1
|
Nagy-Watson NV, Jonz MG. Hypoxia increases intracellular calcium in glutamate-activated horizontal cells of goldfish retina via mitochondrial K ATP channels and intracellular stores. Comp Biochem Physiol A Mol Integr Physiol 2025; 300:111786. [PMID: 39608485 DOI: 10.1016/j.cbpa.2024.111786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Central neurons of the common goldfish (Carassius auratus) are exceptional in their capacity to survive Ca2+-induced excitotoxicity and cell death during hypoxia. Horizontal cells (HCs) are inhibitory interneurons of the retina that are tonically depolarized by the neurotransmitter, glutamate, yet preserve intracellular Ca2+ homeostasis. In HCs isolated from goldfish, and in the absence of glutamatergic input, intracellular Ca2+ concentration ([Ca2+]i) is protected from prolonged exposure to hypoxia by mitochondrial ATP-dependent K+ (mKATP) channel activity. In the present study, we investigated the effects of hypoxia upon [Ca2+]i in isolated HCs during tonic activation by glutamate to better predict the effects of hypoxia in the active retina. Dynamic changes in [Ca2+]i were measured using the ratiometric Ca2+ indicator, Fura-2. Application of 100 μM glutamate during hypoxia (PO2 = 25 mmHg) produced a 1.3-fold greater rise in [Ca2+]i compared to the same glutamate stimulus during normoxia. The hypoxia-dependent increase in [Ca2+]i was abolished by application of 5-hydroxydecanoic acid, which renders mKATP channels inactive. Extracellular Ca2+ did not contribute to the elevated [Ca2+]i observed during hypoxia, as the effect persisted in Ca2+-free solution and during application of verapamil, an L-type Ca2+ channel blocker. By contrast, inhibition of the mitochondrial Ca2+ uniporter or ryanodine receptors (with ruthenium red or ryanodine, respectively) abolished the hypoxia-dependent rise in [Ca2+]i. This study reports an mKATP-dependent rise in [Ca2+]i during hypoxia in HCs activated by glutamate, and suggests roles for the mitochondria and intracellular Ca2+ stores in regulating this mechanism.
Collapse
Affiliation(s)
| | - Michael G Jonz
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada; Brain and Mind Research Institute, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
2
|
Morikawa R, Rodrigues TM, Schreyer HM, Cowan CS, Nadeau S, Graff-Meyer A, Patino-Alvarez CP, Khani MH, Jüttner J, Roska B. The sodium-bicarbonate cotransporter Slc4a5 mediates feedback at the first synapse of vision. Neuron 2024; 112:3715-3733.e9. [PMID: 39317184 PMCID: PMC11602199 DOI: 10.1016/j.neuron.2024.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 07/22/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024]
Abstract
Feedback at the photoreceptor synapse is the first neuronal circuit computation in vision, which influences downstream activity patterns within the visual system. Yet, the identity of the feedback signal and the mechanism of synaptic transmission are still not well understood. Here, we combined perturbations of cell-type-specific genes of mouse horizontal cells with two-photon imaging of the result of light-induced feedback in cones and showed that the electrogenic bicarbonate transporter Slc4a5, but not the electroneutral bicarbonate transporter Slc4a3, both expressed specifically in horizontal cells, is necessary for horizontal cell-to-cone feedback. Pharmacological blockage of bicarbonate transporters and buffering pH also abolished the feedback but blocking sodium-proton exchangers and GABA receptors did not. Our work suggests an unconventional mechanism of feedback at the first visual synapse: changes in horizontal cell voltage modulate bicarbonate transport to the cell, via Slc4a5, which leads to the modulation of feedback to cones.
Collapse
Affiliation(s)
- Rei Morikawa
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland
| | - Tiago M Rodrigues
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland
| | | | - Cameron S Cowan
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland
| | - Sarah Nadeau
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland
| | - Alexandra Graff-Meyer
- Facility for Advanced Imaging and Microscopy, Friedrich Miescher Institute for Biomedical Research, 4056 Basel, Switzerland
| | | | | | - Josephine Jüttner
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland
| | - Botond Roska
- Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland.
| |
Collapse
|
3
|
Zlomuzica A, Plank L, Dere E. A new path to mental disorders: Through gap junction channels and hemichannels. Neurosci Biobehav Rev 2022; 142:104877. [PMID: 36116574 DOI: 10.1016/j.neubiorev.2022.104877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/20/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022]
Abstract
Behavioral disturbances related to emotional regulation, reward processing, cognition, sleep-wake regulation and activity/movement represent core symptoms of most common mental disorders. Increasing empirical and theoretical evidence suggests that normal functioning of these behavioral domains relies on fine graded coordination of neural and glial networks which are maintained and modulated by intercellular gap junction channels and unapposed pannexin or connexin hemichannels. Dysfunctions in these networks might contribute to the development and maintenance of psychopathological and neurobiological features associated with mental disorders. Here we review and discuss the evidence indicating a prominent role of gap junction channel and hemichannel dysfunction in core symptoms of mental disorders. We further discuss how the increasing knowledge on intercellular gap junction channels and unapposed pannexin or connexin hemichannels in the brain might lead to deeper mechanistic insight in common mental disorders and to the development of novel treatment approaches. We further attempt to exemplify what type of future research on this topic could be integrated into multidimensional approaches to understand and cure mental disorders.
Collapse
Affiliation(s)
- Armin Zlomuzica
- Department of Behavioral and Clinical Neuroscience, Ruhr-University Bochum (RUB), Massenbergstraße 9-13, D-44787 Bochum, Germany.
| | - Laurin Plank
- Department of Behavioral and Clinical Neuroscience, Ruhr-University Bochum (RUB), Massenbergstraße 9-13, D-44787 Bochum, Germany
| | - Ekrem Dere
- Department of Behavioral and Clinical Neuroscience, Ruhr-University Bochum (RUB), Massenbergstraße 9-13, D-44787 Bochum, Germany; Sorbonne Université. Institut de Biologie Paris-Seine, (IBPS), Département UMR 8256: Adaptation Biologique et Vieillissement, UFR des Sciences de la Vie, Campus Pierre et Marie Curie, Bâtiment B, 9 quai Saint Bernard, F-75005 Paris, France.
| |
Collapse
|
4
|
Lukowicz-Bedford RM, Farnsworth DR, Miller AC. Connexinplexity: the spatial and temporal expression of connexin genes during vertebrate organogenesis. G3 (BETHESDA, MD.) 2022; 12:jkac062. [PMID: 35325106 PMCID: PMC9073686 DOI: 10.1093/g3journal/jkac062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/24/2022] [Indexed: 11/28/2022]
Abstract
Animal development requires coordinated communication between cells. The Connexin family of proteins is a major contributor to intercellular communication in vertebrates by forming gap junction channels that facilitate the movement of ions, small molecules, and metabolites between cells. Additionally, individual hemichannels can provide a conduit to the extracellular space for paracrine and autocrine signaling. Connexin-mediated communication is widely used in epithelial, neural, and vascular development and homeostasis, and most tissues likely use this form of communication. In fact, Connexin disruptions are of major clinical significance contributing to disorders developing from all major germ layers. Despite the fact that Connexins serve as an essential mode of cellular communication, the temporal and cell-type-specific expression patterns of connexin genes remain unknown in vertebrates. A major challenge is the large and complex connexin gene family. To overcome this barrier, we determined the expression of all connexins in zebrafish using single-cell RNA-sequencing of entire animals across several stages of organogenesis. Our analysis of expression patterns has revealed that few connexins are broadly expressed, but rather, most are expressed in tissue- or cell-type-specific patterns. Additionally, most tissues possess a unique combinatorial signature of connexin expression with dynamic temporal changes across the organism, tissue, and cell. Our analysis has identified new patterns for well-known connexins and assigned spatial and temporal expression to genes with no-existing information. We provide a field guide relating zebrafish and human connexin genes as a critical step toward understanding how Connexins contribute to cellular communication and development throughout vertebrate organogenesis.
Collapse
Affiliation(s)
| | - Dylan R Farnsworth
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Adam C Miller
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
5
|
Li Q, Zhu H, Fan M, Sun J, Reinach PS, Wang Y, Qu J, Zhou X, Zhao F. Form-deprivation myopia downregulates calcium levels in retinal horizontal cells in mice. Exp Eye Res 2022; 218:109018. [PMID: 35240197 DOI: 10.1016/j.exer.2022.109018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/14/2022] [Accepted: 02/25/2022] [Indexed: 11/25/2022]
Abstract
The process of eye axis lengthening in myopic eyes is regulated by multiple mechanisms in the retina, and horizontal cells (HCs) are an essential interneuron in the visual regulatory system. Wherein intracellular Ca2+ plays an important role in the events involved in the regulatory role of HCs in the retinal neural network. It is unknown if intracellular Ca2+ regulation in HCs mediates changes in the retinal neural network during myopia progression. We describe here a novel calcium fluorescence indicator system that monitors HCs' intracellular Ca2+ levels during form-deprivation myopia (FDM) in mice. AAV injection of GCaMP6s, as a protein calcium sensor, into a Gja10-Cre mouse monitored the changes in Ca2+signaling in HC that accompany FDM progression in mice. An alternative Gja10-Cre/Ai96-GCaMP6s mouse model was created by cross mating Gja10-Cre with Ai96 mice. Immunofluorescence imaging and live imaging of the retinal cells verified the identity of these animal models. Changes in retinal horizontal cellular Ca2+ levels were resolved during FDM development. The numbers of GCaMP6s and the proportion of HCs were tracked based on profiling changes in GCaMP6s+calbindin+/calbindin+ coimmunostaining patterns. They significantly decreased more after either two days (P < 0.01) or two weeks (P < 0.001) in form deprived eyes than in the untreated fellow eyes. These decreases in their proportion reached significance only in the retinal central region rather than also in the retinal periphery. A novel approach employing a GCaMP6s mouse model was developed that may ultimately clarify if HCs mediate Ca2+ signals that contribute to controlling FDM progression in mice. The results indicate so far that FDM progression is associated with declines in HC Ca2+ signaling activity.
Collapse
Affiliation(s)
- Qihang Li
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - He Zhu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Miaomiao Fan
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Jing Sun
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Peter S Reinach
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Yuhan Wang
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Jia Qu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China; Research Unit of Myopia Basic Research and Clinical Prevention and Control, Chinese Academy of Medical Sciences (2019RU025), Wenzhou, Zhejiang, China; Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | - Xiangtian Zhou
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China; Research Unit of Myopia Basic Research and Clinical Prevention and Control, Chinese Academy of Medical Sciences (2019RU025), Wenzhou, Zhejiang, China; Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China.
| | - Fuxin Zhao
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China.
| |
Collapse
|
6
|
Ding YQ, Qi JG. Sensory root demyelination: Transforming touch into pain. Glia 2021; 70:397-413. [PMID: 34549463 DOI: 10.1002/glia.24097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 11/12/2022]
Abstract
The normal feeling of touch is vital for nearly every aspect of our daily life. However, touching is not always felt as touch, but also abnormally as pain under numerous diseased conditions. For either mechanistic understanding of the faithful feeling of touch or clinical management of chronic pain, there is an essential need to thoroughly dissect the neuropathological changes that lead to painful touch or tactile allodynia and their corresponding cellular and molecular underpinnings. In recent years, we have seen remarkable progress in our understanding of the neural circuits for painful touch, with an increasing emphasis on the upstream roles of non-neuronal cells. As a highly specialized form of axon ensheathment by glial cells in jawed vertebrates, myelin sheaths not only mediate their outstanding neural functions via saltatory impulse propagation of temporal and spatial precision, but also support long-term neuronal/axonal integrity via metabolic and neurotrophic coupling. Therefore, myelinopathies have been implicated in diverse neuropsychiatric diseases, which are traditionally recognized as a result of the dysfunctions of neural circuits. However, whether myelinopathies can transform touch into pain remains a long-standing question. By summarizing and reframing the fragmentary but accumulating evidence so far, the present review indicates that sensory root demyelination represents a hitherto underappreciated neuropathological change for most neuropathic conditions of painful touch and offers an insightful window into faithful tactile sensation as well as a potential therapeutic target for intractable painful touch.
Collapse
Affiliation(s)
- You-Quan Ding
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jian-Guo Qi
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Baer SM, Chang S, Crook SM, Gardner CL, Jones JR, Ringhofer C, Nelson RF. A multiscale continuum model of the vertebrate outer retina: The temporal dynamics of background-induced flicker enhancement. J Theor Biol 2021; 525:110763. [PMID: 34000285 PMCID: PMC11385586 DOI: 10.1016/j.jtbi.2021.110763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 04/28/2021] [Accepted: 05/07/2021] [Indexed: 11/25/2022]
Abstract
The retina is a part of the central nervous system that is accessible, well documented, and studied by researchers spanning the clinical, experimental, and theoretical sciences. Here, we mathematically model the subcircuits of the outer plexiform layer of the retina on two spatial scales: that of an individual synapse and that of the scale of the receptive field (hundreds to thousands of synapses). To this end we formulate a continuum spine model (a partial differential equation system) that incorporates the horizontal cell syncytium and its numerous processes (spines) within cone pedicles. With this multiscale modeling approach, detailed biophysical mechanisms at the synaptic level are retained while scaling up to the receptive field level. As an example of its utility, the model is applied to study background-induced flicker enhancement in which the onset of a dim background enhances the center flicker response of horizontal cells. Simulation results, in comparison with flicker enhancement data for square, slit, and disk test regions, suggest that feedback mechanisms that are voltage-axis modulators of cone calcium channels (for example, ephaptic and/or pH feedback) are robust in capturing the temporal dynamics of background-induced flicker enhancement. The value and potential of this continuum spine approach is that it provides a framework for mathematically modeling the input-output properties of the entire receptive field of the outer retina while implementing the latest models for transmission mechanisms at the synaptic level.
Collapse
Affiliation(s)
- Steven M Baer
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States.
| | - Shaojie Chang
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States; The High School Affiliated to Beijing Normal University, Beijing 100052, PR China
| | - Sharon M Crook
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States
| | - Carl L Gardner
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States
| | - Jeremiah R Jones
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States
| | - Christian Ringhofer
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85287, United States
| | - Ralph F Nelson
- Neural Circuits Unit, Basic Neuroscience Program, NINDS, NIH, Bethesda, MD 20892, United States
| |
Collapse
|
8
|
Vila A, Shihabeddin E, Zhang Z, Santhanam A, Ribelayga CP, O’Brien J. Synaptic Scaffolds, Ion Channels and Polyamines in Mouse Photoreceptor Synapses: Anatomy of a Signaling Complex. Front Cell Neurosci 2021; 15:667046. [PMID: 34393723 PMCID: PMC8356055 DOI: 10.3389/fncel.2021.667046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/05/2021] [Indexed: 12/29/2022] Open
Abstract
Synaptic signaling complexes are held together by scaffold proteins, each of which is selectively capable of interacting with a number of other proteins. In previous studies of rabbit retina, we found Synapse-Associated Protein-102 (SAP102) and Channel Associated Protein of Synapse-110 (Chapsyn110) selectively localized in the tips of horizontal cell processes at contacts with rod and cone photoreceptors, along with several interacting ion channels. We have examined the equivalent suites of proteins in mouse retina and found similarities and differences. In the mouse retina we identified Chapsyn110 as the scaffold selectively localized in the tips of horizontal cells contacting photoreceptors, with Sap102 more diffusely present. As in rabbit, the inward rectifier potassium channel Kir2.1 was present with Chapsyn110 on the tips of horizontal cell dendrites within photoreceptor invaginations, where it could provide a hyperpolarization-activated current that could contribute to ephaptic signaling in the photoreceptor synapses. Pannexin 1 and Pannexin 2, thought to play a role in ephaptic and/or pH mediated signaling, were present in the outer plexiform layer, but likely not in the horizontal cells. Polyamines regulate many ion channels and control the degree of rectification of Kir2.1 by imposing a voltage-dependent block. During the day polyamine immunolabeling was unexpectedly high in photoreceptor terminals compared to other areas of the retina. This content was significantly lower at night, when polyamine content was predominantly in Müller glia, indicating daily rhythms of polyamine content. Both rod and cone terminals displayed the same rhythm. While polyamine content was not prominent in horizontal cells, if polyamines are released, they may regulate the activity of Kir2.1 channels located in the tips of HCs. The rhythmic change in polyamine content of photoreceptor terminals suggests that a daily rhythm tunes the behavior of suites of ion channels within the photoreceptor synapses.
Collapse
Affiliation(s)
- Alejandro Vila
- Richard S. Ruiz M.D. Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, TX, United States
- MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Eyad Shihabeddin
- Richard S. Ruiz M.D. Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, TX, United States
- MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Zhijing Zhang
- Richard S. Ruiz M.D. Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Abirami Santhanam
- Richard S. Ruiz M.D. Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Christophe P. Ribelayga
- Richard S. Ruiz M.D. Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, TX, United States
- MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - John O’Brien
- Richard S. Ruiz M.D. Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, TX, United States
- MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
9
|
Natha CM, Vemulapalli V, Fiori MC, Chang CWT, Altenberg GA. Connexin hemichannel inhibitors with a focus on aminoglycosides. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166115. [PMID: 33711451 DOI: 10.1016/j.bbadis.2021.166115] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/06/2021] [Accepted: 02/22/2021] [Indexed: 12/31/2022]
Abstract
Connexins are membrane proteins involved directly in cell-to-cell communication through the formation of gap-junctional channels. These channels result from the head-to-head docking of two hemichannels, one from each of two adjacent cells. Undocked hemichannels are also present at the plasma membrane where they mediate the efflux of molecules that participate in autocrine and paracrine signaling, but abnormal increase in hemichannel activity can lead to cell damage in disorders such as cardiac infarct, stroke, deafness, cataracts, and skin diseases. For this reason, connexin hemichannels have emerged as a valid therapeutic target. Know small molecule hemichannel inhibitors are not ideal leads for the development of better drugs for clinical use because they are not specific and/or have toxic effects. Newer inhibitors are more selective and include connexin mimetic peptides, anti-connexin antibodies and drugs that reduce connexin expression such as antisense oligonucleotides. Re-purposed drugs and their derivatives are also promising because of the significant experience with their clinical use. Among these, aminoglycoside antibiotics have been identified as inhibitors of connexin hemichannels that do not inhibit gap-junctional channels. In this review, we discuss connexin hemichannels and their inhibitors, with a focus on aminoglycoside antibiotics and derivatives of kanamycin A that inhibit connexin hemichannels, but do not have antibiotic effect.
Collapse
Affiliation(s)
- Cristina M Natha
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Varun Vemulapalli
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Mariana C Fiori
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Cheng-Wei T Chang
- Department of Chemistry and Biochemistry, Utah State University, Logan, UT, USA
| | - Guillermo A Altenberg
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
10
|
Barnes S, Grove JCR, McHugh CF, Hirano AA, Brecha NC. Horizontal Cell Feedback to Cone Photoreceptors in Mammalian Retina: Novel Insights From the GABA-pH Hybrid Model. Front Cell Neurosci 2020; 14:595064. [PMID: 33328894 PMCID: PMC7672006 DOI: 10.3389/fncel.2020.595064] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 09/24/2020] [Indexed: 01/20/2023] Open
Abstract
How neurons in the eye feed signals back to photoreceptors to optimize sensitivity to patterns of light appears to be mediated by one or more unconventional mechanisms. Via these mechanisms, horizontal cells control photoreceptor synaptic gain and enhance key aspects of temporal and spatial center-surround receptive field antagonism. After the transduction of light energy into an electrical signal in photoreceptors, the next key task in visual processing is the transmission of an optimized signal to the follower neurons in the retina. For this to happen, the release of the excitatory neurotransmitter glutamate from photoreceptors is carefully regulated via horizontal cell feedback, which acts as a thermostat to keep the synaptic transmission in an optimal range during changes to light patterns and intensities. Novel findings of a recently described model that casts a classical neurotransmitter system together with ion transport mechanisms to adjust the alkaline milieu outside the synapse are reviewed. This novel inter-neuronal messaging system carries feedback signals using two separate, but interwoven regulated systems. The complex interplay between these two signaling modalities, creating synaptic modulation-at-a-distance, has obscured it’s being defined. The foundations of our understanding of the feedback mechanism from horizontal cells to photoreceptors have been long established: Horizontal cells have broad receptive fields, suitable for providing surround inhibition, their membrane potential, a function of stimulus intensity and size, regulates inhibition of photoreceptor voltage-gated Ca2+ channels, and strong artificial pH buffering eliminates this action. This review compares and contrasts models of how these foundations are linked, focusing on a recent report in mammals that shows tonic horizontal cell release of GABA activating Cl− and HCO3− permeable GABA autoreceptors. The membrane potential of horizontal cells provides the driving force for GABAR-mediated HCO3− efflux, alkalinizing the cleft when horizontal cells are hyperpolarized by light or adding to their depolarization in darkness and contributing to cleft acidification via NHE-mediated H+ efflux. This model challenges interpretations of earlier studies that were considered to rule out a role for GABA in feedback to cones.
Collapse
Affiliation(s)
- Steven Barnes
- Doheny Eye Institute, Los Angeles, CA, United States.,Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - James C R Grove
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, United States
| | | | - Arlene A Hirano
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Nicholas C Brecha
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
Controlling Horizontal Cell-Mediated Lateral Inhibition in Transgenic Zebrafish Retina with Chemogenetic Tools. eNeuro 2020; 7:ENEURO.0022-20.2020. [PMID: 33060180 PMCID: PMC7665903 DOI: 10.1523/eneuro.0022-20.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 08/11/2020] [Accepted: 08/28/2020] [Indexed: 12/03/2022] Open
Abstract
Horizontal cells (HCs) form reciprocal synapses with rod and cone photoreceptors, an arrangement that underlies lateral inhibition in the retina. HCs send negative and positive feedback signals to photoreceptors, but how HCs initiate these signals remains unclear. Unfortunately, because HCs have no unique neurotransmitter receptors, there are no pharmacological treatments for perturbing membrane potential specifically in HCs. Here we use transgenic zebrafish whose HCs express alien receptors, enabling cell-type-specific control by cognate alien agonists. To depolarize HCs, we used the Phe-Met-Arg-Phe-amide (FMRFamide)-gated Na+ channel (FaNaC) activated by the invertebrate neuropeptide FMRFamide. To hyperpolarize HCs we used a pharmacologically selective actuator module (PSAM)-glycine receptor (GlyR), an engineered Cl– selective channel activated by a synthetic agonist. Expression of FaNaC or PSAM-GlyR was restricted to HCs with the cell-type selective promoter for connexin-55.5. We assessed HC-feedback control of photoreceptor synapses in three ways. First, we measured presynaptic exocytosis from photoreceptor terminals using the fluorescent dye FM1-43. Second, we measured the electroretinogram (ERG) b-wave, a signal generated by postsynaptic responses. Third, we used Ca2+ imaging in retinal ganglion cells (RGCs) expressing the Ca2+ indicator GCaMP6. Addition of FMRFamide significantly decreased FM1-43 destaining in darkness, whereas the addition of PSAM-GlyR significantly increased it. However, both agonists decreased the light-elicited ERG b-wave and eliminated surround inhibition of the Ca2+ response of RGCs. Taken together, our findings show that chemogenetic tools can selectively manipulate negative feedback from HCs, providing a platform for understanding its mechanism and helping to elucidate its functional roles in visual information processing at a succession of downstream stages.
Collapse
|
12
|
Dere D, Zlomuzica A, Dere E. Channels to consciousness: a possible role of gap junctions in consciousness. Rev Neurosci 2020; 32:/j/revneuro.ahead-of-print/revneuro-2020-0012/revneuro-2020-0012.xml. [PMID: 32853172 DOI: 10.1515/revneuro-2020-0012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022]
Abstract
The neurophysiological basis of consciousness is still unknown and one of the most challenging questions in the field of neuroscience and related disciplines. We propose that consciousness is characterized by the maintenance of mental representations of internal and external stimuli for the execution of cognitive operations. Consciousness cannot exist without working memory, and it is likely that consciousness and working memory share the same neural substrates. Here, we present a novel psychological and neurophysiological framework that explains the role of consciousness for cognition, adaptive behavior, and everyday life. A hypothetical architecture of consciousness is presented that is organized as a system of operation and storage units named platforms that are controlled by a consciousness center (central executive/online platform). Platforms maintain mental representations or contents, are entrusted with different executive functions, and operate at different levels of consciousness. The model includes conscious-mode central executive/online and mental time travel platforms and semiconscious steady-state and preconscious standby platforms. Mental representations or contents are represented by neural circuits and their support cells (astrocytes, oligodendrocytes, etc.) and become conscious when neural circuits reverberate, that is, fire sequentially and continuously with relative synchronicity. Reverberatory activity in neural circuits may be initiated and maintained by pacemaker cells/neural circuit pulsars, enhanced electronic coupling via gap junctions, and unapposed hemichannel opening. The central executive/online platform controls which mental representations or contents should become conscious by recruiting pacemaker cells/neural network pulsars, the opening of hemichannels, and promoting enhanced neural circuit coupling via gap junctions.
Collapse
Affiliation(s)
- Dorothea Dere
- Département UMR 8256 Adaptation Biologique et Vieillissement, Sorbonne Université, Institut de Biologie Paris-Seine, (IBPS), UFR des Sciences de la Vie, Campus Pierre et Marie Curie, Bâtiment B, 9 quai Saint Bernard, F-75005 Paris Cedex, France
| | - Armin Zlomuzica
- Faculty of Psychology, Behavioral and Clinical Neuroscience, University of Bochum, Massenbergstraße 9-13, D-44787 Bochum, Germany
| | - Ekrem Dere
- Département UMR 8256 Adaptation Biologique et Vieillissement, Sorbonne Université, Institut de Biologie Paris-Seine, (IBPS), UFR des Sciences de la Vie, Campus Pierre et Marie Curie, Bâtiment B, 9 quai Saint Bernard, F-75005 Paris Cedex, France
| |
Collapse
|
13
|
Furukawa T, Ueno A, Omori Y. Molecular mechanisms underlying selective synapse formation of vertebrate retinal photoreceptor cells. Cell Mol Life Sci 2020; 77:1251-1266. [PMID: 31586239 PMCID: PMC11105113 DOI: 10.1007/s00018-019-03324-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/21/2019] [Accepted: 09/25/2019] [Indexed: 11/29/2022]
Abstract
In vertebrate central nervous systems (CNSs), highly diverse neurons are selectively connected via synapses, which are essential for building an intricate neural network. The vertebrate retina is part of the CNS and is comprised of a distinct laminar organization, which serves as a good model system to study developmental synapse formation mechanisms. In the retina outer plexiform layer, rods and cones, two types of photoreceptor cells, elaborate selective synaptic contacts with ON- and/or OFF-bipolar cell terminals as well as with horizontal cell terminals. In the mouse retina, three photoreceptor subtypes and at least 15 bipolar subtypes exist. Previous and recent studies have significantly progressed our understanding of how selective synapse formation, between specific subtypes of photoreceptor and bipolar cells, is designed at the molecular level. In the ON pathway, photoreceptor-derived secreted and transmembrane proteins directly interact in trans with the GRM6 (mGluR6) complex, which is localized to ON-bipolar cell dendritic terminals, leading to selective synapse formation. Here, we review our current understanding of the key factors and mechanisms underlying selective synapse formation of photoreceptor cells with bipolar and horizontal cells in the retina. In addition, we describe how defects/mutations of the molecules involved in photoreceptor synapse formation are associated with human retinal diseases and visual disorders.
Collapse
Affiliation(s)
- Takahisa Furukawa
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Akiko Ueno
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshihiro Omori
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
14
|
Abstract
At the first retinal synapse, horizontal cells (HCs) contact both photoreceptor terminals and bipolar cell dendrites, modulating information transfer between these two cell types to enhance spatial contrast and mediate color opponency. The synaptic mechanisms through which these modulations occur are still debated. The initial hypothesis of a GABAergic feedback from HCs to cones has been challenged by pharmacological inconsistencies. Surround antagonism has been demonstrated to occur via a modulation of cone calcium channels through ephaptic signaling and pH changes in the synaptic cleft. GABAergic transmission between HCs and cones has been reported in some lower vertebrates, like the turtle and tiger salamander. In these reports, it was revealed that GABA is released from HCs through reverse transport and target GABA receptors are located at the cone terminals. In mammalian retinas, there is growing evidence that HCs can release GABA through conventional vesicular transmission, acting both on autaptic GABA receptors and on receptors expressed at the dendritic tips of the bipolar cells. The presence of GABA receptors on mammalian cone terminals remains equivocal. Here, we looked specifically for functional GABA receptors in mouse photoreceptors by recording in the whole-cell or amphotericin/gramicidin-perforated patch clamp configurations. Cones could be differentiated from rods through morphological criteria. Local GABA applications evoked a Cl- current in cones but not in rods. It was blocked by the GABAA receptor antagonist bicuculline methiodide and unaffected by the GABAC receptor antagonist TPMPA [(1,2,5,6-tetrahydropyridin-4-yl)methylphosphinic acid]. The voltage dependency of the current amplitude was as expected from a direct action of GABA on cone pedicles but not from an indirect modulation of cone currents following the activation of the GABA receptors of HCs. This supports a direct role of GABA released from HCs in the control of cone activity in the mouse retina.
Collapse
|
15
|
Analysis of zebrafish cryptochrome 2 and 4 expression in UV cone photoreceptors. Gene Expr Patterns 2020; 35:119100. [DOI: 10.1016/j.gep.2020.119100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 12/30/2019] [Accepted: 02/12/2020] [Indexed: 01/11/2023]
|
16
|
Thoreson WB, Dacey DM. Diverse Cell Types, Circuits, and Mechanisms for Color Vision in the Vertebrate Retina. Physiol Rev 2019; 99:1527-1573. [PMID: 31140374 PMCID: PMC6689740 DOI: 10.1152/physrev.00027.2018] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 01/13/2023] Open
Abstract
Synaptic interactions to extract information about wavelength, and thus color, begin in the vertebrate retina with three classes of light-sensitive cells: rod photoreceptors at low light levels, multiple types of cone photoreceptors that vary in spectral sensitivity, and intrinsically photosensitive ganglion cells that contain the photopigment melanopsin. When isolated from its neighbors, a photoreceptor confounds photon flux with wavelength and so by itself provides no information about color. The retina has evolved elaborate color opponent circuitry for extracting wavelength information by comparing the activities of different photoreceptor types broadly tuned to different parts of the visible spectrum. We review studies concerning the circuit mechanisms mediating opponent interactions in a range of species, from tetrachromatic fish with diverse color opponent cell types to common dichromatic mammals where cone opponency is restricted to a subset of specialized circuits. Distinct among mammals, primates have reinvented trichromatic color vision using novel strategies to incorporate evolution of an additional photopigment gene into the foveal structure and circuitry that supports high-resolution vision. Color vision is absent at scotopic light levels when only rods are active, but rods interact with cone signals to influence color perception at mesopic light levels. Recent evidence suggests melanopsin-mediated signals, which have been identified as a substrate for setting circadian rhythms, may also influence color perception. We consider circuits that may mediate these interactions. While cone opponency is a relatively simple neural computation, it has been implemented in vertebrates by diverse neural mechanisms that are not yet fully understood.
Collapse
Affiliation(s)
- Wallace B Thoreson
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha, Nebraska ; and Department of Biological Structure, Washington National Primate Research Center, University of Washington , Seattle, Washington
| | - Dennis M Dacey
- Department of Ophthalmology and Visual Sciences, Truhlsen Eye Institute, University of Nebraska Medical Center , Omaha, Nebraska ; and Department of Biological Structure, Washington National Primate Research Center, University of Washington , Seattle, Washington
| |
Collapse
|
17
|
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
|
18
|
An Alternative Splice Variant of Zebrafish Cx52.6 is Expressed in Retinal Horizontal Cells. Neuroscience 2018; 388:191-202. [PMID: 30048782 DOI: 10.1016/j.neuroscience.2018.07.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/27/2018] [Accepted: 07/16/2018] [Indexed: 11/22/2022]
Abstract
Retinal horizontal cells (HCs) are inhibitory neurons, which modulate the transmission of light-elicited signals from photoreceptors to bipolar cells in the outer retina. HCs of the same physiological type are extensively coupled via gap junctions. In the zebrafish retina, the population of HCs comprises up to four morphologically distinct subtypes. Four different connexins (Cx52.6, Cx52.7, Cx52.9 and Cx55.5) were detected in these cells with overlapping expression patterns. In this study, we show that Cx52.6 is alternatively spliced in the retina, resulting in an additional isoform, designated as Cx53.4, which differs from the originally described Cx52.6 only by the final C-terminal peptide (12 vs. 4 aa). Further protein sequence alignments revealed that Cx53.4 represents the counterpart of alternatively spliced mouse Cx57 and human Cx62. RT-PCR analyses of mRNA expression in different adult zebrafish tissues showed that Cx53.4 is expressed exclusively in the retina. The localization of Cx53.4 protein within the retina was analyzed using a specific antibody. Immunofluorescence analyses demonstrated that the expression of Cx53.4 is restricted to HCs of all four subtypes. Further, immunoelectron microscopy confirmed the presence of Cx53.4 in gap junctions between HC dendrites and between their axon terminals.
Collapse
|
19
|
Petralia RS, Wang YX, Mattson MP, Yao PJ. Invaginating Structures in Mammalian Synapses. Front Synaptic Neurosci 2018; 10:4. [PMID: 29674962 PMCID: PMC5895750 DOI: 10.3389/fnsyn.2018.00004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/19/2018] [Indexed: 12/26/2022] Open
Abstract
Invaginating structures at chemical synapses in the mammalian nervous system exist in presynaptic axon terminals, postsynaptic spines or dendrites, and glial processes. These invaginating structures can be divided into three categories. The first category includes slender protrusions invaginating into axonal terminals, postsynaptic spines, or glial processes. Best known examples of this category are spinules extending from postsynaptic spines into presynaptic terminals in forebrain synapses. Another example of this category are protrusions from inhibitory presynaptic terminals invaginating into postsynaptic neuronal somas. Regardless of the direction and location, the invaginating structures of the first category do not have synaptic active zones within the invagination. The second category includes postsynaptic spines invaginating into presynaptic terminals, whereas the third category includes presynaptic terminals invaginating into postsynaptic spines or dendrites. Unlike the first category, the second and third categories have active zones within the invagination. An example of the second category are mossy terminal synapses of the hippocampal CA3 region, in which enlarged spine-like structures invaginate partly or entirely into mossy terminals. An example of the third category is the neuromuscular junction (NMJ) where substantial invaginations of the presynaptic terminals invaginate into the muscle fibers. In the retina, rod and cone synapses have invaginating processes from horizontal and bipolar cells. Because horizontal cells act both as post and presynaptic structures, their invaginating processes represent both the second and third category. These invaginating structures likely play broad yet specialized roles in modulating neuronal cell signaling.
Collapse
Affiliation(s)
| | - Ya-Xian Wang
- Advanced Imaging Core, NIDCD/NIH, Bethesda, MD, United States
| | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging, Intramural Research Program, Baltimore, MD, United States
| | - Pamela J Yao
- Laboratory of Neurosciences, National Institute on Aging, Intramural Research Program, Baltimore, MD, United States
| |
Collapse
|
20
|
Rhett JM, Yeh ES. The Potential for Connexin Hemichannels to Drive Breast Cancer Progression through Regulation of the Inflammatory Response. Int J Mol Sci 2018; 19:ijms19041043. [PMID: 29601539 PMCID: PMC5979453 DOI: 10.3390/ijms19041043] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/12/2022] Open
Abstract
Over the past few decades, connexin hemichannels have become recognized as major players in modulating the inflammatory response. Chronic inflammation is documented to promote tumorigenesis and is a critical component of tumor progression. Furthermore, inflammation is strongly linked to angiogenesis, immunotolerance, invasiveness, metastasis, and resistance in breast cancers. In this review, the literature on the role of connexin hemichannels in inflammation is summarized, and the potential role for hemichannel-mediated inflammation in driving breast cancer progression is discussed. Lastly, the potential for connexin-based therapeutics to modulate the inflammatory component of the tumor microenvironment as an avenue for the treatment of breast cancer is also discussed.
Collapse
Affiliation(s)
- J Matthew Rhett
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29412, USA.
| | - Elizabeth S Yeh
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29412, USA.
| |
Collapse
|
21
|
Cenedese V, de Graaff W, Csikós T, Poovayya M, Zoidl G, Kamermans M. Pannexin 1 Is Critically Involved in Feedback from Horizontal Cells to Cones. Front Mol Neurosci 2017; 10:403. [PMID: 29375296 PMCID: PMC5770619 DOI: 10.3389/fnmol.2017.00403] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/20/2017] [Indexed: 11/18/2022] Open
Abstract
Retinal horizontal cells (HCs) feed back negatively to cone photoreceptors and in that way generate the center/surround organization of bipolar cell receptive fields. The mechanism by which HCs inhibit photoreceptors is a matter of debate. General consensus exists that horizontal cell activity leads to the modulation of the cone Ca-current. This modulation has two components, one fast and the other slow. Several mechanisms for this modulation have been proposed: a fast ephaptic mechanism, and a slow pH mediated mechanism. Here we test the hypothesis that the slow negative feedback signal from HCs to cones is mediated by Panx1 channels expressed at the tips of the dendrites of horizontal cell. We generated zebrafish lacking Panx1 and found that the slow component of the feedback signal was strongly reduced in the mutants showing that Panx1 channels are a fundamental part of the negative feedback pathway from HCs to cones.
Collapse
Affiliation(s)
- Valentina Cenedese
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Wim de Graaff
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Tamás Csikós
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Mitali Poovayya
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Georg Zoidl
- Department of Biology, York University, Toronto, ON, Canada
| | - Maarten Kamermans
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, Netherlands.,Department of Biomedical Physics and Biomedical Optics, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
22
|
Kim Y, Griffin JM, Nor MNM, Zhang J, Freestone PS, Danesh-Meyer HV, Rupenthal ID, Acosta M, Nicholson LFB, O'Carroll SJ, Green CR. Tonabersat Prevents Inflammatory Damage in the Central Nervous System by Blocking Connexin43 Hemichannels. Neurotherapeutics 2017; 14:1148-1165. [PMID: 28560708 PMCID: PMC5722754 DOI: 10.1007/s13311-017-0536-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The cis benzopyran compound tonabersat (SB-220453) has previously been reported to inhibit connexin26 expression in the brain by attenuating the p38-mitogen-activated protein kinase pathway. We show here that tonabersat directly inhibits connexin43 hemichannel opening. Connexin43 hemichannels have been called "pathological pores" based upon their role in secondary lesion spread, edema, inflammation, and neuronal loss following central nervous system injuries, as well as in chronic inflammatory disease. Both connexin43 hemichannels and pannexin channels released adenosine triphosphate (ATP) during ischemia in an in vitro ischemia model, but only connexin43 hemichannels contributed to ATP release during reperfusion. Tonabersat inhibited connexin43 hemichannel-mediated ATP release during both ischemia and reperfusion phases, with direct channel block confirmed using electrophysiology. Tonabersat also reduced connexin43 gap junction coupling in vitro, but only at higher concentrations, with junctional plaques internalized and degraded via the lysosomal pathway. Systemic delivery of tonabersat in a rat bright-light retinal damage model (a model for dry age-related macular degeneration) resulted in significantly improved functional outcomes assessed using electroretinography. Tonabersat also prevented thinning of the retina, especially the outer nuclear layer and choroid, assessed using optical coherence tomography. We conclude that tonabersat, already given orally to over 1000 humans in clinical trials (as a potential treatment for, and prophylactic treatment of, migraine because it was thought to inhibit cortical spreading depression), is a connexin hemichannel inhibitor and may have the potential to be a novel treatment of central nervous system injury and chronic neuroinflammatory disease.
Collapse
Affiliation(s)
- Yeri Kim
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
- New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
| | - Jarred M Griffin
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
| | - Mohd N Mat Nor
- New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
- School of Optometry and Vision Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
- Faculty of Medicine, University Sultan Zainal Abidin, Kuala Terengganu, Malaysia
| | - Jie Zhang
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
- New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
| | - Peter S Freestone
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
| | - Helen V Danesh-Meyer
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
- New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
| | - Ilva D Rupenthal
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
- New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
- Buchanan Ocular Therapeutics Unit, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
| | - Monica Acosta
- New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
- School of Optometry and Vision Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
| | - Louise F B Nicholson
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
| | - Simon J O'Carroll
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand
| | - Colin R Green
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand.
- New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, 1142, New Zealand.
| |
Collapse
|
23
|
Petralia RS, Wang YX, Mattson MP, Yao PJ. Invaginating Presynaptic Terminals in Neuromuscular Junctions, Photoreceptor Terminals, and Other Synapses of Animals. Neuromolecular Med 2017; 19:193-240. [PMID: 28612182 PMCID: PMC6518423 DOI: 10.1007/s12017-017-8445-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 06/01/2017] [Indexed: 10/19/2022]
Abstract
Typically, presynaptic terminals form a synapse directly on the surface of postsynaptic processes such as dendrite shafts and spines. However, some presynaptic terminals invaginate-entirely or partially-into postsynaptic processes. We survey these invaginating presynaptic terminals in all animals and describe several examples from the central nervous system, including giant fiber systems in invertebrates, and cup-shaped spines, electroreceptor synapses, and some specialized auditory and vestibular nerve terminals in vertebrates. We then examine mechanoreceptors and photoreceptors, concentrating on the complex of pre- and postsynaptic processes found in basal invaginations of the cell. We discuss in detail the role of vertebrate invaginating horizontal cell processes in both chemical and electrical feedback mechanisms. We also discuss the common presence of indenting or invaginating terminals in neuromuscular junctions on muscles of most kinds of animals, and especially discuss those of Drosophila and vertebrates. Finally, we consider broad questions about the advantages of possessing invaginating presynaptic terminals and describe some effects of aging and disease, especially on neuromuscular junctions. We suggest that the invagination is a mechanism that can enhance both chemical and electrical interactions at the synapse.
Collapse
Affiliation(s)
- Ronald S Petralia
- Advanced Imaging Core, NIDCD/NIH, 35A Center Drive, Room 1E614, Bethesda, MD, 20892-3729, USA.
| | - Ya-Xian Wang
- Advanced Imaging Core, NIDCD/NIH, 35A Center Drive, Room 1E614, Bethesda, MD, 20892-3729, USA
| | - Mark P Mattson
- Laboratory of Neurosciences, NIA/NIH, Baltimore, MD, 21224, USA
| | - Pamela J Yao
- Laboratory of Neurosciences, NIA/NIH, Baltimore, MD, 21224, USA
| |
Collapse
|
24
|
Chaffiol A, Ishii M, Cao Y, Mangel SC. Dopamine Regulation of GABA A Receptors Contributes to Light/Dark Modulation of the ON-Cone Bipolar Cell Receptive Field Surround in the Retina. Curr Biol 2017; 27:2600-2609.e4. [PMID: 28844643 DOI: 10.1016/j.cub.2017.07.063] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 05/22/2017] [Accepted: 07/27/2017] [Indexed: 10/19/2022]
Abstract
Cone bipolar cells are interneurons that receive synaptic input from cone photoreceptor cells and provide the output of the first synaptic layer of the retina. These cells exhibit center-surround receptive fields, a prototype of lateral inhibition between neighboring sensory cells in which stimulation of the receptive field center excites the cell whereas stimulation of the surrounding region laterally inhibits the cell. This fundamental sensory coding mechanism facilitates spatial discrimination and detection of stimulus edges. However, although it is well established that the receptive field surround is strongest when ambient or background illumination is most intense, e.g., at midday, and that the surround is minimal following maintained darkness, the synaptic mechanisms that produce and modulate the surround have not been resolved. Using electrical recording of bipolar cells under experimental conditions in which the cells exhibited surround light responses, and light and electron microscopic immunocytochemistry, we show in the rabbit retina that bright-light-induced activation of dopamine D1 receptors located on ON-center cone bipolar cell dendrites increases the expression and activity of GABAA receptors on the dendrites of the cells and that surround light responses depend on endogenous GABAA receptor activation. We also show that maintained darkness and D1 receptor blockade following maintained illumination and D1 receptor activation result in minimal GABAA receptor expression and activity and greatly diminished surrounds. Modulation of the D1/GABAA receptor signaling pathway of ON-cBC dendrites by the ambient light level facilitates detection of spatial details on bright days and large dim objects on moonless nights.
Collapse
Affiliation(s)
- Antoine Chaffiol
- Department of Neuroscience, The Ohio State University College Of Medicine, Columbus, OH 43210, USA
| | - Masaaki Ishii
- Department of Neuroscience, The Ohio State University College Of Medicine, Columbus, OH 43210, USA
| | - Yu Cao
- Department of Neuroscience, The Ohio State University College Of Medicine, Columbus, OH 43210, USA
| | - Stuart C Mangel
- Department of Neuroscience, The Ohio State University College Of Medicine, Columbus, OH 43210, USA.
| |
Collapse
|
25
|
Versatile functional roles of horizontal cells in the retinal circuit. Sci Rep 2017; 7:5540. [PMID: 28717219 PMCID: PMC5514144 DOI: 10.1038/s41598-017-05543-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 05/31/2017] [Indexed: 01/13/2023] Open
Abstract
In the retinal circuit, environmental light signals are converted into electrical signals that can be decoded properly by the brain. At the first synapse of the visual system, information flow from photoreceptors to bipolar cells is modulated by horizontal cells (HCs), however, their functional contribution to retinal output and individual visual function is not fully understood. In the current study, we investigated functional roles for HCs in retinal ganglion cell (RGC) response properties and optokinetic responses by establishing a HC-depleted mouse line. We observed that HC depletion impairs the antagonistic center-surround receptive field formation of RGCs, supporting a previously reported HC function revealed by pharmacological approaches. In addition, we found that HC loss reduces both the ON and OFF response diversities of RGCs, impairs adjustment of the sensitivity to ambient light at the retinal output level, and alters spatial frequency tuning at an individual level. Taken together, our current study suggests multiple functional aspects of HCs crucial for visual processing.
Collapse
|
26
|
Lapato AS, Tiwari-Woodruff SK. Connexins and pannexins: At the junction of neuro-glial homeostasis & disease. J Neurosci Res 2017; 96:31-44. [PMID: 28580666 DOI: 10.1002/jnr.24088] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/08/2017] [Accepted: 05/01/2017] [Indexed: 12/15/2022]
Abstract
In the central nervous system (CNS), connexin (Cx)s and pannexin (Panx)s are an integral component of homeostatic neuronal excitability and synaptic plasticity. Neuronal Cx gap junctions form electrical synapses across biochemically similar GABAergic networks, allowing rapid and extensive inhibition in response to principle neuron excitation. Glial Cx gap junctions link astrocytes and oligodendrocytes in the pan-glial network that is responsible for removing excitotoxic ions and metabolites. In addition, glial gap junctions help constrain excessive excitatory activity in neurons and facilitate astrocyte Ca2+ slow wave propagation. Panxs do not form gap junctions in vivo, but Panx hemichannels participate in autocrine and paracrine gliotransmission, alongside Cx hemichannels. ATP and other gliotransmitters released by Cx and Panx hemichannels maintain physiologic glutamatergic tone by strengthening synapses and mitigating aberrant high frequency bursting. Under pathological depolarizing and inflammatory conditions, gap junctions and hemichannels become dysregulated, resulting in excessive neuronal firing and seizure. In this review, we present known contributions of Cxs and Panxs to physiologic neuronal excitation and explore how the disruption of gap junctions and hemichannels lead to abnormal glutamatergic transmission, purinergic signaling, and seizures.
Collapse
Affiliation(s)
- Andrew S Lapato
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, 92521.,Center for Glial-Neuronal Interactions, University of California Riverside, Riverside, CA, 92521
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, 92521.,Center for Glial-Neuronal Interactions, University of California Riverside, Riverside, CA, 92521.,Neuroscience Graduate Program, University of California Riverside, Riverside, CA, 92521
| |
Collapse
|
27
|
Chapot CA, Euler T, Schubert T. How do horizontal cells 'talk' to cone photoreceptors? Different levels of complexity at the cone-horizontal cell synapse. J Physiol 2017; 595:5495-5506. [PMID: 28378516 DOI: 10.1113/jp274177] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/27/2017] [Indexed: 11/08/2022] Open
Abstract
The first synapse of the retina plays a fundamental role in the visual system. Due to its importance, it is critical that it encodes information from the outside world with the greatest accuracy and precision possible. Cone photoreceptor axon terminals contain many individual synaptic sites, each represented by a presynaptic structure called a 'ribbon'. These synapses are both highly sophisticated and conserved. Each ribbon relays the light signal to one ON cone bipolar cell and several OFF cone bipolar cells, while two dendritic processes from a GABAergic interneuron, the horizontal cell, modulate the cone output via parallel feedback mechanisms. The presence of these three partners within a single synapse has raised numerous questions, and its anatomical and functional complexity is still only partially understood. However, the understanding of this synapse has recently evolved, as a consequence of progress in understanding dendritic signal processing and its role in facilitating global versus local signalling. Indeed, for the downstream retinal network, dendritic processing in horizontal cells may be essential, as they must support important functional operations such as contrast enhancement, which requires spatial averaging of the photoreceptor array, while at the same time preserving accurate spatial information. Here, we review recent progress made towards a better understanding of the cone synapse, with an emphasis on horizontal cell function, and discuss why such complexity might be necessary for early visual processing.
Collapse
Affiliation(s)
- Camille A Chapot
- Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany.,Centre for Integrative Neuroscience, University of Tübingen, 72076, Tübingen, Germany.,Graduate Training Centre of Neuroscience, University of Tübingen, 72076, Tübingen, Germany
| | - Thomas Euler
- Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany.,Centre for Integrative Neuroscience, University of Tübingen, 72076, Tübingen, Germany.,Bernstein Centre for Computational Neuroscience, University of Tübingen, 72076, Tübingen, Germany
| | - Timm Schubert
- Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany.,Centre for Integrative Neuroscience, University of Tübingen, 72076, Tübingen, Germany
| |
Collapse
|
28
|
Bargiello TA, Oh S, Tang Q, Bargiello NK, Dowd TL, Kwon T. Gating of Connexin Channels by transjunctional-voltage: Conformations and models of open and closed states. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:22-39. [PMID: 28476631 DOI: 10.1016/j.bbamem.2017.04.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/26/2017] [Accepted: 04/28/2017] [Indexed: 11/18/2022]
Abstract
Voltage is an important physiologic regulator of channels formed by the connexin gene family. Connexins are unique among ion channels in that both plasma membrane inserted hemichannels (undocked hemichannels) and intercellular channels (aggregates of which form gap junctions) have important physiological roles. The hemichannel is the fundamental unit of gap junction voltage-gating. Each hemichannel displays two distinct voltage-gating mechanisms that are primarily sensitive to a voltage gradient formed along the length of the channel pore (the transjunctional voltage) rather than sensitivity to the absolute membrane potential (Vm or Vi-o). These transjunctional voltage dependent processes have been termed Vj- or fast-gating and loop- or slow-gating. Understanding the mechanism of voltage-gating, defined as the sequence of voltage-driven transitions that connect open and closed states, first and foremost requires atomic resolution models of the end states. Although ion channels formed by connexins were among the first to be characterized structurally by electron microscopy and x-ray diffraction in the early 1980's, subsequent progress has been slow. Much of the current understanding of the structure-function relations of connexin channels is based on two crystal structures of Cx26 gap junction channels. Refinement of crystal structure by all-atom molecular dynamics and incorporation of charge changing protein modifications has resulted in an atomic model of the open state that arguably corresponds to the physiologic open state. Obtaining validated atomic models of voltage-dependent closed states is more challenging, as there are currently no methods to solve protein structure while a stable voltage gradient is applied across the length of an oriented channel. It is widely believed that the best approach to solve the atomic structure of a voltage-gated closed ion channel is to apply different but complementary experimental and computational methods and to use the resulting information to derive a consensus atomic structure that is then subjected to rigorous validation. In this paper, we summarize our efforts to obtain and validate atomic models of the open and voltage-driven closed states of undocked connexin hemichannels. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Thaddeus A Bargiello
- Dominic P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States.
| | - Seunghoon Oh
- Department of Physiology, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Qingxiu Tang
- Dominic P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Nicholas K Bargiello
- Dominic P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Terry L Dowd
- Department of Chemistry, Brooklyn College, Brooklyn, NY 11210, United States
| | - Taekyung Kwon
- Dominic P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| |
Collapse
|
29
|
Kim Y, Davidson JO, Green CR, Nicholson LFB, O'Carroll SJ, Zhang J. Connexins and Pannexins in cerebral ischemia. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:224-236. [PMID: 28347700 DOI: 10.1016/j.bbamem.2017.03.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/24/2017] [Accepted: 03/22/2017] [Indexed: 12/12/2022]
Abstract
A common cause of mortality and long-term adult disability, cerebral ischemia or brain ischemia imposes a significant health and financial burden on communities worldwide. Cerebral ischemia is a condition that arises from a sudden loss of blood flow and consequent failure to meet the high metabolic demands of the brain. The lack of blood flow initiates a sequelae of cell death mechanisms, including the activation of the inflammatory pathway, which can ultimately result in irreversible brain tissue damage. In particular, Connexins and Pannexins are non-selective channels with a large pore that have shown to play time-dependent roles in the perpetuation of ischaemic injury. This review highlights the roles of Connexin and Pannexin channels in cell death mechanisms as a promising therapeutic target in cerebral ischemia, and in particular connexin hemichannels which may contribute most of the ATP release as a result of ischemia as well as during reperfusion. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Yeri Kim
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland
| | - Joanne O Davidson
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland
| | - Colin R Green
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland
| | - Louise F B Nicholson
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland
| | - Simon J O'Carroll
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland
| | - Jie Zhang
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland.
| |
Collapse
|
30
|
Miller AC, Pereda AE. The electrical synapse: Molecular complexities at the gap and beyond. Dev Neurobiol 2017; 77:562-574. [PMID: 28170151 DOI: 10.1002/dneu.22484] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/04/2017] [Accepted: 01/04/2017] [Indexed: 12/21/2022]
Abstract
Gap junctions underlie electrical synaptic transmission between neurons. Generally perceived as simple intercellular channels, "electrical synapses" have demonstrated to be more functionally sophisticated and structurally complex than initially anticipated. Electrical synapses represent an assembly of multiple molecules, consisting of channels, adhesion complexes, scaffolds, regulatory machinery, and trafficking proteins, all required for their proper function and plasticity. Additionally, while electrical synapses are often viewed as strictly symmetric structures, emerging evidence has shown that some components forming electrical synapses can be differentially distributed at each side of the junction. We propose that the molecular complexity and asymmetric distribution of proteins at the electrical synapse provides rich potential for functional diversity. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 562-574, 2017.
Collapse
Affiliation(s)
- Adam C Miller
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, Oregon
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
31
|
Watanabe M. Gap Junction in the Teleost Fish Lineage: Duplicated Connexins May Contribute to Skin Pattern Formation and Body Shape Determination. Front Cell Dev Biol 2017; 5:13. [PMID: 28271062 PMCID: PMC5318405 DOI: 10.3389/fcell.2017.00013] [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: 12/16/2016] [Accepted: 02/07/2017] [Indexed: 11/13/2022] Open
Abstract
Gap junctions are intercellular channels that allow passage of ions and small molecules between adjacent cells. Gap junctions in vertebrates are composed of connexons, which are an assembly of six proteins, connexins. Docking of two connexons on the opposite cell surfaces forms a gap junction between the cytoplasm of two neighboring cells. Connexins compose a family of structurally related four-pass transmembrane proteins. In mammals, there are ~20 connexins, each of which contributes to unique permeability of gap junctions, and mutations of some connexin-encoding genes are associated with human diseases. Zebrafish has been predicted to contain 39 connexin-encoding genes; the high number can be attributed to gene duplication during fish evolution, which resulted in diversified functions of gap junctions in teleosts. The determination of body shapes and skin patterns in animal species is an intriguing question. Mathematical models suggest principle mechanisms explaining the diversification of animal morphology. Recent studies have revealed the involvement of gap junctions in fish morphological diversity, including skin pattern formation and body shape determination. This review focuses on connexins in teleosts, which are integrated in the mathematical models explaining morphological diversity of animal skin patterns and body shapes.
Collapse
|
32
|
Country MW, Jonz MG. Calcium dynamics and regulation in horizontal cells of the vertebrate retina: lessons from teleosts. J Neurophysiol 2017; 117:523-536. [PMID: 27832601 PMCID: PMC5288477 DOI: 10.1152/jn.00585.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/02/2016] [Indexed: 01/20/2023] Open
Abstract
Horizontal cells (HCs) are inhibitory interneurons of the vertebrate retina. Unlike typical neurons, HCs are chronically depolarized in the dark, leading to a constant influx of Ca2+ Therefore, mechanisms of Ca2+ homeostasis in HCs must differ from neurons elsewhere in the central nervous system, which undergo excitotoxicity when they are chronically depolarized or stressed with Ca2+ HCs are especially well characterized in teleost fish and have been used to unlock mysteries of the vertebrate retina for over one century. More recently, mammalian models of the retina have been increasingly informative for HC physiology. We draw from both teleost and mammalian models in this review, using a comparative approach to examine what is known about Ca2+ pathways in vertebrate HCs. We begin with a survey of Ca2+-permeable ion channels, exchangers, and pumps and summarize Ca2+ influx and efflux pathways, buffering, and intracellular stores. This includes evidence for Ca2+-permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors and N-methyl-d-aspartate receptors and for voltage-gated Ca2+ channels. Special attention is given to interactions between ion channels, to differences among species, and in which subtypes of HCs these channels have been found. We then discuss a number of unresolved issues pertaining to Ca2+ dynamics in HCs, including a potential role for Ca2+ in feedback to photoreceptors, the role for Ca2+-induced Ca2+ release, and the properties and functions of Ca2+-based action potentials. This review aims to highlight the unique Ca2+ dynamics in HCs, as these are inextricably tied to retinal function.
Collapse
Affiliation(s)
- Michael W Country
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Michael G Jonz
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
33
|
Greb H, Hermann S, Dirks P, Ommen G, Kretschmer V, Schultz K, Zoidl G, Weiler R, Janssen-Bienhold U. Complexity of gap junctions between horizontal cells of the carp retina. Neuroscience 2016; 340:8-22. [PMID: 27793781 DOI: 10.1016/j.neuroscience.2016.10.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 10/11/2016] [Accepted: 10/18/2016] [Indexed: 11/17/2022]
Abstract
In the vertebrate retina, horizontal cells (HCs) reveal homologous coupling by gap junctions (gj), which are thought to consist of different connexins (Cx). However, recent studies in mouse, rabbit and zebrafish retina indicate that individual HCs express more than one connexin. To provide further insights into the composition of gj connecting HCs and to determine whether HCs express multiple connexins, we examined the molecular identity and distribution of gj between HCs of the carp retina. We have cloned four carp connexins designated Cx49.5, Cx55.5, Cx52.6 and Cx53.8 with a close relationship to connexins previously reported in HCs of mouse, rabbit and zebrafish, respectively. Using in situ hybridization, Cx49.5 expression was detected in different subpopulations of retinal neurons including HCs, whereas the Cx52.6 transcript was localized exclusively in HCs. Using specific antibodies, Cx55.5 and Cx53.8 were detected on dendrites of all four HC subtypes and axon terminals. Immunoelectron microscopy confirmed the presence of Cx55.5 and Cx53.8 in gap junctions between these processes and Cx55.5 was additionally observed in HC dendrites invaginating cone pedicles, suggesting its participation in the modulation of photoreceptor output in the carp retina. Furthermore, using single-cell RT-PCR, all four connexins were detected in different subtypes of HCs, suggesting overlapping expression patterns. Thus, the composition of gj mediating homologous coupling between subtypes of carp HCs appears to be more complex than expected. Moreover, BLAST searches of the preliminary carp genome, using novel sequences as query, suggest that most of the analyzed connexin genes are duplicated in carp.
Collapse
Affiliation(s)
- H Greb
- Visual Neuroscience, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany
| | - S Hermann
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany
| | - P Dirks
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany
| | - G Ommen
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany
| | - V Kretschmer
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany; Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg University of Mainz, D-55128 Mainz, Germany
| | - K Schultz
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany
| | - G Zoidl
- Department of Psychology, Faculty of Health, York University, Toronto, Canada
| | - R Weiler
- Neurobiology, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany; Research Center Neurosensory Science, University of Oldenburg, D-26111 Oldenburg, Germany
| | - U Janssen-Bienhold
- Visual Neuroscience, Department of Neuroscience, University of Oldenburg, D-26111 Oldenburg, Germany; Research Center Neurosensory Science, University of Oldenburg, D-26111 Oldenburg, Germany.
| |
Collapse
|
34
|
Orellana JA. Physiological Functions of Glial Cell Hemichannels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:93-108. [DOI: 10.1007/978-3-319-40764-7_5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
35
|
Klaassen LJ, de Graaff W, van Asselt JB, Klooster J, Kamermans M. Specific connectivity between photoreceptors and horizontal cells in the zebrafish retina. J Neurophysiol 2016; 116:2799-2814. [PMID: 27707811 DOI: 10.1152/jn.00449.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/30/2016] [Indexed: 11/22/2022] Open
Abstract
The functional and morphological connectivity between various horizontal cell (HC) types (H1, H2, H3, and H4) and photoreceptors was studied in zebrafish retina. Since HCs are strongly coupled by gap junctions and feedback from HCs to photoreceptors depends strongly on connexin (Cx) hemichannels, we characterized the various HC Cxs (Cx52.6, Cx52.7, Cx52.9, and Cx55.5) in Xenopus oocytes. All Cxs formed hemichannels that were conducting at physiological membrane potentials. The Cx hemichannels differed in kinetic properties and voltage dependence, allowing for specific tuning of the coupling of HCs and the feedback signal from HCs to cones. The morphological connectivity between HC layers and cones was determined next. We used zebrafish expressing green fluorescent protein under the control of Cx promoters. We found that all HCs showed Cx55.5 promoter activity. Cx52.7 promoter activity was exclusively present in H4 cells, while Cx52.9 promoter activity occurred only in H1 cells. Cx52.6 promoter activity was present in H4 cells and in the ventral quadrant of the retina also in H1 cells. Finally, we determined the spectral sensitivities of the HC layers. Three response types were found. Monophasic responses were generated by HCs that contacted all cones (H1 cells), biphasic responses were generated by HCs that contacted M, S, and UV cones (H2 cells), and triphasic responses were generated by HCs that contacted either S and UV cones (H3 cells) or rods and UV cones (H4 cells). Electron microscopy confirms that H4 cells innervate cones. This indicates that rod-driven HCs process spectral information during photopic and luminance information during scotopic conditions.
Collapse
Affiliation(s)
- Lauw J Klaassen
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; and
| | - Wim de Graaff
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; and
| | - Jorrit B van Asselt
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; and
| | - Jan Klooster
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; and
| | - Maarten Kamermans
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; and .,Department of Genome Analysis, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
36
|
Kinetics of Inhibitory Feedback from Horizontal Cells to Photoreceptors: Implications for an Ephaptic Mechanism. J Neurosci 2016; 36:10075-88. [PMID: 27683904 DOI: 10.1523/jneurosci.1090-16.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/12/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Inhibitory feedback from horizontal cells (HCs) to cones generates center-surround receptive fields and color opponency in the retina. Mechanisms of HC feedback remain unsettled, but one hypothesis proposes that an ephaptic mechanism may alter the extracellular electrical field surrounding photoreceptor synaptic terminals, thereby altering Ca(2+) channel activity and photoreceptor output. An ephaptic voltage change produced by current flowing through open channels in the HC membrane should occur with no delay. To test for this mechanism, we measured kinetics of inhibitory feedback currents in Ambystoma tigrinum cones and rods evoked by hyperpolarizing steps applied to synaptically coupled HCs. Hyperpolarizing HCs stimulated inward feedback currents in cones that averaged 8-9 pA and exhibited a biexponential time course with time constants averaging 14-17 ms and 120-220 ms. Measurement of feedback-current kinetics was limited by three factors: (1) HC voltage-clamp speed, (2) cone voltage-clamp speed, and (3) kinetics of Ca(2+) channel activation or deactivation in the photoreceptor terminal. These factors totaled ∼4-5 ms in cones meaning that the true fast time constants for HC-to-cone feedback currents were 9-13 ms, slower than expected for ephaptic voltage changes. We also compared speed of feedback to feedforward glutamate release measured at the same cone/HC synapses and found a latency for feedback of 11-14 ms. Inhibitory feedback from HCs to rods was also significantly slower than either measurement kinetics or feedforward release. The finding that inhibitory feedback from HCs to photoreceptors involves a significant delay indicates that it is not due to previously proposed ephaptic mechanisms. SIGNIFICANCE STATEMENT Lateral inhibitory feedback from horizontal cells (HCs) to photoreceptors creates center-surround receptive fields and color-opponent interactions. Although underlying mechanisms remain unsettled, a longstanding hypothesis proposes that feedback is due to ephaptic voltage changes that regulate photoreceptor synaptic output by altering Ca(2+) channel activity. Ephaptic processes should occur with no delay. We measured kinetics of inhibitory feedback currents evoked in photoreceptors with voltage steps applied to synaptically coupled HCs and found that feedback is too slow to be explained by ephaptic voltage changes generated by current flowing through continuously open channels in HC membranes. By eliminating the proposed ephaptic mechanism for HC feedback regulation of photoreceptor Ca(2+) channels, our data support earlier proposals that synaptic cleft pH changes are more likely responsible.
Collapse
|
37
|
Gajardo-Gómez R, Labra VC, Orellana JA. Connexins and Pannexins: New Insights into Microglial Functions and Dysfunctions. Front Mol Neurosci 2016; 9:86. [PMID: 27713688 PMCID: PMC5031785 DOI: 10.3389/fnmol.2016.00086] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/05/2016] [Indexed: 12/11/2022] Open
Abstract
Under physiological conditions, microglia adopt a resting phenotype associated with the production of anti-inflammatory and neurotrophic factors. In response to a wide variety of insults, these cells shift to an activated phenotype that is necessary for the proper restoration of brain homeostasis. However, when the intensity of a threat is relatively high, microglial activation worsens the progression of damage rather than providing protection, with potentially significant consequences for neuronal survival. Coordinated interactions among microglia and other brain cells, including astrocytes and neurons, are critical for the development of timely and optimal inflammatory responses in the brain parenchyma. Tissue synchronization is in part mediated by connexins and pannexins, which are protein families that form different plasma membrane channels to communicate with neighboring cells. Gap junction channels (which are exclusively formed by connexins in vertebrates) connect the cytoplasm of contacting cells to coordinate electrical and metabolic coupling. Hemichannels (HCs) and pannexons (which are formed by connexins and pannexins, respectively) communicate the intra- and extracellular compartments and serve as diffusion pathways for the exchange of ions and small molecules. In this review article, we discuss the available evidence concerning the functional expression and regulation of connexin- and pannexin-based channels in microglia and their contributions to microglial function and dysfunction. Specifically, we focus on the possible implications of these channels in microglia-to-microglia, microglia-to-astrocyte and neuron-to-microglia interactions in the inflamed brain.
Collapse
Affiliation(s)
- Rosario Gajardo-Gómez
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Valeria C Labra
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Juan A Orellana
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
38
|
Johnson RG, Le HC, Evenson K, Loberg SW, Myslajek TM, Prabhu A, Manley AM, O’Shea C, Grunenwald H, Haddican M, Fitzgerald PM, Robinson T, Cisterna BA, Sáez JC, Liu TF, Laird DW, Sheridan JD. Connexin Hemichannels: Methods for Dye Uptake and Leakage. J Membr Biol 2016; 249:713-741. [DOI: 10.1007/s00232-016-9925-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/22/2016] [Indexed: 01/18/2023]
|
39
|
Yoshikawa S, Vila A, Segelken J, Lin YP, Mitchell CK, Nguyen D, O'Brien J. Zebrafish connexin 79.8 (Gja8a): A lens connexin used as an electrical synapse in some neurons. Dev Neurobiol 2016; 77:548-561. [PMID: 27402207 DOI: 10.1002/dneu.22418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/23/2016] [Accepted: 07/05/2016] [Indexed: 11/07/2022]
Abstract
In the mammalian central nervous system, a remarkably small number of connexins is used in electrical synapses, with the majority formed from Cx36. A larger number has been detected in teleosts, with some seeming to serve restricted roles. Here, we report the discovery of a new connexin expressed in the zebrafish lens and a limited set of neurons. Zebrafish cx79.8 (gja8a), previously annotated incorrectly as cx50.5 based on a partial cDNA sequence, is a homologue of mammalian Cx50 (Gja8). We examined its expression through transgenic promoter-reporter constructs, in situ hybridization, and immunolabeling, and examined regulation of coupling in transfected HeLa cells. cx79.8 was expressed most strongly in the lens, but expression was also found in several groups of neurons in the cerebellum and related areas at the midbrain-hindbrain boundary, in cone photoreceptors, and in neurons in the retinal inner nuclear and ganglion cell layers. Labeling in the retina with antibodies against two C-terminal epitopes revealed numerous small punctate spots in the inner plexiform layer and along the somata of cones. Abundant gap junctions were labeled in the outer 1/3 of the lens, but were absent from the center, suggesting that the epitopes or the entire protein was absent from the center. Cx79.8 tracer coupling was strongly regulated by phosphorylation, and was extremely low in control conditions in HeLa cells due to protein phosphatase 2A activity. These properties allow coupling to be strongly restricted in situ, a frequently observed property for electrical synapses. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 548-561, 2017.
Collapse
Affiliation(s)
- Shunichi Yoshikawa
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas
| | - Alejandro Vila
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas.,University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Jasmin Segelken
- Visual Neuroscience, Faculty of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Ya-Ping Lin
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas
| | - Cheryl K Mitchell
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas
| | - Duc Nguyen
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas
| | - John O'Brien
- Richard S. Ruiz M.D., Department of Ophthalmology and Visual Science, University of Texas Health Science Center at Houston, Houston, Texas.,University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
40
|
Abstract
Electrical synapses are an omnipresent feature of nervous systems, from the simple nerve nets of cnidarians to complex brains of mammals. Formed by gap junction channels between neurons, electrical synapses allow direct transmission of voltage signals between coupled cells. The relative simplicity of this arrangement belies the sophistication of these synapses. Coupling via electrical synapses can be regulated by a variety of mechanisms on times scales ranging from milliseconds to days, and active properties of the coupled neurons can impart emergent properties such as signal amplification, phase shifts and frequency-selective transmission. This article reviews the biophysical characteristics of electrical synapses and some of the core mechanisms that control their plasticity in the vertebrate central nervous system.
Collapse
Affiliation(s)
- Sebastian Curti
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | - John O'Brien
- Department of Ophthalmology & Visual Science, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
41
|
Klooster J, Kamermans M. An Ultrastructural and Immunohistochemical Analysis of the Outer Plexiform Layer of the Retina of the European Silver Eel (Anguilla anguilla L). PLoS One 2016; 11:e0152967. [PMID: 27032102 PMCID: PMC4816530 DOI: 10.1371/journal.pone.0152967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/22/2016] [Indexed: 11/29/2022] Open
Abstract
Here we studied the ultrastructural organization of the outer retina of the European silver eel, a highly valued commercial fish species. The retina of the European eel has an organization very similar to most vertebrates. It contains both rod and cone photoreceptors. Rods are abundantly present and immunoreactive for rhodopsin. Cones are sparsely present and only show immunoreactivity for M-opsin and not for L-, S- or UV-cone opsins. As in all other vertebrate retinas, Müller cells span the width of the retina. OFF-bipolar cells express the ionotropic glutamate receptor GluR4 and ON-bipolar cells, as identified by their PKCα immunoreactivity, express the metabotropic receptor mGluR6. Both the ON- and the OFF-bipolar cell dendrites innervate the cone pedicle and rod spherule. Horizontal cells are surrounded by punctate Cx53.8 immunoreactivity indicating that the horizontal cells are strongly electrically coupled by gap-junctions. Connexin-hemichannels were found at the tips of the horizontal cell dendrites invaginating the photoreceptor synapse. Such hemichannels are implicated in the feedback pathway from horizontal cells to cones. Finally, horizontal cells are surrounded by tyrosine hydroxylase immunoreactivity, illustrating a strong dopaminergic input from interplexiform cells.
Collapse
Affiliation(s)
- Jan Klooster
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Maarten Kamermans
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
- Department of Neurogenetics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
42
|
Kim Y, Davidson JO, Gunn KC, Phillips AR, Green CR, Gunn AJ. Role of Hemichannels in CNS Inflammation and the Inflammasome Pathway. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 104:1-37. [DOI: 10.1016/bs.apcsb.2015.12.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
43
|
Abstract
Lateral inhibition at the first synapse in the retina is important for visual perception, enhancing image contrast, color discrimination, and light adaptation. Despite decades of research, the feedback signal from horizontal cells to photoreceptors that generates lateral inhibition remains uncertain. GABA, protons, or an ephaptic mechanism have all been suggested as the primary mediator of feedback. However, the complexity of the reciprocal cone to horizontal cell synapse has left the identity of the feedback signal an unsolved mystery.
Collapse
|
44
|
Becker DL, Phillips AR, Duft BJ, Kim Y, Green CR. Translating connexin biology into therapeutics. Semin Cell Dev Biol 2015; 50:49-58. [PMID: 26688335 DOI: 10.1016/j.semcdb.2015.12.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 12/07/2015] [Indexed: 12/26/2022]
Abstract
It is 45 years since gap junctions were first described. Universities face increasing commercial pressures and declining federal funding, with governments and funding foundations showing greater interest in gaining return on their investments. This review outlines approaches taken to translate gap junction research to clinical application and the challenges faced. The need for commercialisation is discussed and key concepts behind research patenting briefly described. Connexin channel roles in disease and injury are also discussed, as is identification of the connexin hemichannel as a therapeutic target which appears to play a role in both the start and perpetuation of the inflammasome pathway. Furthermore connexin hemichannel opening results in vascular dieback in acute injury and chronic disease. Translation to human indications is illustrated from the perspective of one connexin biotechnology company, CoDa Therapeutics, Inc.
Collapse
Affiliation(s)
- David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | | | | | - Yeri Kim
- Department of Ophthalmology and New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Colin R Green
- Department of Ophthalmology and New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.
| |
Collapse
|
45
|
Berkowitz BA, Bissig D, Roberts R. MRI of rod cell compartment-specific function in disease and treatment in vivo. Prog Retin Eye Res 2015; 51:90-106. [PMID: 26344734 DOI: 10.1016/j.preteyeres.2015.09.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/26/2015] [Accepted: 09/01/2015] [Indexed: 10/23/2022]
Abstract
Rod cell oxidative stress is a major pathogenic factor in retinal disease, such as diabetic retinopathy (DR) and retinitis pigmentosa (RP). Personalized, non-destructive, and targeted treatment for these diseases remains elusive since current imaging methods cannot analytically measure treatment efficacy against rod cell compartment-specific oxidative stress in vivo. Over the last decade, novel MRI-based approaches that address this technology gap have been developed. This review summarizes progress in the development of MRI since 2006 that enables earlier evaluation of the impact of disease on rod cell compartment-specific function and the efficacy of anti-oxidant treatment than is currently possible with other methods. Most of the new assays of rod cell compartment-specific function are based on endogenous contrast mechanisms, and this is expected to facilitate their translation into patients with DR and RP, and other oxidative stress-based retinal diseases.
Collapse
Affiliation(s)
- Bruce A Berkowitz
- Dept. of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI, USA; Dept. Of Ophthalmology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - David Bissig
- Dept. of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robin Roberts
- Dept. Of Ophthalmology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
46
|
Decrock E, De Bock M, Wang N, Bultynck G, Giaume C, Naus CC, Green CR, Leybaert L. Connexin and pannexin signaling pathways, an architectural blueprint for CNS physiology and pathology? Cell Mol Life Sci 2015; 72:2823-51. [PMID: 26118660 PMCID: PMC11113968 DOI: 10.1007/s00018-015-1962-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 02/06/2023]
Abstract
The central nervous system (CNS) is composed of a highly heterogeneous population of cells. Dynamic interactions between different compartments (neuronal, glial, and vascular systems) drive CNS function and allow to integrate and process information as well as to respond accordingly. Communication within this functional unit, coined the neuro-glio-vascular unit (NGVU), typically relies on two main mechanisms: direct cell-cell coupling via gap junction channels (GJCs) and paracrine communication via the extracellular compartment, two routes to which channels composed of transmembrane connexin (Cx) or pannexin (Panx) proteins can contribute. Multiple isoforms of both protein families are present in the CNS and each CNS cell type is characterized by a unique Cx/Panx portfolio. Over the last two decades, research has uncovered a multilevel platform via which Cxs and Panxs can influence different cellular functions within a tissue: (1) Cx GJCs enable a direct cell-cell communication of small molecules, (2) Cx hemichannels and Panx channels can contribute to autocrine/paracrine signaling pathways, and (3) different structural domains of these proteins allow for channel-independent functions, such as cell-cell adhesion, interactions with the cytoskeleton, and the activation of intracellular signaling pathways. In this paper, we discuss current knowledge on their multifaceted contribution to brain development and to specific processes in the NGVU, including synaptic transmission and plasticity, glial signaling, vasomotor control, and blood-brain barrier integrity in the mature CNS. By highlighting both physiological and pathological conditions, it becomes evident that Cxs and Panxs can play a dual role in the CNS and that an accurate fine-tuning of each signaling mechanism is crucial for normal CNS physiology.
Collapse
Affiliation(s)
- Elke Decrock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Marijke De Bock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Nan Wang
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, 75231 Paris Cedex 05, France
- University Pierre et Marie
Curie, ED, N°158, 75005 Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, 75005 Paris, France
| | - Christian C. Naus
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Colin R. Green
- Department of Ophthalmology, The University of Auckland, Auckland, New Zealand
| | - Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| |
Collapse
|
47
|
Dorgau B, Herrling R, Schultz K, Greb H, Segelken J, Ströh S, Bolte P, Weiler R, Dedek K, Janssen-Bienhold U. Connexin50 couples axon terminals of mouse horizontal cells by homotypic gap junctions. J Comp Neurol 2015; 523:2062-81. [PMID: 25823610 DOI: 10.1002/cne.23779] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 02/12/2015] [Accepted: 03/24/2015] [Indexed: 11/06/2022]
Abstract
Horizontal cells in the mouse retina are of the axon-bearing B-type and contribute to the gain control of photoreceptors and to the center-surround organization of bipolar cells by providing feedback and feedforward signals to photoreceptors and bipolar cells, respectively. Horizontal cells form two independent networks, coupled by dendro-dendritic and axo-axonal gap junctions composed of connexin57 (Cx57). In Cx57-deficient mice, occasionally the residual tracer coupling of horizontal cell somata was observed. Also, negative feedback from horizontal cells to photoreceptors, potentially mediated by connexin hemichannels, appeared unaffected. These results point to the expression of a second connexin in mouse horizontal cells. We investigated the expression of Cx50, which was recently identified in axonless A-type horizontal cells of the rabbit retina. In the mouse retina, Cx50-immunoreactive puncta were predominantly localized on large axon terminals of horizontal cells. Electron microscopy did not reveal any Cx50-immunolabeling at the membrane of horizontal cell tips invaginating photoreceptor terminals, ruling out the involvement of Cx50 in negative feedback. Moreover, Cx50 colocalized only rarely with Cx57 on horizontal cell processes, indicating that both connexins form homotypic rather than heterotypic or heteromeric gap junctions. To check whether the expression of Cx50 is changed when Cx57 is lacking, we compared the Cx50 expression in wildtype and Cx57-deficient mice. However, Cx50 expression was unaffected in Cx57-deficient mice. In summary, our results indicate that horizontal cell axon terminals form two independent sets of homotypic gap junctions, a feature which might be important for light adaptation in the retina.
Collapse
Affiliation(s)
- Birthe Dorgau
- Department of Neurobiology, University of Oldenburg, D-26111, Oldenburg, Germany.,Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | - Regina Herrling
- Department of Neurobiology, University of Oldenburg, D-26111, Oldenburg, Germany
| | - Konrad Schultz
- Department of Neurobiology, University of Oldenburg, D-26111, Oldenburg, Germany
| | - Helena Greb
- Department of Neurobiology, University of Oldenburg, D-26111, Oldenburg, Germany
| | - Jasmin Segelken
- Department of Neurobiology, University of Oldenburg, D-26111, Oldenburg, Germany
| | - Sebastian Ströh
- Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | - Petra Bolte
- Institute of Genetic Medicine, Newcastle University, Newcastle, UK.,Animal Navigation, University of Oldenburg, Oldenburg, Germany
| | | | - Karin Dedek
- Department of Neurobiology, University of Oldenburg, D-26111, Oldenburg, Germany
| | | |
Collapse
|
48
|
Vroman R, Kamermans M. Feedback-induced glutamate spillover enhances negative feedback from horizontal cells to cones. J Physiol 2015; 593:2927-40. [PMID: 25820622 DOI: 10.1113/jp270158] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/24/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS In the retina, horizontal cells feed back negatively to cone photoreceptors. Glutamate released from cones can spill over to neighbouring cones. Here we show that cone glutamate release induced by negative feedback can also spill over to neighbouring cones. This glutamate activates the glutamate transporter-associated chloride current in these neighbouring cones, which leads to a change in their membrane potential and thus modulates their output. In this way, feedback-induced glutamate spillover enhances negative feedback from horizontal cells to cones, thus forming an additional feedback pathway. This effect will be particularly prominent in cones that are strongly hyperpolarized by light. ABSTRACT Inhibition in the outer retina functions via an unusual mechanism. When horizontal cells hyperpolarize the activation potential of the Ca(2+) current of cones shifts to more negative potentials. The underlying mechanism consists of an ephaptic component and a Panx1/ATP-mediated component. Here we identified a third feedback component, which remains active outside the operating range of the Ca(2+) current. We show that the glutamate transporters of cones can be activated by glutamate released from their neighbours. This pathway can be triggered by negative feedback from horizontal cells to cones, thus providing an additional feedback pathway. This additional pathway is mediated by a Cl(-) current, can be blocked by either removing the gradient of K(+) or by adding the glutamate transporter blocker TBOA, or low concentrations of Zn(2+) . These features point to a glutamate transporter-associated Cl(-) current. The pathway has a delay of 4.7 ± 1.7 ms. The effectiveness of this pathway in modulating the cone output depends on the equilibrium potential of Cl(-) (ECl ) and the membrane potential of the cone. Because estimates of ECl show that it is around the dark resting membrane potential of cones, the activation of the glutamate transporter-associated Cl(-) current will be most effective in changing the membrane potential during strong hyperpolarization of cones. This means that negative feedback would particularly be enhanced by this pathway when cones are hyperpolarized. Spatially, this pathway does not reach further than the direct neighbouring cones. The consequence is that this feedback pathway transmits information between cones of different spectral type.
Collapse
Affiliation(s)
- Rozan Vroman
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Maarten Kamermans
- Retinal Signal Processing Lab, Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.,Department of Neurogenetics, University of Amsterdam, Academic Medical Centre, Meibergdreef 15, 1105 AZ, Amsterdam, Netherlands
| |
Collapse
|
49
|
Montero TD, Orellana JA. Hemichannels: new pathways for gliotransmitter release. Neuroscience 2014; 286:45-59. [PMID: 25475761 DOI: 10.1016/j.neuroscience.2014.11.048] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/14/2014] [Accepted: 11/20/2014] [Indexed: 01/16/2023]
Abstract
Growing evidence suggests that glial cells express virtually all known types of neurotransmitter receptors, enabling them to sense neuronal activity and microenvironment changes by responding locally via the Ca(2+)-dependent release of bioactive molecules, known as "gliotransmitters". Several mechanisms of gliotransmitter release have been documented. One of these mechanisms involves the opening of plasma membrane channels, known as hemichannels. These channels are composed of six protein subunits consisting of connexins or pannexins, two highly conserved protein families encoded by 21 or 3 genes, respectively, in humans. Most data indicate that under physiological conditions, glial cell hemichannels have low activity, but this activity is sufficient to ensure the release of relevant quantities of gliotransmitters to the extracellular milieu, including ATP, glutamate, adenosine and glutathione. Nevertheless, it has been suggested that dysregulations of hemichannel properties could be critical in the beginning and during the maintenance of homeostatic imbalances observed in several brain diseases. In this study, we review the current knowledge on the hemichannel-dependent release of gliotransmitters in the physiology and pathophysiology of the CNS.
Collapse
Affiliation(s)
- T D Montero
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - J A Orellana
- Departamento de Neurología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
50
|
Cheung G, Chever O, Rouach N. Connexons and pannexons: newcomers in neurophysiology. Front Cell Neurosci 2014; 8:348. [PMID: 25408635 PMCID: PMC4219455 DOI: 10.3389/fncel.2014.00348] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/06/2014] [Indexed: 11/14/2022] Open
Abstract
Connexin hemichannels are single membrane channels which have been traditionally thought to work in pairs to form gap junction channels across two opposing cells. In astrocytes, gap junction channels allow direct intercellular communication and greatly facilitate the transmission of signals. Recently, there has been growing evidence demonstrating that connexin hemichannels, as well as pannexin channels, on their own are open in various conditions. They allow bidirectional flow of ions and signaling molecules and act as release sites for transmitters like ATP and glutamate into the extracellular space. While much attention has focused on the function of connexin hemichannels and pannexons during pathological situations like epilepsy, inflammation, neurodegeneration or ischemia, their potential roles in physiology is often ignored. In order to fully understand the dynamic properties and roles of connexin hemichannels and pannexons in the brain, it is essential to decipher whether they also have some physiological functions and contribute to normal cerebral processes. Here, we present recent studies in the CNS suggesting emerging physiological functions of connexin hemichannels and pannexons in normal neuronal activity and behavior. We also discuss how these pioneer studies pave the way for future research to extend the physiological relevance of connexons and pannexons, and some fundamental issues yet to be addressed.
Collapse
Affiliation(s)
- Giselle Cheung
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University Paris, France
| | - Oana Chever
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University Paris, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University Paris, France
| |
Collapse
|