survey presented that the human retina displayed two characteristic features, a pronounced ON dominance of Cx36 plaques in the IPL and prevalent Cx36 plaque conglomerates in the OPL. While many species showed either of these features, only the guinea pig retina shared both. The observed similarities and subtle differences in Cx36 plaque distribution across mammals do not correspond to evolutionary distances but may reflect accomodation to lifestyles of examined species.
Collapse
Affiliation(s)
- Tamás Kovács-Öller
- Department of Experimental Zoology and Neurobiology, University of PécsPécs, Hungary; János Szentágothai Research CenterPécs, Hungary; Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary
| | - Gábor Debertin
- Department of Experimental Zoology and Neurobiology, University of PécsPécs, Hungary; János Szentágothai Research CenterPécs, Hungary; Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary
| | - Márton Balogh
- Department of Experimental Zoology and Neurobiology, University of PécsPécs, Hungary; János Szentágothai Research CenterPécs, Hungary; Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary
| | - Alma Ganczer
- Department of Experimental Zoology and Neurobiology, University of PécsPécs, Hungary; János Szentágothai Research CenterPécs, Hungary; Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary
| | - József Orbán
- János Szentágothai Research CenterPécs, Hungary; Department of Biophysics, University of PécsPécs, Hungary; High-Field Terahertz Research Group, Hungarian Academy of Sciences (MTA-PTE)Pécs, Hungary
| | - Miklós Nyitrai
- János Szentágothai Research CenterPécs, Hungary; Department of Biophysics, University of PécsPécs, Hungary; Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences (MTA-PTE)Pécs, Hungary
| | - Lajos Balogh
- National Research Institute for Radiobiology and Radiohygiene Budapest, Hungary
| | - Orsolya Kántor
- Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary; Department of Anatomy, Histology and Embryology, Semmelweis UniversityBudapest, Hungary; Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Faculty of Medicine, University of FreiburgFreiburg, Germany
| | - Béla Völgyi
- Department of Experimental Zoology and Neurobiology, University of PécsPécs, Hungary; János Szentágothai Research CenterPécs, Hungary; Retinal Electrical Synapses Research Group, Hungarian Academy of Sciences (MTA-PTE NAP B)Pécs, Hungary; Department of Ophthalmology, New York University Langone Medical Center, New YorkNY, USA
| |
Collapse
|
25
|
Asteriti S, Gargini C, Cangiano L. Connexin 36 expression is required for electrical coupling between mouse rods and cones. Vis Neurosci 2017; 34:E006. [PMID: 28965521 DOI: 10.1017/s0952523817000037] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Rod-cone gap junctions mediate the so-called "secondary rod pathway", one of three routes that convey rod photoreceptor signals across the retina. Connexin 36 (Cx36) is expressed at these gap junctions, but an unidentified connexin protein also seems to be expressed. Cx36 knockout mice have been used extensively in the quest to dissect the roles in vision of all three pathways, with the assumption, never directly tested, that rod-cone electrical coupling is abolished by deletion of this connexin isoform. We previously showed that when wild type mouse cones couple to rods, their apparent dynamic range is extended toward lower light intensities, with the appearance of large responses to dim flashes (up to several mV) originating in rods. Here we recorded from the cones of Cx36del[LacZ]/del[LacZ] mice and found that dim flashes of the same intensity evoked at most small sub-millivolt responses. Moreover, these residual responses originated in the cones themselves, since: (i) their spectral preference matched that of the recorded cone and not of rods, (ii) their time-to-peak was shorter than in coupled wild type cones, (iii) a pharmacological block of gap junctions did not reduce their amplitude. Taken together, our data show that rod signals are indeed absent in the cones of Cx36 knockout mice. This study is the first direct demonstration that Cx36 is crucial for the assembly of functional rod-cone gap junctional channels, implying that its genetic deletion is a reliable experimental approach to eliminate rod-cone coupling.
Collapse
Affiliation(s)
- Sabrina Asteriti
- Department of Translational Research,University of Pisa,Pisa,Italy
| | | | - Lorenzo Cangiano
- Department of Translational Research,University of Pisa,Pisa,Italy
| |
Collapse
|
26
|
Retinal gap junctions are involved in rhythmogenesis of neuronal activity at remote locations – Study on infra-slow oscillations in the rat olivary pretectal nucleus. Neuroscience 2016; 339:150-161. [DOI: 10.1016/j.neuroscience.2016.09.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/21/2016] [Accepted: 09/21/2016] [Indexed: 12/21/2022]
|
27
|
Tideman JWL, Fan Q, Polling JR, Guo X, Yazar S, Khawaja A, Höhn R, Lu Y, Jaddoe VWV, Yamashiro K, Yoshikawa M, Gerhold-Ay A, Nickels S, Zeller T, He M, Boutin T, Bencic G, Vitart V, Mackey DA, Foster PJ, MacGregor S, Williams C, Saw SM, Guggenheim JA, Klaver CCW. When do myopia genes have their effect? Comparison of genetic risks between children and adults. Genet Epidemiol 2016; 40:756-766. [PMID: 27611182 DOI: 10.1002/gepi.21999] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 07/05/2016] [Accepted: 07/17/2016] [Indexed: 01/10/2023]
Abstract
Previous studies have identified many genetic loci for refractive error and myopia. We aimed to investigate the effect of these loci on ocular biometry as a function of age in children, adolescents, and adults. The study population consisted of three age groups identified from the international CREAM consortium: 5,490 individuals aged <10 years; 5,000 aged 10-25 years; and 16,274 aged >25 years. All participants had undergone standard ophthalmic examination including measurements of axial length (AL) and corneal radius (CR). We examined the lead SNP at all 39 currently known genetic loci for refractive error identified from genome-wide association studies (GWAS), as well as a combined genetic risk score (GRS). The beta coefficient for association between SNP genotype or GRS versus AL/CR was compared across the three age groups, adjusting for age, sex, and principal components. Analyses were Bonferroni-corrected. In the age group <10 years, three loci (GJD2, CHRNG, ZIC2) were associated with AL/CR. In the age group 10-25 years, four loci (BMP2, KCNQ5, A2BP1, CACNA1D) were associated; and in adults 20 loci were associated. Association with GRS increased with age; β = 0.0016 per risk allele (P = 2 × 10-8 ) in <10 years, 0.0033 (P = 5 × 10-15 ) in 10- to 25-year-olds, and 0.0048 (P = 1 × 10-72 ) in adults. Genes with strongest effects (LAMA2, GJD2) had an early effect that increased with age. Our results provide insights on the age span during which myopia genes exert their effect. These insights form the basis for understanding the mechanisms underlying high and pathological myopia.
Collapse
Affiliation(s)
- J Willem L Tideman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Qiao Fan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | - Jan Roelof Polling
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Orthoptics, School of Applied Science Utrecht, Rotterdam, The Netherlands
| | - Xiaobo Guo
- Department of Statistical Science, School of Mathematics & Computational Science, Sun Yat-Sen University, Guangzhou, GD, China
- SYSU-CMU Shunde International Joint Research Institute, Guangzhou, GD, China
- Southern China Research Center of Statistical Science, Sun Yat-Sen University, Guangzhou, GD, China
| | - Seyhan Yazar
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Anthony Khawaja
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - René Höhn
- Department of Ophthalmology, University Medical Center, Mainz, Germany
- Department of Ophthalmology, Inselspital, Bern, Switzerland
| | - Yi Lu
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Vincent W V Jaddoe
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Kenji Yamashiro
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Munemitsu Yoshikawa
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Aslihan Gerhold-Ay
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center Mainz, Mainz, Germany
| | - Stefan Nickels
- Department of Ophthalmology, University Medical Center, Mainz, Germany
| | - Tanja Zeller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Mingguang He
- Centre for Eye Research Australia, University of Melbourne, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Thibaud Boutin
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Goran Bencic
- Department of Ophthalmology, Sisters of Mercy University Hospital, Zagreb, Croatia
| | - Veronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - David A Mackey
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Paul J Foster
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, United Kingdom
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Cathy Williams
- School of Social and Community Medicine, University of Bristol, Bristol, England
| | - Seang Mei Saw
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
- National University of Singapore Saw Swee Hock School of Public Health, Singapore Eye Research Institute, Singapore National Eye Centre, Singapore
| | | | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
28
|
Kántor O, Benkő Z, Énzsöly A, Dávid C, Naumann A, Nitschke R, Szabó A, Pálfi E, Orbán J, Nyitrai M, Németh J, Szél Á, Lukáts Á, Völgyi B. Characterization of connexin36 gap junctions in the human outer retina. Brain Struct Funct 2016; 221:2963-84. [PMID: 26173976 DOI: 10.1007/s00429-015-1082-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 07/06/2015] [Indexed: 10/23/2022]
Abstract
Retinal connexins (Cx) form gap junctions (GJ) in key circuits that transmit average or synchronize signals. Expression of Cx36, -45, -50 and -57 have been described in many species but there is still a disconcerting paucity of information regarding the Cx makeup of human retinal GJs. We used well-preserved human postmortem samples to characterize Cx36 GJ constituent circuits of the outer plexiform layer (OPL). Based on their location, morphometric characteristics and co-localizations with outer retinal neuronal markers, we distinguished four populations of Cx36 plaques in the human OPL. Three of these were comprised of loosely scattered Cx36 plaques; the distalmost population 1 formed cone-to-rod GJs, population 2 in the mid-OPL formed cone-to-cone GJs, whereas the proximalmost population 4 likely connected bipolar cell dendrites. The fourth population (population 3) of Cx36 plaques conglomerated beneath cone pedicles and connected dendritic tips of bipolar cells that shared a common presynaptic cone. Overall, we show that the human outer retina displays a diverse cohort of Cx36 GJ that follows the general mammalian scheme and display a great functional diversity.
Collapse
Affiliation(s)
- Orsolya Kántor
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - Zsigmond Benkő
- Department of Theory, Wigner Research Center for Physics of the Hungarian Academy of Sciences, Budapest, 1121, Hungary
- Semmelweis University School of Ph.D. Studies, Budapest, 1085, Hungary
| | - Anna Énzsöly
- Department of Ophthalmology, Semmelweis University, Budapest, 1085, Hungary
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Csaba Dávid
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Angela Naumann
- Life Imaging Center, Center for Biological Systems Analysis, Albert-Ludwigs University, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, 79104, Freiburg, Germany
| | - Roland Nitschke
- Life Imaging Center, Center for Biological Systems Analysis, Albert-Ludwigs University, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, 79104, Freiburg, Germany
| | - Arnold Szabó
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Emese Pálfi
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, 1094, Hungary
| | - József Orbán
- Department of Biophysics, University of Pécs, Pécs, 7624, Hungary
- János Szentágothai Research Center, University of Pécs, Ifjúság str. 6, 7624, Pécs, Hungary
| | - Miklós Nyitrai
- Department of Biophysics, University of Pécs, Pécs, 7624, Hungary
- János Szentágothai Research Center, University of Pécs, Ifjúság str. 6, 7624, Pécs, Hungary
| | - János Németh
- Department of Ophthalmology, Semmelweis University, Budapest, 1085, Hungary
| | - Ágoston Szél
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Ákos Lukáts
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, 1094, Hungary
| | - Béla Völgyi
- János Szentágothai Research Center, University of Pécs, Ifjúság str. 6, 7624, Pécs, Hungary.
- MTA-PTE NAP B Retinal Electrical Synapses Research Group, Pécs, 7624, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, 7624, Hungary.
- Department of Ophthalmology, New York University Langone Medical Center, New York, NY, 10016, USA.
| |
Collapse
|
29
|
Calcium buffer proteins are specific markers of human retinal neurons. Cell Tissue Res 2016; 365:29-50. [PMID: 26899253 DOI: 10.1007/s00441-016-2376-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 02/04/2016] [Indexed: 10/22/2022]
Abstract
Ca(2+)-buffer proteins (CaBPs) modulate the temporal and spatial characteristics of transient intracellular Ca(2+)-concentration changes in neurons in order to fine-tune the strength and duration of the output signal. CaBPs have been used as neurochemical markers to identify and trace neurons of several brain loci including the mammalian retina. The CaBP content of retinal neurons, however, varies between species and, thus, the results inferred from animal models cannot be utilised directly by clinical ophthalmologists. Moreover, the shortage of well-preserved human samples greatly impedes human retina studies at the cellular and network level. Our purpose has therefore been to examine the distribution of major CaBPs, including calretinin, calbindin-D28, parvalbumin and the recently discovered secretagogin in exceptionally well-preserved human retinal samples. Based on a combination of immunohistochemistry, Neurolucida tracing and Lucifer yellow injections, we have established a database in which the CaBP marker composition can be defined for morphologically identified cell types of the human retina. Hence, we describe the full CaBP make-up for a number of human retinal neurons, including HII horizontal cells, AII amacrine cells, type-1 tyrosine-hydroxylase-expressing amacrine cells and other lesser known neurons. We have also found a number of unidentified cells whose morphology remains to be characterised. We present several examples of the colocalisation of two or three CaBPs with slightly different subcellular distributions in the same cell strongly suggesting a compartment-specific division of labour of Ca(2+)-buffering by CaBPs. Our work thus provides a neurochemical framework for future ophthalmological studies and renders new information concerning the cellular and subcellular distribution of CaBPs for experimental neuroscience.
Collapse
|
30
|
HCN1 Channels Enhance Rod System Responsivity in the Retina under Conditions of Light Exposure. PLoS One 2016; 11:e0147728. [PMID: 26807953 PMCID: PMC4725747 DOI: 10.1371/journal.pone.0147728] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/07/2016] [Indexed: 11/20/2022] Open
Abstract
Purpose Vision originates in rods and cones at the outer retina. Already at these early stages, diverse processing schemes shape and enhance image information to permit perception over a wide range of lighting conditions. In this work, we address the role of hyperpolarization-activated and cyclic nucleotide-gated channels 1 (HCN1) in rod photoreceptors for the enhancement of rod system responsivity under conditions of light exposure. Methods To isolate HCN1 channel actions in rod system responses, we generated double mutant mice by crossbreeding Hcn1-/- mice with Cnga3-/- mice in which cones are non-functional. Retinal function in the resulting Hcn1-/-Cnga3-/- animals was followed by means of electroretinography (ERG) up to the age of four month. Retinal imaging via scanning laser ophthalmoscopy (SLO) and optical coherence tomography (OCT) was also performed to exclude potential morphological alterations. Results This study on Hcn1-/-Cnga3-/- mutant mice complements our previous work on HCN1 channel function in the retina. We show here in a functional rod-only setting that rod responses following bright light exposure terminate without the counteraction of HCN channels much later than normal. The resulting sustained signal elevation does saturate the retinal network due to an intensity-dependent reduction in the dynamic range. In addition, the lack of rapid adaptational feedback modulation of rod photoreceptor output via HCN1 in this double mutant limits the ability to follow repetitive (flicker) stimuli, particularly under mesopic conditions. Conclusions This work corroborates the hypothesis that, in the absence of HCN1-mediated feedback, the amplitude of rod signals remains at high levels for a prolonged period of time, leading to saturation of the retinal pathways. Our results demonstrate the importance of HCN1 channels for regular vision.
Collapse
|
31
|
Bolte P, Herrling R, Dorgau B, Schultz K, Feigenspan A, Weiler R, Dedek K, Janssen-Bienhold U. Expression and Localization of Connexins in the Outer Retina of the Mouse. J Mol Neurosci 2015; 58:178-92. [PMID: 26453550 DOI: 10.1007/s12031-015-0654-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/08/2015] [Indexed: 01/30/2023]
Abstract
The identification of the proteins that make up the gap junction channels between rods and cones is of crucial importance to understand the functional role of photoreceptor coupling within the retinal network. In vertebrates, connexin proteins constitute the structural components of gap junction channels. Connexin36 is known to be expressed in cones whereas extensive investigations have failed to identify the corresponding connexin expressed in rods. Using immunoelectron microscopy, we demonstrate that connexin36 (Cx36) is present in gap junctions of cone but not rod photoreceptors in the mouse retina. To identify the rod connexin, we used nested reverse transcriptase polymerase chain reaction and tested retina and photoreceptor samples for messenger RNA (mRNA) expression of all known connexin genes. In addition to connexin36, we detected transcripts for connexin32, connexin43, connexin45, connexin50, and connexin57 in photoreceptor samples. Immunohistochemistry showed that connexin43, connexin45, connexin50, and connexin57 proteins are expressed in the outer plexiform layer. However, none of these connexins was detected at gap junctions between rods and cones as a counterpart of connexin36. Therefore, the sought-after rod protein must be either an unknown connexin sequence, a connexin36 splice product not detected by our antibodies, or a protein from a further gap junction protein family.
Collapse
Affiliation(s)
- Petra Bolte
- Neurobiology Group, Department for Neuroscience, School of Medicine and Health Sciences, University of Oldenburg, 26111, Oldenburg, Germany.,Animal Navigation, University of Oldenburg, 26111, Oldenburg, Germany
| | - Regina Herrling
- Neurobiology Group, Department for Neuroscience, School of Medicine and Health Sciences, University of Oldenburg, 26111, Oldenburg, Germany
| | - Birthe Dorgau
- Neurobiology Group, Department for Neuroscience, School of Medicine and Health Sciences, University of Oldenburg, 26111, Oldenburg, Germany.,Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Konrad Schultz
- Neurobiology Group, Department for Neuroscience, School of Medicine and Health Sciences, University of Oldenburg, 26111, Oldenburg, Germany
| | - Andreas Feigenspan
- Neurobiology Group, Department for Neuroscience, School of Medicine and Health Sciences, University of Oldenburg, 26111, Oldenburg, Germany.,Animal Physiology, FAU Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Reto Weiler
- Neurobiology Group, Department for Neuroscience, School of Medicine and Health Sciences, University of Oldenburg, 26111, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, 26111, Oldenburg, Germany
| | - Karin Dedek
- Neurobiology Group, Department for Neuroscience, School of Medicine and Health Sciences, University of Oldenburg, 26111, Oldenburg, Germany. .,Research Center Neurosensory Science, University of Oldenburg, 26111, Oldenburg, Germany.
| | - Ulrike Janssen-Bienhold
- Neurobiology Group, Department for Neuroscience, School of Medicine and Health Sciences, University of Oldenburg, 26111, Oldenburg, Germany. .,Research Center Neurosensory Science, University of Oldenburg, 26111, Oldenburg, Germany.
| |
Collapse
|
32
|
Identification of myopia-associated WNT7B polymorphisms provides insights into the mechanism underlying the development of myopia. Nat Commun 2015; 6:6689. [DOI: 10.1038/ncomms7689] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 02/20/2015] [Indexed: 11/08/2022] Open
|
33
|
Kovács-Öller T, Raics K, Orbán J, Nyitrai M, Völgyi B. Developmental changes in the expression level of connexin36 in the rat retina. Cell Tissue Res 2014; 358:289-302. [PMID: 25110193 DOI: 10.1007/s00441-014-1967-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 07/09/2014] [Indexed: 02/03/2023]
Abstract
Connexin36 (Cx36) is the major gap junction forming protein in the brain and the retina; thus, alterations in its expression indicate changes in the corresponding circuitry. Many structural changes occur in the early postnatal retina before functional neuronal circuits are finalized, including those that incorporate gap junctions. To reveal the time-lapse formation of inner retinal gap junctions, we examine the developing postnatal rat retina from birth (P0) to young adult age (P20) and follow the expression of Cx36 in the mRNA and protein levels. We found a continuous elevation in the expression of both the Cx36 transcript and protein between P0 and P20 and a somewhat delayed Cx36 plaque formation throughout the inner plexiform layer (IPL) starting at P10. By using tristratificated calretinin positive (CaR(+)) fibers in the IPL as a guide, we detected a clear preference of Cx36 plaques for the ON sublamina from the earliest time of detection. This distributional preference became more pronounced at P15 and P20 due to the emergence and widespread expression of large (>0.1 μm(2)) Cx36 plaques in the ON sublamina. Finally, we showed that parvalbumin-positive (PV(+)) AII amacrine cell dendrites colocalize with Cx36 plaques as early as P10 in strata 3 and 4, whereas colocalizations in stratum 5 became characteristic only around P20. We conclude that Cx36 expression in the rat IPL displays a characteristic succession of changes during retinogenesis reflecting the formation of the underlying electrical synaptic circuitry. In particular, AII cell gap junctions, first formed with ON cone bipolar cells and later with other AII amacrine cells, accounted for the observed Cx36 expressional changes.
Collapse
Affiliation(s)
- Tamás Kovács-Öller
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Ifjúság street 6, Hungary
| | | | | | | | | |
Collapse
|
34
|
Palacios-Muñoz A, Escobar MJ, Vielma A, Araya J, Astudillo A, Valdivia G, García IE, Hurtado J, Schmachtenberg O, Martínez AD, Palacios AG. Role of connexin channels in the retinal light response of a diurnal rodent. Front Cell Neurosci 2014; 8:249. [PMID: 25202238 PMCID: PMC4142540 DOI: 10.3389/fncel.2014.00249] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 08/05/2014] [Indexed: 01/02/2023] Open
Abstract
Several studies have shown that connexin channels play an important role in retinal neural coding in nocturnal rodents. However, the contribution of these channels to signal processing in the retina of diurnal rodents remains unclear. To gain insight into this problem, we studied connexin expression and the contribution of connexin channels to the retinal light response in the diurnal rodent Octodon degus (degu) compared to rat, using in vivo ERG recording under scotopic and photopic light adaptation. Analysis of the degu genome showed that the common retinal connexins present a high degree of homology to orthologs expressed in other mammals, and expression of Cx36 and Cx43 was confirmed in degu retina. Cx36 localized mainly to the outer and inner plexiform layers (IPLs), while Cx43 was expressed mostly in cells of the retinal pigment epithelium. Under scotopic conditions, the b-wave response amplitude was strongly reduced by 18-β-glycyrrhetinic acid (β-GA) (−45.1% in degu, compared to −52.2% in rat), suggesting that connexins are modulating this response. Remarkably, under photopic adaptation, β-GA increased the ERG b-wave amplitude in degu (+107.2%) while reducing it in rat (−62.3%). Moreover, β-GA diminished the spontaneous action potential firing rate in ganglion cells (GCs) and increased the response latency of ON and OFF GCs. Our results support the notion that connexins exert a fine-tuning control of the retinal light response and have an important role in retinal neural coding.
Collapse
Affiliation(s)
- Angelina Palacios-Muñoz
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Maria J Escobar
- Departamento de Electrónica, Universidad Técnico Federico Santa María Valparaíso, Chile
| | - Alex Vielma
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Joaquín Araya
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Aland Astudillo
- Departamento de Electrónica, Universidad Técnico Federico Santa María Valparaíso, Chile
| | - Gonzalo Valdivia
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Isaac E García
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - José Hurtado
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile ; Instituto de Sistemas Complejos de Valparaíso Valparaíso, Chile
| | - Oliver Schmachtenberg
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Agustín D Martínez
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Adrian G Palacios
- Facultad de Ciencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile ; Instituto de Sistemas Complejos de Valparaíso Valparaíso, Chile
| |
Collapse
|
35
|
Hysi PG, Mahroo OA, Cumberland P, Wojciechowski R, Williams KM, Young TL, Mackey DA, Rahi JS, Hammond CJ. Common mechanisms underlying refractive error identified in functional analysis of gene lists from genome-wide association study results in 2 European British cohorts. JAMA Ophthalmol 2014; 132:50-6. [PMID: 24264139 DOI: 10.1001/jamaophthalmol.2013.6022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
IMPORTANCE To date, relatively few genes responsible for a fraction of heritability have been identified by means of large genetic association studies of refractive error. OBJECTIVE To explore the genetic mechanisms that lead to refractive error in the general population. DESIGN, SETTING, AND PARTICIPANTS Genome-wide association studies were carried out in 2 British population-based independent cohorts (N = 5928 participants) to identify genes moderately associated with refractive error. MAIN OUTCOMES AND MEASURES Enrichment analyses were used to identify sets of genes overrepresented in both cohorts. Enriched groups of genes were compared between both participating cohorts as a further measure against random noise. RESULTS Groups of genes enriched at highly significant statistical levels were remarkably consistent in both cohorts. In particular, these results indicated that plasma membrane (P = 7.64 × 10⁻³⁰), cell-cell adhesion (P = 2.42 × 10⁻¹⁸), synaptic transmission (P = 2.70 × 10⁻¹⁴), calcium ion binding (P = 3.55 × 10⁻¹⁵), and cation channel activity (P = 2.77 × 10⁻¹⁴) were significantly overrepresented in relation to refractive error. CONCLUSIONS AND RELEVANCE These findings provide evidence that development of refractive error in the general population is related to the intensity of photosignal transduced from the retina, which may have implications for future interventions to minimize this disorder. Pathways connected to the procession of the nerve impulse are major mechanisms involved in the development of refractive error in populations of European origin.
Collapse
Affiliation(s)
- Pirro G Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, England
| | - Omar A Mahroo
- Department of Ophthalmology, King's College London, London, England3Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, England
| | - Phillippa Cumberland
- Centre for Paediatric Epidemiology and Biostatistics, Institute of Child Health, University College London, London, England
| | | | - Katie M Williams
- Department of Ophthalmology, King's College London, London, England
| | - Terri L Young
- Center for Human Genetics, Duke University Medical Center, Durham, North Carolina
| | - David A Mackey
- Lions Eye Institute, University of Western Australia, Centre for Ophthalmology and Visual Science, Perth, Australia
| | - Jugnoo S Rahi
- Centre for Paediatric Epidemiology and Biostatistics, Institute of Child Health, University College London, London, England
| | - Christopher J Hammond
- Department of Twin Research and Genetic Epidemiology, King's College London, London, England2Department of Ophthalmology, King's College London, London, England
| |
Collapse
|
36
|
Abstract
Rod and cone photoreceptors are coupled by gap junctions (GJs), relatively large channels able to mediate both electrical and molecular communication. Despite their critical location in our visual system and evidence that they are dynamically gated for dark/light adaptation, the full impact that rod–cone GJs can have on cone function is not known. We recorded the photovoltage of mouse cones and found that the initial level of rod input increased spontaneously after obtaining intracellular access. This process allowed us to explore the underlying coupling capacity to rods, revealing that fully coupled cones acquire a striking rod-like phenotype. Calcium, a candidate mediator of the coupling process, does not appear to be involved on the cone side of the junctional channels. Our findings show that the anatomical substrate is adequate for rod–cone coupling to play an important role in vision and, possibly, in biochemical signaling among photoreceptors. DOI:http://dx.doi.org/10.7554/eLife.01386.001 People can see in a range of light levels—from dim moonlight to bright midday sun—because our eyes contain two types of light-sensitive cells: rods and cones. Rods are more plentiful than cones, and while they are sensitive at low light levels, rods can only provide grey-scale vision. Further, bright light can rapidly ‘dazzle’ the ability of rods to see in near-darkness, and they are slow to recover when this happens. In contrast, cones need bright light to function, but allow us to see in colour. The signals received by rods and cones are sent through the optic nerve to the brain, where they are interpreted as vision. However, ‘gap junctions’ that connect the rods and cones allow for electrical and chemical ‘crosstalk’ between these cells, before the signals then travel along the optic nerve. Furthermore, even though it is thought that the connections between rods and cones are regulated in response to light, the body’s daily rhythms and other biochemical signals, their importance for vision is not known. Now, Asteriti et al. have taken tissue slices from the retinas at the back of mice eyes, and measured the electrical signals generated when cones are exposed to light. This revealed that the rod-cone coupling is strong enough to make the cones responsive to dim light, just like rods. Moreover, the cones also recovered slowly after being exposed to flashes of bright light. When chemical inhibitors were used to block the gap junctions, the cones stopped behaving like rods and became less sensitive to dim light. The findings of Asteriti et al. show that rod-cone coupling is sufficient to play an important role in vision. The next challenge is to find out what this role is, and how it might be affected by different physiological conditions, including stress and injury. DOI:http://dx.doi.org/10.7554/eLife.01386.002
Collapse
Affiliation(s)
- Sabrina Asteriti
- Department of Translational Research, University of Pisa, Pisa, Italy
| | | | | |
Collapse
|
37
|
Weng S, Estevez ME, Berson DM. Mouse ganglion-cell photoreceptors are driven by the most sensitive rod pathway and by both types of cones. PLoS One 2013; 8:e66480. [PMID: 23762490 PMCID: PMC3676382 DOI: 10.1371/journal.pone.0066480] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/07/2013] [Indexed: 11/21/2022] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (iprgcs) are depolarized by light by two mechanisms: directly, through activation of their photopigment melanopsin; and indirectly through synaptic circuits driven by rods and cones. To learn more about the rod and cone circuits driving ipRGCs, we made multielectrode array (MEA) and patch-clamp recordings in wildtype and genetically modified mice. Rod-driven ON inputs to ipRGCs proved to be as sensitive as any reaching the conventional ganglion cells. These signals presumably pass in part through the primary rod pathway, involving rod bipolar cells and AII amacrine cells coupled to ON cone bipolar cells through gap junctions. Consistent with this interpretation, the sensitive rod ON input to ipRGCs was eliminated by pharmacological or genetic disruption of gap junctions, as previously reported for conventional ganglion cells. A presumptive cone input was also detectable as a brisk, synaptically mediated ON response that persisted after disruption of rod ON pathways. This was roughly three log units less sensitive than the rod input. Spectral analysis revealed that both types of cones, the M- and S-cones, contribute to this response and that both cone types drive ON responses. This contrasts with the blue-OFF, yellow-ON chromatic opponency reported in primate ipRGCs. The cone-mediated response was surprisingly persistent during steady illumination, echoing the tonic nature of both the rod input to ipRGCs and their intrinsic, melanopsin-based phototransduction. These synaptic inputs greatly expand the dynamic range and spectral bandpass of the non-image-forming visual functions for which ipRGCs provide the principal retinal input.
Collapse
Affiliation(s)
- Shijun Weng
- Department of Neuroscience, Brown University, Providence, Rhode Island, United States of America
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, P.R. China
| | - Maureen E. Estevez
- Department of Neuroscience, Brown University, Providence, Rhode Island, United States of America
| | - David M. Berson
- Department of Neuroscience, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
38
|
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: 105] [Impact Index Per Article: 8.8] [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
|
39
|
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
|
40
|
Gap-junctional coupling of mammalian rod photoreceptors and its effect on visual detection. J Neurosci 2012; 32:3552-62. [PMID: 22399777 DOI: 10.1523/jneurosci.2144-11.2012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The presence of gap junctions between rods in mammalian retina suggests a role for rod-rod coupling in human vision. Rod coupling is known to reduce response variability, but because junctional conductances are not known, the downstream effects on visual performance are uncertain. Here we assessed rod coupling in guinea pig retina by measuring: (1) the variability in responses to dim flashes, (2) Neurobiotin tracer coupling, and (3) junctional conductances. Results were consolidated into an electrical network model and a model of human psychophysical detection. Guinea pig rods form tracer pools of 1 to ∼20 rods, with junctional conductances averaging ∼350 pS. We calculate that coupling will reduce human dark-adapted sensitivity ∼10% by impairing the noise filtering of the synapse between rods and rod bipolar cells. However, coupling also mitigates synaptic saturation and is thus calculated to improve sensitivity when stimuli are spatially restricted or are superimposed over background illumination.
Collapse
|
41
|
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
|
42
|
Modulation of rod photoreceptor output by HCN1 channels is essential for regular mesopic cone vision. Nat Commun 2011; 2:532. [PMID: 22068599 DOI: 10.1038/ncomms1540] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 10/10/2011] [Indexed: 11/08/2022] Open
Abstract
Retinal photoreceptors permit visual perception over a wide range of lighting conditions. Rods work best in dim, and cones in bright environments, with considerable functional overlap at intermediate (mesopic) light levels. At many sites in the outer and inner retina where rod and cone signals interact, gap junctions, particularly those containing Connexin36, have been identified. However, little is known about the dynamic processes associated with the convergence of rod and cone system signals into ON- and OFF-pathways. Here we show that proper cone vision under mesopic conditions requires rapid adaptational feedback modulation of rod output via hyperpolarization-activated and cyclic nucleotide-gated channels 1. When these channels are absent, sustained rod responses following bright light exposure saturate the retinal network, resulting in a loss of downstream cone signalling. By specific genetic and pharmacological ablation of key signal processing components, regular cone signalling can be restored, thereby identifying the sites involved in functional rod-cone interactions.
Collapse
|
43
|
Lehner M, Wisłowska-Stanek A, Skórzewska A, Maciejak P, Szyndler J, Turzyńska D, Sobolewska A, Płaźnik A. Differences in the density of GABA-A receptor alpha-2 subunits and gephyrin in brain structures of rats selected for low and high anxiety in basal and fear-stimulated conditions, in a model of contextual fear conditioning. Neurobiol Learn Mem 2010; 94:499-508. [DOI: 10.1016/j.nlm.2010.09.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2010] [Revised: 08/31/2010] [Accepted: 09/01/2010] [Indexed: 11/27/2022]
|
44
|
Bergholz R, Staks T, Rüther K. Effects of the AMPA antagonist ZK 200775 on visual function: a randomized controlled trial. PLoS One 2010; 5:e12111. [PMID: 20711429 PMCID: PMC2920815 DOI: 10.1371/journal.pone.0012111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Accepted: 06/10/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND ZK 200775 is an antagonist at the alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptor and had earned attention as a possible neuroprotective agent in cerebral ischemia. Probands receiving the agent within phase I trials reported on an alteration of visual perception. In this trial, the effects of ZK 200775 on the visual system were analyzed in detail. METHODOLOGY In a randomised controlled trial we examined eyes and vision before and after the intravenous administration of two different doses of ZK 200775 and placebo. There were 3 groups of 6 probands each: Group 1 recieved 0.03 mg/kg/h, group 2 0.75 mg/kg/h of ZK 200775, the control group received 0.9% sodium chloride solution. Probands were healthy males aged between 57 and 69 years. The following methods were applied: clinical examination, visual acuity, ophthalmoscopy, colour vision, rod absolute threshold, central visual field, pattern-reversal visual evoked potentials (pVEP), ON-OFF and full-field electroretinogram (ERG). PRINCIPAL FINDINGS No effect of ZK 200775 was seen on eye position or motility, stereopsis, pupillary function or central visual field testing. Visual acuity and dark vision deteriorated significantly in both treated groups. Color vision was most remarkably impaired. The dark-adapted ERG revealed a reduction of oscillatory potentials (OP) and partly of the a- and b-wave, furthermore an alteration of b-wave morphology and an insignificantly elevated b/a-ratio. Cone-ERG modalities showed decreased amplitudes and delayed implicit times. In the ON-OFF ERG the ON-answer amplitudes increased whereas the peak times of the OFF-answer were reduced. The pattern VEP exhibited lower amplitudes and prolonged peak times. CONCLUSIONS The AMPA receptor blockade led to a strong impairment of typical OFF-pathway functions like color vision and the cone ERG. On the other hand the ON-pathway as measured by dark vision and the scotopic ERG was affected as well. This further elucidates the interdependence of both pathways. TRIAL REGISTRATION ClinicalTrials.gov NCT00999284.
Collapse
Affiliation(s)
- Richard Bergholz
- Department of Ophthalmology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.
| | | | | |
Collapse
|
45
|
Maciejak P, Szyndler J, Lehner M, Turzyńska D, Sobolewska A, Bidziński A, Płaźnik A. The differential effects of protein synthesis inhibition on the expression and reconsolidation of pentylenetetrazole kindled seizures. Epilepsy Behav 2010; 18:193-200. [PMID: 20605533 DOI: 10.1016/j.yebeh.2010.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 04/06/2010] [Accepted: 04/07/2010] [Indexed: 11/26/2022]
Abstract
This work attempted to answer the question whether the central processes engaged in the memory formation and the epilepsy development are governed by the overlapping mechanisms. The effects of the protein synthesis inhibitor cycloheximide (CHX) were examined on the expression and reconsolidation of pentylenetetrazole (PTZ) - induced kindled seizures and for comparative purposes, on the reconsolidation of conditioned fear response (conditioned freezing). It was found that post-test intracerebroventricular administration of CHX (125microg/5microl) significantly attenuated the expression of a conditioned fear response examined 24h later. Thus, inhibition of de novo brain protein synthesis interfered with the reconsolidation of a conditioned response. CHX given at the same dose repeatedly to fully kindled rats immediately after three consecutive sessions of PTZ-induced seizures (35mg/kg ip) did not modify the strength of convulsions. On the other hand, CHX significantly attenuated the strength of convulsions when the drug was administered 1h before the PTZ injection, which occurred every second day for three consecutive sessions. However, when CHX was omitted in a consecutive session, PTZ induced a fully developed expression of tonic-clonic convulsions, thereby indicating that CHX-induced changes in seizure intensity were transitory. Western Blot analysis confirmed that CHX potently inhibited PTZ-induced protein synthesis (c-Fos) in the rat brain, examined 60min after CHX and PTZ administration. The present findings suggest that the mechanisms underlying kindling are resistant to modification, even under the influence of protein synthesis inhibitors, and that there are important differences between the processes of learning and kindling seizures.
Collapse
Affiliation(s)
- Piotr Maciejak
- Department of Neurochemistry, Institute of Psychiatry and Neurology, 9 Sobieskiego Street, 02-957 Warsaw, Poland.
| | | | | | | | | | | | | |
Collapse
|
46
|
Expression and modulation of connexin 30.2, a novel gap junction protein in the mouse retina. Vis Neurosci 2010; 27:91-101. [PMID: 20537217 DOI: 10.1017/s0952523810000131] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mammalian retinae express multiple connexins that mediate the metabolic and electrical coupling of various cell types. In retinal neurons, only connexin 36, connexin 45, connexin 50, and connexin 57 have been described so far. Here, we present an analysis of a novel retinal connexin, connexin 30.2 (Cx30.2), and its regulation in the mouse retina. To analyze the expression of Cx30.2, we used a transgenic mouse line in which the coding region of Cx30.2 was replaced by lacZ reporter DNA. We detected the lacZ signal in the nuclei of neurons located in the inner nuclear layer and the ganglion cell layer (GCL). In this study, we focused on the GCL and characterized the morphology of the Cx30.2-expressing cells. Using immunocytochemistry and intracellular dye injections, we found six different types of Cx30.2-expressing ganglion cells: one type of ON-OFF, three types of OFF, and two types of ON ganglion cells; among the latter was the RG A1 type. We show that RG A1 cells were heterologously coupled to numerous displaced amacrine cells. Our results suggest that these gap junction channels may be heterotypic, involving Cx30.2 and a connexin yet unidentified in the mouse retina. Gap junction coupling can be modulated by protein kinases, a process that plays a major role in retinal adaptation. Therefore, we studied the protein kinase-induced modulation of coupling between RG A1 and displaced amacrine cells. Our data provide evidence that coupling of RG A1 cells to displaced amacrine cells is mediated by Cx30.2 and that the extent of this coupling is modulated by protein kinase C.
Collapse
|
47
|
Pan F, Paul DL, Bloomfield SA, Völgyi B. Connexin36 is required for gap junctional coupling of most ganglion cell subtypes in the mouse retina. J Comp Neurol 2010; 518:911-27. [PMID: 20058323 PMCID: PMC2860380 DOI: 10.1002/cne.22254] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Converging evidence indicates that electrical synaptic transmission via gap junctions plays a crucial role in signal processing in the retina. In particular, amacrine and ganglion cells express numerous gap junctions, resulting in extensive electrical networks in the proximal retina. Both connexin36 (Cx36) and connexin45 (Cx45) subunits are widely distributed in the inner plexiform layer (IPL) and therefore are likely contribute to gap junctions formed by a number of ganglion cell subtypes. In the present study, we used the gap junction-permeant tracer Neurobiotin to compare the coupling pattern of different ganglion cell subtypes in wild-type (WT) and Cx36 knockout (KO) mouse retinas. We found that homologous ganglion-to-ganglion cell coupling was lost for two subtypes after deletion of Cx36, whereas two other ganglion cell subtypes retained homologous coupling in the KO mouse. In contrast, deletion of Cx36 resulted in a partial or complete loss of ganglion-to-amacrine cell heterologous coupling in 9 of 10 ganglion cell populations studied. Overall, our results indicate that Cx36 is the predominant subunit of gap junctions in the proximal mouse retina, expressed by most ganglion cell subtypes, and thereby likely plays a major role in the concerted activity generated by electrical synapses.
Collapse
Affiliation(s)
- Feng Pan
- Department of Ophthalmology, New York University School of Medicine, New York, New York 10016
- Department of Physiology and Neuroscience, New York University School of Medicine, New York, New York 10016
| | - David L. Paul
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115
| | - Stewart A. Bloomfield
- Department of Ophthalmology, New York University School of Medicine, New York, New York 10016
- Department of Physiology and Neuroscience, New York University School of Medicine, New York, New York 10016
| | - Béla Völgyi
- Department of Ophthalmology, New York University School of Medicine, New York, New York 10016
| |
Collapse
|
48
|
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: 69] [Impact Index Per Article: 4.3] [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
|
49
|
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]
|
50
|
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: 139] [Impact Index Per Article: 8.7] [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
|