1
|
Onyak JR, Vergara MN, Renna JM. Retinal organoid light responsivity: current status and future opportunities. Transl Res 2022; 250:98-111. [PMID: 35690342 DOI: 10.1016/j.trsl.2022.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022]
Abstract
The ability to generate human retinas in vitro from pluripotent stem cells opened unprecedented opportunities for basic science and for the development of therapeutic approaches for retinal degenerative diseases. Retinal organoid models not only mimic the histoarchitecture and cellular composition of the native retina, but they can achieve a remarkable level of maturation that allows them to respond to light stimulation. However, studies evaluating the nature, magnitude, and properties of light-evoked responsivity from each cell type, in each retinal organoid layer, have been sparse. In this review we discuss the current understanding of retinal organoid function, the technologies used for functional assessment in human retinal organoids, and the challenges and opportunities that lie ahead.
Collapse
Affiliation(s)
| | - M Natalia Vergara
- CellSight Ocular Stem Cell and Regeneration Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, Colorado.
| | - Jordan M Renna
- Department of Biology, The University of Akron, Akron, Ohio.
| |
Collapse
|
2
|
Zhu Y, Cao B, Tolone A, Yan J, Christensen G, Arango-Gonzalez B, Ueffing M, Paquet-Durand F. In vitro Model Systems for Studies Into Retinal Neuroprotection. Front Neurosci 2022; 16:938089. [PMID: 35873807 PMCID: PMC9301112 DOI: 10.3389/fnins.2022.938089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Therapy development for neurodegenerative diseases of the retina constitutes a major unmet medical need, and this may be particularly relevant for inherited diseases of the retina, which are largely untreatable to this day. Therapy development necessitates appropriate models to improve the understanding of the underlying degenerative mechanisms, as well as for the testing and evaluation of novel treatment approaches. This review provides an overview of various in vitro model systems used to study retinal neuroprotection. The in vitro methods and technologies discussed range from primary retinal cell cultures and cell lines, to retinal organoids and organotypic retinal explants, to the cultivation of whole eyeballs. The advantages and disadvantages of these methods are compared and evaluated, also in view of the 3R principles (i.e., the refinement, reduction, and replacement of live animal testing), to identify suitable in vitro alternatives for in vivo experimentation. The article further expands on the use of in vitro models to test and evaluate neuroprotective treatments and to aid the development of retinal drug delivery systems. Among the pharmacological agents tested and characterized in vitro are such that interfere with aberrant cyclic guanosine monophosphate (cGMP) -signaling or such that inhibit the activities of poly (ADP-ribose) polymerase (PARP), histone deacetylases (HDAC), calpain-type proteases, as well as unfolded protein response-related stress. We then introduce nanoparticle-based drug delivery systems and discuss how different in vitro systems may be used to assess their efficacy in the treatment of retinal diseases. The summary provides a brief comparison of available in vitro models and relates their advantages and limitations to the various experimental requirements, for instance, for studies into disease mechanisms, novel treatments, or retinal toxicity. In many cases, combinations of different in vitro models may be required to obtain a comprehensive view of the efficacy of a given retinal neuroprotection approach.
Collapse
Affiliation(s)
- Yu Zhu
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, Germany
| | - Bowen Cao
- Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, Germany
- Molecular Biology of Retinal Degenerations, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Arianna Tolone
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Jie Yan
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, Germany
| | - Gustav Christensen
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, Germany
| | - Blanca Arango-Gonzalez
- Molecular Biology of Retinal Degenerations, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Marius Ueffing
- Molecular Biology of Retinal Degenerations, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- *Correspondence: Marius Ueffing,
| | - François Paquet-Durand
- Cell Death Mechanisms Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- François Paquet-Durand,
| |
Collapse
|
3
|
Li L, Zhao H, Xie H, Akhtar T, Yao Y, Cai Y, Dong K, Gu Y, Bao J, Chen J, Zhang M, Zhong K, Xu W, Xue T. Electrophysiological characterization of photoreceptor-like cells in human inducible pluripotent stem cell-derived retinal organoids during in vitro maturation. STEM CELLS (DAYTON, OHIO) 2021; 39:959-974. [PMID: 33662144 DOI: 10.1002/stem.3363] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/10/2021] [Indexed: 11/10/2022]
Abstract
Retinal organoids (ROs) derived from human inducible pluripotent stem cells (hiPSCs) exhibit considerable therapeutic potential. However, current quality control of ROs during in vitro differentiation is largely limited to the detection of molecular markers, often by immunostaining, polymerase chain reaction (PCR) assays and sequencing, often without proper functional assessments. As such, in the current study, we systemically characterized the physiological maturation of photoreceptor-like cells in hiPSC-derived ROs. By performing patch-clamp recordings from photoreceptor-like cells in ROs at distinct differentiation stages (ie, Differentiation Day [D]90, D150, and D200), we determined the electrophysiological properties of the plasma membrane and several characteristic ion channels closely associated with the physiological functions of the photoreceptors. Ionic hallmarks, such as hyperpolarization-activated cyclic nucleotide-gated (HCN) channels and cyclic nucleotide-gated (CNG) channels, matured progressively during differentiation. After D200 in culture, these characteristic currents closely resembled those in macaque or human native photoreceptors. Furthermore, we demonstrated that the hyperpolarization-activated inward current/depolarization-activated outward current ratio (I-120 /I+40 ), termed as the inward-outward current (IOC) ratio hereon, accurately represented the maturity of photoreceptors and could serve as a sensitive indicator of pathological state. Thus, this study provides a comprehensive dataset describing the electrophysiological maturation of photoreceptor-like cells in hiPSC-derived ROs for precise and sensitive quality control during RO differentiation.
Collapse
Affiliation(s)
- Lingyun Li
- Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China.,CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China
| | - Huan Zhao
- School of Biology, Food, and Environment, Hefei University, Hefei, People's Republic of China
| | - Haohuan Xie
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China
| | - Tasneem Akhtar
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China
| | - Yichuan Yao
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China
| | - Yuan Cai
- Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Kai Dong
- Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Yonghao Gu
- Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Jin Bao
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, People's Republic of China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Jutao Chen
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, People's Republic of China
| | - Mei Zhang
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, People's Republic of China
| | - Kai Zhong
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, People's Republic of China.,Key Laboratory of Anhui Province for High Field Magnetic Resonance Imaging, Hefei, People's Republic of China
| | - Weiping Xu
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, People's Republic of China.,The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Tian Xue
- Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China.,CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, People's Republic of China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, People's Republic of China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
4
|
Bryman GS, Liu A, Do MTH. Optimized Signal Flow through Photoreceptors Supports the High-Acuity Vision of Primates. Neuron 2020; 108:335-348.e7. [PMID: 32846139 DOI: 10.1016/j.neuron.2020.07.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/24/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023]
Abstract
The fovea is a neural specialization that endows humans and other primates with the sharpest vision among mammals. This performance originates in the foveal cones, which are extremely narrow and long to form a high-resolution pixel array. Puzzlingly, this form is predicted to impede electrical conduction to an extent that appears incompatible with vision. We observe the opposite: signal flow through even the longest cones (0.4-mm axons) is essentially lossless. Unlike in most neurons, amplification and impulse generation by voltage-gated channels are dispensable. Rather, sparse channel activity preserves intracellular current, which flows as if unobstructed by organelles. Despite these optimizations, signaling would degrade if cones were lengthier. Because cellular packing requires that cone elongation accompanies foveal expansion, this degradation helps explain why the fovea is a constant, miniscule size despite multiplicative changes in eye size through evolution. These observations reveal how biophysical mechanisms tailor form-function relationships for primate behavioral performance.
Collapse
Affiliation(s)
- Gregory S Bryman
- F.M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital and Harvard Medical School, Center for Life Science 12061, 3 Blackfan Circle, Boston, MA 02115, USA.
| | - Andreas Liu
- F.M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital and Harvard Medical School, Center for Life Science 12061, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Michael Tri H Do
- F.M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital and Harvard Medical School, Center for Life Science 12061, 3 Blackfan Circle, Boston, MA 02115, USA.
| |
Collapse
|
5
|
Van Hook MJ, Nawy S, Thoreson WB. Voltage- and calcium-gated ion channels of neurons in the vertebrate retina. Prog Retin Eye Res 2019; 72:100760. [PMID: 31078724 PMCID: PMC6739185 DOI: 10.1016/j.preteyeres.2019.05.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/25/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
In this review, we summarize studies investigating the types and distribution of voltage- and calcium-gated ion channels in the different classes of retinal neurons: rods, cones, horizontal cells, bipolar cells, amacrine cells, interplexiform cells, and ganglion cells. We discuss differences among cell subtypes within these major cell classes, as well as differences among species, and consider how different ion channels shape the responses of different neurons. For example, even though second-order bipolar and horizontal cells do not typically generate fast sodium-dependent action potentials, many of these cells nevertheless possess fast sodium currents that can enhance their kinetic response capabilities. Ca2+ channel activity can also shape response kinetics as well as regulating synaptic release. The L-type Ca2+ channel subtype, CaV1.4, expressed in photoreceptor cells exhibits specific properties matching the particular needs of these cells such as limited inactivation which allows sustained channel activity and maintained synaptic release in darkness. The particular properties of K+ and Cl- channels in different retinal neurons shape resting membrane potentials, response kinetics and spiking behavior. A remaining challenge is to characterize the specific distributions of ion channels in the more than 100 individual cell types that have been identified in the retina and to describe how these particular ion channels sculpt neuronal responses to assist in the processing of visual information by the retina.
Collapse
Affiliation(s)
- Matthew J Van Hook
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Scott Nawy
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department Pharmacology & Experimental Neuroscience(2), University of Nebraska Medical Center, Omaha, NE, USA
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department Pharmacology & Experimental Neuroscience(2), University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
6
|
Choudhury S, Strang CE, Alexander JJ, Scalabrino ML, Lynch Hill J, Kasuga DT, Witherspoon CD, Boye SL, Gamlin PD, Boye SE. Novel Methodology for Creating Macaque Retinas with Sortable Photoreceptors and Ganglion Cells. Front Neurosci 2016; 10:551. [PMID: 27990105 PMCID: PMC5131003 DOI: 10.3389/fnins.2016.00551] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/16/2016] [Indexed: 01/08/2023] Open
Abstract
Purpose: The ability to generate macaque retinas with sortable cell populations would be of great benefit to both basic and translational studies of the primate retina. The purpose of our study was therefore to develop methods to achieve this goal by selectively labeling, in life, photoreceptors (PRs) and retinal ganglion cells (RGCs) with separate fluorescent markers. Methods: Labeling of macaque (Macaca fascicularis) PRs and RGCs was accomplished by subretinal delivery of AAV5-hGRK1-GFP, and retrograde transport of micro-ruby™ from the lateral geniculate nucleus, respectively. Retinas were anatomically separated into different regions. Dissociation conditions were optimized, and cells from each region underwent fluorescent activated cell sorting (FACS). Expression of retinal cell type- specific genes was assessed by quantitative real-time PCR to characterize isolated cell populations. Results: We show that macaque PRs and RGCs can be simultaneously labeled in-life and enriched populations isolated by FACS. Recovery from different retinal regions indicated efficient isolation/enrichment for PRs and RGCs, with the macula being particularly amendable to this technique. Conclusions: The methods and materials presented here allow for the identification of novel reagents designed to target RGCs and/or photoreceptors in a species that is phylogenetically and anatomically similar to human. These techniques will enable screening of intravitreally-delivered AAV capsid libraries for variants with increased tropism for PRs and/or RGCs and the evaluation of vector tropism and/or cellular promoter activity of gene therapy vectors in a clinically relevant species.
Collapse
Affiliation(s)
| | - Christianne E Strang
- Department of Psychology, University of Alabama at Birmingham Birmingham, AL, USA
| | | | | | - Julie Lynch Hill
- Department of Ophthalmology, University of Alabama at Birmingham Birmingham, AL, USA
| | - Daniel T Kasuga
- Department of Ophthalmology, University of Alabama at Birmingham Birmingham, AL, USA
| | - C Douglas Witherspoon
- Department of Ophthalmology, University of Alabama at Birmingham Birmingham, AL, USA
| | - Sanford L Boye
- Department of Ophthalmology, University of Florida Gainesville, FL, USA
| | - Paul D Gamlin
- Department of Human Genetics, Emory University Atlanta, GA, USA
| | - Shannon E Boye
- Department of Ophthalmology, University of Florida Gainesville, FL, USA
| |
Collapse
|
7
|
Teotia P, Chopra DA, Dravid SM, Van Hook MJ, Qiu F, Morrison J, Rizzino A, Ahmad I. Generation of Functional Human Retinal Ganglion Cells with Target Specificity from Pluripotent Stem Cells by Chemically Defined Recapitulation of Developmental Mechanism. Stem Cells 2016; 35:572-585. [PMID: 27709736 DOI: 10.1002/stem.2513] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/06/2016] [Accepted: 09/12/2016] [Indexed: 01/07/2023]
Abstract
Glaucoma is a complex group of diseases wherein a selective degeneration of retinal ganglion cells (RGCs) lead to irreversible loss of vision. A comprehensive approach to glaucomatous RGC degeneration may include stem cells to functionally replace dead neurons through transplantation and understand RGCs vulnerability using a disease in a dish stem cell model. Both approaches require the directed generation of stable, functional, and target-specific RGCs from renewable sources of cells, that is, the embryonic stem cells and induced pluripotent stem cells. Here, we demonstrate a rapid and safe, stage-specific, chemically defined protocol that selectively generates RGCs across species, including human, by recapitulating the developmental mechanism. The de novo generated RGCs from pluripotent cells are similar to native RGCs at the molecular, biochemical, functional levels. They also express axon guidance molecules, and discriminate between specific and nonspecific targets, and are nontumorigenic. Stem Cells 2017;35:572-585.
Collapse
Affiliation(s)
- Pooja Teotia
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Divyan A Chopra
- Department of Pharmacology, Creighton University, Omaha, Nebraska, USA
| | | | - Matthew J Van Hook
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Fang Qiu
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - John Morrison
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Angie Rizzino
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Iqbal Ahmad
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
8
|
Pannicke T, Ivo Chao T, Reisenhofer M, Francke M, Reichenbach A. Comparative electrophysiology of retinal Müller glial cells-A survey on vertebrate species. Glia 2016; 65:533-568. [PMID: 27767232 DOI: 10.1002/glia.23082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/15/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022]
Abstract
Müller cells are the dominant macroglial cells in the retina of all vertebrates. They fulfill a variety of functions important for retinal physiology, among them spatial buffering of K+ ions and uptake of glutamate and other neurotransmitters. To this end, Müller cells express inwardly rectifying K+ channels and electrogenic glutamate transporters. Moreover, a lot of voltage- and ligand-gated ion channels, aquaporin water channels, and electrogenic transporters are expressed in Müller cells, some of them in a species-specific manner. For example, voltage-dependent Na+ channels are found exclusively in some but not all mammalian species. Whereas a lot of data exist from amphibians and mammals, the results from other vertebrates are sparse. It is the aim of this review to present a survey on Müller cell electrophysiology covering all classes of vertebrates. The focus is on functional studies, mainly performed using the whole-cell patch-clamp technique. However, data about the expression of membrane channels and transporters from immunohistochemistry are also included. Possible functional roles of membrane channels and transporters are discussed. Obviously, electrophysiological properties involved in the main functions of Müller cells developed early in vertebrate evolution. GLIA 2017;65:533-568.
Collapse
Affiliation(s)
- Thomas Pannicke
- Paul-Flechsig-Institut für Hirnforschung, Abteilung Pathophysiologie der Neuroglia, Universität Leipzig, Germany
| | - T Ivo Chao
- Institute of Anatomy and Cell Biology, Medical School Göttingen, Germany
| | - Miriam Reisenhofer
- Department of Chemistry, University of Zürich, Switzerland
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Mike Francke
- Paul-Flechsig-Institut für Hirnforschung, Abteilung Pathophysiologie der Neuroglia, Universität Leipzig, Germany
- Sächsischer Inkubator für klinische Translation (SIKT), Universität Leipzig, Germany
| | - Andreas Reichenbach
- Paul-Flechsig-Institut für Hirnforschung, Abteilung Pathophysiologie der Neuroglia, Universität Leipzig, Germany
| |
Collapse
|
9
|
Long Y, Bordt AS, Liu WS, Davis EP, Lee SJ, Tseng L, Chuang AZ, Whitaker CM, Massey SC, Sherman MB, Marshak DW. Wide-field diffuse amacrine cells in the monkey retina contain immunoreactive Cocaine- and Amphetamine-Regulated Transcript (CART). Peptides 2016; 84:22-35. [PMID: 27568514 PMCID: PMC5037056 DOI: 10.1016/j.peptides.2016.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 07/19/2016] [Accepted: 08/22/2016] [Indexed: 10/21/2022]
Abstract
The goals of this study were to localize the neuropeptide Cocaine- and Amphetamine-Regulated Transcript (CART) in primate retinas and to describe the morphology, neurotransmitter content and synaptic connections of the neurons that contain it. Using in situ hybridization, light and electron microscopic immunolabeling, CART was localized to GABAergic amacrine cells in baboon retinas. The CART-positive cells had thin, varicose dendrites that gradually descended through the inner plexiform layer and ramified extensively in the innermost stratum. They resembled two types of wide-field diffuse amacrine cells that had been described previously in macaque retinas using the Golgi method and also A17, serotonin-accumulating and waterfall cells of other mammals. The CART-positive cells received synapses from rod bipolar cell axons and made synapses onto the axons in a reciprocal configuration. The CART-positive cells also received synapses from other amacrine cells. Some of these were located on their primary dendrites, and the presynaptic cells there included dopaminergic amacrine cells. Although some CART-positive somas were localized in the ganglion cell layer, they did not contain the ganglion cell marker RNA binding protein with multiple splicing (RBPMS). Based on these results and electrophysiological studies in other mammals, the CART-positive amacrine cells would be expected to play a major role in the primary rod pathway of primates, providing feedback inhibition to rod bipolar cells.
Collapse
Affiliation(s)
- Ye Long
- Department of Neurobiology and Anatomy, McGovern Medical School, Houston, TX
| | - Andrea S. Bordt
- Department of Neurobiology and Anatomy, McGovern Medical School, Houston, TX
| | - Weiley S. Liu
- Department of Neurobiology and Anatomy, McGovern Medical School, Houston, TX
| | - Elizabeth P. Davis
- Department of Neurobiology and Anatomy, McGovern Medical School, Houston, TX
| | - Stephen J. Lee
- Department of Neurobiology and Anatomy, McGovern Medical School, Houston, TX
| | - Luke Tseng
- Department of Neurobiology and Anatomy, McGovern Medical School, Houston, TX
| | - Alice Z. Chuang
- Department of Ophthalmology and Visual Science, McGovern Medical School, Houston, TX
| | | | - Stephen C. Massey
- Department of Ophthalmology and Visual Science, McGovern Medical School, Houston, TX
| | - Michael B. Sherman
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX
| | - David W. Marshak
- Department of Neurobiology and Anatomy, McGovern Medical School, Houston, TX
- Department of Ophthalmology and Visual Science, McGovern Medical School, Houston, TX
| |
Collapse
|
10
|
Parameswaran S, Dravid SM, Teotia P, Krishnamoorthy RR, Qiu F, Toris C, Morrison J, Ahmad I. Continuous non-cell autonomous reprogramming to generate retinal ganglion cells for glaucomatous neuropathy. Stem Cells 2015; 33:1743-58. [PMID: 25753398 PMCID: PMC4524556 DOI: 10.1002/stem.1987] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/06/2015] [Indexed: 02/03/2023]
Abstract
Glaucoma, where the retinal ganglion cells (RGCs) carrying the visual signals from the retina to the visual centers in the brain are progressively lost, is the most common cause of irreversible blindness. The management approaches, whether surgical, pharmacological, or neuroprotective do not reverse the degenerative changes. The stem cell approach to replace dead RGCs is a viable option but currently faces several barriers, such as the lack of a renewable, safe, and ethical source of RGCs that are functional and could establish contacts with bona fide targets. To address these barriers, we have derived RGCs from the easily accessible adult limbal cells, reprogrammed to pluripotency by a non-nucleic acid approach, thus circumventing the risk of insertional mutagenesis. The generation of RGCs from the induced pluripotent stem (iPS) cells, also accomplished non-cell autonomously, recapitulated the developmental mechanism, ensuring the predictability and stability of the acquired phenotype, comparable to that of native RGCs at biochemical, molecular, and functional levels. More importantly, the induced RGCs expressed axonal guidance molecules and demonstrated the potential to establish contacts with specific targets. Furthermore, when transplanted in the rat model of ocular hypertension, these cells incorporated into the host RGC layer and expressed RGC-specific markers. Transplantation of these cells in immune-deficient mice did not produce tumors. Together, our results posit retinal progenitors generated from non-nucleic acid-derived iPS cells as a safe and robust source of RGCs for replacing dead RGCs in glaucoma.
Collapse
Affiliation(s)
- Sowmya Parameswaran
- Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE
| | | | - Pooja Teotia
- Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE
| | | | - Fang Qiu
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE
| | - Carol Toris
- Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE
| | - John Morrison
- Casey Eye Institute, Oregon Health & Science University, Portland, OR
| | - Iqbal Ahmad
- Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
11
|
|
12
|
NaV1.1 channels in axon initial segments of bipolar cells augment input to magnocellular visual pathways in the primate retina. J Neurosci 2013; 33:16045-59. [PMID: 24107939 DOI: 10.1523/jneurosci.1249-13.2013] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In the primate visual system, the ganglion cells of the magnocellular pathway underlie motion and flicker detection and are relatively transient, while the more sustained ganglion cells of the parvocellular pathway have comparatively lower temporal resolution, but encode higher spatial frequencies. Although it is presumed that functional differences in bipolar cells contribute to the tuning of the two pathways, the properties of the relevant bipolar cells have not yet been examined in detail. Here, by making patch-clamp recordings in acute slices of macaque retina, we show that the bipolar cells within the magnocellular pathway, but not the parvocellular pathway, exhibit voltage-gated sodium (NaV), T-type calcium (CaV), and hyperpolarization-activated, cyclic nucleotide-gated (HCN) currents, and can generate action potentials. Using immunohistochemistry in macaque and human retinae, we show that NaV1.1 is concentrated in an axon initial segment (AIS)-like region of magnocellular pathway bipolar cells, a specialization not seen in transient bipolar cells of other vertebrates. In contrast, CaV3.1 channels were localized to the somatodendritic compartment and proximal axon, but were excluded from the AIS, while HCN1 channels were concentrated in the axon terminal boutons. Simulations using a compartmental model reproduced physiological results and indicate that magnocellular pathway bipolar cells initiate spikes in the AIS. Finally, we demonstrate that NaV channels in bipolar cells augment excitatory input to parasol ganglion cells of the magnocellular pathway. Overall, the results demonstrate that selective expression of voltage-gated channels contributes to the establishment of parallel processing in the major visual pathways of the primate retina.
Collapse
|
13
|
Maturana MI, Kameneva T, Burkitt AN, Meffin H, Grayden DB. The effect of morphology upon electrophysiological responses of retinal ganglion cells: simulation results. J Comput Neurosci 2013; 36:157-75. [PMID: 23835760 PMCID: PMC3950609 DOI: 10.1007/s10827-013-0463-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 04/15/2013] [Accepted: 05/14/2013] [Indexed: 11/30/2022]
Abstract
Retinal ganglion cells (RGCs) display differences in their morphology and intrinsic electrophysiology. The goal of this study is to characterize the ionic currents that explain the behavior of ON and OFF RGCs and to explore if all morphological types of RGCs exhibit the phenomena described in electrophysiological data. We extend our previous single compartment cell models of ON and OFF RGCs to more biophysically realistic multicompartment cell models and investigate the effect of cell morphology on intrinsic electrophysiological properties. The membrane dynamics are described using the Hodgkin - Huxley type formalism. A subset of published patch-clamp data from isolated intact mouse retina is used to constrain the model and another subset is used to validate the model. Two hundred morphologically distinct ON and OFF RGCs are simulated with various densities of ionic currents in different morphological neuron compartments. Our model predicts that the differences between ON and OFF cells are explained by the presence of the low voltage activated calcium current in OFF cells and absence of such in ON cells. Our study shows through simulation that particular morphological types of RGCs are capable of exhibiting the full range of phenomena described in recent experiments. Comparisons of outputs from different cells indicate that the RGC morphologies that best describe recent experimental results are ones that have a larger ratio of soma to total surface area.
Collapse
Affiliation(s)
- Matias I Maturana
- Centre for Neural Engineering, University of Melbourne, 203 Bouverie St, Carlton, Vic, 3053, Australia
| | | | | | | | | |
Collapse
|
14
|
Yang XF, Miao Y, Ping Y, Wu HJ, Yang XL, Wang Z. Melatonin inhibits tetraethylammonium-sensitive potassium channels of rod ON type bipolar cells via MT2 receptors in rat retina. Neuroscience 2010; 173:19-29. [PMID: 21094224 DOI: 10.1016/j.neuroscience.2010.11.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 10/29/2010] [Accepted: 11/13/2010] [Indexed: 11/16/2022]
Abstract
By challenging specific receptors, melatonin synthesized and released by photoreceptors regulates various physiological functions in the vertebrate retina. Here, we studied modulatory effects of melatonin on K+ currents of rod-dominant ON type bipolar cells (Rod-ON-BCs) in rat retinal slices by patch-clamp techniques. Double immunofluorescence experiments conducted in isolated cell and retinal section preparations showed that the melatonin MT₂ receptor was expressed in somata, dendrites and axon terminals of rat Rod-ON-BCs. Electrophysiologically, application of melatonin selectively inhibited the tetraethylammonium (TEA)-sensitive K+ current component, but did not show any effect on the 4-aminopyridine (4-AP)-sensitive component. Consistent with the immunocytochemical result, the melatonin effect was blocked by co-application of 4-phenyl-2-propionamidotetralin (4-P-PDOT), a specific MT₂ receptor antagonist. Neither protein kinase A (PKA) nor protein kinase G (PKG) seemed to be involved because both the PKA inhibitor Rp-cAMP and the PKG inhibitor KT5823 did not block the melatonin-induced suppression of the K+ currents. In contrast, application of the phospholipase C (PLC) inhibitor U73122 or the protein kinase C (PKC) inhibitor bisindolylmaleimide IV (Bis IV) eliminated the melatonin effect, and when the Ca²+ chelator BAPTA-containing pipette was used, melatonin failed to inhibit the K+ currents. These results suggest that suppression of the TEA-sensitive K+ current component via activation of MT₂ receptors expressed on rat Rod-ON-BCs may be mediated by a Ca²+-dependent PLC/inositol 1,4,5-trisphosphate (IP₃/PKC signaling pathway.
Collapse
Affiliation(s)
- X-F Yang
- Institutes of Brain Science, Institute of Neurobiology and State Key Laboratory of Medical Neurobiology, Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | | | | | | | | | | |
Collapse
|
15
|
Moriondo A, Rispoli G. The contribution of cationic conductances to the potential of rod photoreceptors. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2009; 39:889-902. [PMID: 19234695 DOI: 10.1007/s00249-009-0419-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2008] [Revised: 01/27/2009] [Accepted: 02/02/2009] [Indexed: 11/29/2022]
Abstract
The contribution of cationic conductances in shaping the rod photovoltage was studied in light adapted cells recorded under whole-cell voltage- or current-clamp conditions. Depolarising current steps (of size comparable to the light-regulated current) produced monotonic responses when the prepulse holding potential (V (h)) was -40 mV (i.e. corresponding to the membrane potential in the dark). At V (h) = -60 mV (simulating the steady-state response to an intense background of light) current injections <35 pA (mimicking a light decrement) produced instead an initial depolarisation that declined to a plateau, and voltage transiently overshot V (h) at the stimulus offset. Current steps >40 pA produced a steady depolarisation to approximately -16 mV at both V (h). The difference between the responses at the two V (h) was primarily generated by the slow delayed-rectifier-like K(+) current (I (Kx)), which therefore strongly affects both the photoresponse rising and falling phase. The steady voltage observed at both V (h) in response to large current injections was instead generated by Ca-activated K(+) channels (I (KCa)), as previously found. Both I (Kx) and I (KCa) oppose the cation influx, occurring at the light stimulus offset through the cGMP-gated channels and the voltage-activated Ca(2+) channels (I (Ca)). This avoids that the cation influx could erratically depolarise the rod past its normal resting value, thus allowing a reliable dim stimuli detection, without slowing down the photovoltage recovery kinetics. The latter kinetics was instead accelerated by the hyperpolarisation-activated, non-selective current (I (h)) and I (Ca). Blockade of all K(+) currents with external TEA unmasked a I (Ca)-dependent regenerative behaviour.
Collapse
Affiliation(s)
- Andrea Moriondo
- Dipartimento di Biologia ed Evoluzione, Sezione di Fisiologia e Biofisica, National Institute of Neuroscience and Neuroscience Center, Università di Ferrara, Via L. Borsari 46, Ferrara, Italy
| | | |
Collapse
|
16
|
Yu YC, Satoh H, Wu SM, Marshak DW. Histamine enhances voltage-gated potassium currents of ON bipolar cells in macaque retina. Invest Ophthalmol Vis Sci 2008; 50:959-65. [PMID: 18836167 DOI: 10.1167/iovs.08-2746] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The goal was to understand the functions of retinopetal axons containing histamine. In prior work, type 3 histamine receptors (HR3) have been localized to the tips of ON bipolar cell dendrites in macaque retinas. Voltage-gated potassium channels have also been localized to bipolar cell dendrites, and the hypothesis tested in the present study was that these are modulated by histamine. METHODS Whole-cell recordings of potassium currents were made from bipolar cells in slice preparations of macaque retina. In voltage-clamp mode, the cells were held at -60 mV and stepped to values from -60 to 80 mV. Recordings of the membrane potential were also made in current-clamp mode. Histamine, the HR3 agonist (R) alpha-methylhistamine (RAMH), tetraethyl ammonium (TEA), and 4-aminopyridine (4-AP) were applied in the superfusate. RESULTS Histamine produced a dose-dependent increase in potassium currents in a subset of bipolar cells. At 5 microM, histamine increased the currents by 15% or more in the ON bipolar cells but not in the OFF bipolar cells. RAMH at 5 microM increased the amplitude of the potassium currents in the ON bipolar cells. In 10 mM TEA, potassium currents were reduced in all the bipolar cells, and there was no effect of histamine. Histamine hyperpolarized the resting membrane potential of the ON bipolar cells by 5 mV. CONCLUSIONS By enhancing potassium currents in the ON bipolar cells, histamine is expected to reduce the amplitude of the light responses and limit their duration. The hyperpolarization of the resting membrane potential would also reduce neurotransmitter release at their output synapses.
Collapse
Affiliation(s)
- Yong-Chun Yu
- Department of Neurobiology and Anatomy, University of Texas Medical School, Houston, Texas 77225, USA
| | | | | | | |
Collapse
|
17
|
Giovannelli A, Di Marco S, Maccarone R, Bisti S. Long-term dark rearing induces permanent reorganization in retinal circuitry. Biochem Biophys Res Commun 2007; 365:349-54. [PMID: 17999915 DOI: 10.1016/j.bbrc.2007.10.204] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Accepted: 10/31/2007] [Indexed: 11/15/2022]
Abstract
Recent data challenged the assumption that light has little effect on retina development. Here, we report evidence that dark rearing permanently changes the synaptic input to GCs. A reduced spontaneous postsynaptic currents (SPSCs) frequency was found in retinal GCs from rats born and raised in the dark for three months. Glutamate antagonists (CNQX and AP-5) reversibly reduced SPSCs frequency in control and dark-reared (DR) retinae. The GABA antagonist picrotoxin (PTX) reduced SPSCs frequency in control retinas, but increased SPSCs frequency in DR, mainly by presynaptic action on excitatory currents. In DR animals exposed to normal cyclic light for 3 months, SPSCs frequency remained lower then in control rats and increased following PTX, suggesting that long-term dark rearing induces permanent modifications of the retinal circuitry. Our results strongly support the idea that light stimulation plays a role in establishing normal synaptic input to GCs.
Collapse
Affiliation(s)
- Aldo Giovannelli
- Dipartimento di Medicina Sperimentale, Università di L'Aquila, via Vetoio, Coppito 2, L'Aquila 67100, Italy.
| | | | | | | |
Collapse
|
18
|
Ohkuma M, Kawai F, Horiguchi M, Miyachi EI. Patch-clamp recording of human retinal photoreceptors and bipolar cells. Photochem Photobiol 2007; 83:317-22. [PMID: 16995772 DOI: 10.1562/2006-06-15-ra-923] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Photoreceptors and retinal bipolar cells are considered as nonspiking neurons; however, we recently showed that human rod photoreceptors can generate sodium action potentials in response to membrane depolarization from membrane potentials of -60 or -70 mV (Kawai et al., Neuron 30 [2001] 451). We performed patch-clamp recording of human cone photoreceptors and retinal bipolar cells to examine whether functional voltage-gated sodium channels are expressed in these cells as well as rod photoreceptors. Under current-clamp conditions, the injection of depolarizing current steps into a cone photoreceptor-induced marked action potentials. These action potentials were blocked by 1 microM tetrodotoxin, a voltage-gated sodium channel blocker. Under voltage-clamp conditions, depolarizing voltage steps-induced a fast transient inward current in several bipolar cells (n = 4/78). This current was activated from -70 to + 20 mV (maximal at -10 mV) and inactivated within 5 ms. The 10-90% rise time of this current was shorter than another inward current (less than one-hundredth). These results indicate that human cones and bipolar cells express voltage-gated sodium channels as rod photoreceptors. Sodium channels may serve to amplify the release of a neurotransmitter and to accelerate the light-dark change in photosignals.
Collapse
Affiliation(s)
- Mahito Ohkuma
- Department of Physiology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan.
| | | | | | | |
Collapse
|
19
|
Shen W. Repetitive light stimulation inducing glycine receptor plasticity in the retinal neurons. J Neurophysiol 2005; 94:2231-8. [PMID: 16105957 DOI: 10.1152/jn.01099.2004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neurotransmitter receptor plasticity is a mechanism that can regulate the temporal and intensity encoding of a synapse. While this has been extensively studied as a mechanism of learning, less is known about such processes in sensory systems. This study examines modulation of glycine receptor function at the first synapse in the retina. It was found that horizontal cells, which are postsynaptic to photoreceptors, have glycine receptor currents that are enhanced when internal calcium is elevated. This can be achieved by glutamatergic synaptic input or by activation of voltage-gated calcium channels. When the retina was maintained in a dark-adapted state, the calcium levels in horizontal cells were relatively low. After a series of brief light stimuli, the internal calcium concentration in horizontal cells was elevated, and the glycine currents were faster and greater in amplitude. The increase of internal calcium levels was caused by increased transmitter release from photoreceptors. Thus glycine receptor function is state dependent and can be rapidly altered by synaptic input from photoreceptors. Light stimulation drives glycine receptor plasticity in the retinal neural network.
Collapse
Affiliation(s)
- Wen Shen
- Department of Biomedical Science, Florida Atlantic Univ., Bldg. BC-71, Rm. 229, 777 Glades Rd., Boca Raton, Florida 33431, USA.
| |
Collapse
|
20
|
Ugarte G, Delgado R, O'Day PM, Farjah F, Cid LP, Vergara C, Bacigalupo J. Putative ClC-2 Chloride Channel Mediates Inward Rectification in Drosophila Retinal Photoreceptors. J Membr Biol 2005; 207:151-60. [PMID: 16550486 DOI: 10.1007/s00232-005-0810-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2005] [Revised: 10/06/2005] [Indexed: 10/24/2022]
Abstract
We report that Drosophila retinal photoreceptors express inwardly rectifying chloride channels that seem to be orthologous to mammalian ClC-2 inward rectifier channels. We measured inwardly rectifying Cl(-) currents in photoreceptor plasma membranes: Hyperpolarization under whole-cell tight-seal voltage clamp induced inward Cl(-) currents; and hyperpolarization of voltage-clamped inside-out patches excised from plasma membrane induced Cl(-) currents that have a unitary channel conductance of approximately 3.7 pS. The channel was inhibited by 1 mM: Zn(2+) and by 1 mM: 9-anthracene, but was insensitive to DIDS. Its anion permeability sequence is Cl(-) = SCN(-)> Br(-)>> I(-), characteristic of ClC-2 channels. Exogenous polyunsaturated fatty acid, linolenic acid, enhanced or activated the inward rectifier Cl(-) currents in both whole-cell and excised patch-clamp recordings. Using RT-PCR, we found expression in Drosophila retina of a ClC-2 gene orthologous to mammalian ClC-2 channels. Antibodies to rat ClC-2 channels labeled Drosophila photoreceptor plasma membranes and synaptic regions. Our results provide evidence that the inward rectification in Drosophila retinal photoreceptors is mediated by ClC-2-like channels in the non-transducing (extra-rhabdomeral) plasma membrane, and that this inward rectification can be modulated by polyunsaturated fatty acid.
Collapse
Affiliation(s)
- G Ugarte
- Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|
21
|
Han Y, Massey SC. Electrical synapses in retinal ON cone bipolar cells: subtype-specific expression of connexins. Proc Natl Acad Sci U S A 2005; 102:13313-8. [PMID: 16150718 PMCID: PMC1201596 DOI: 10.1073/pnas.0505067102] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2005] [Accepted: 07/29/2005] [Indexed: 11/18/2022] Open
Abstract
Retinal bipolar cells are known to form a complex, interconnecting network through electrical synapses that are either heterologous (with amacrine cells) or homologous (with other bipolar cells). These electrical synapses can be functionally as important as chemical synapses because their distinct properties provide a different character for the network. Much less is known, however, about electrical synapses in retinal bipolar cells than about chemical synapses. Here we report the molecular basis for electrical synapses in retinal bipolar cells, particularly ON cone bipolar cells. We have found variable connexin 36 (cx36) expression in different types of ON cone bipolar cells: cx36 message was found in some, but not all, ON cone bipolar cells (4 of 14 cells). In one specific type of ON cone bipolar cell (BPGus-GFP), however, cx36 was detected in 17 of 19 cells. Moreover, we have located cx36 puncta at the axonal terminals of BPGus-GFP cells, and we have found that these BPGus-GFP-associated cx36 puncta always colocalized with AII amacrine cell processes. Molecular and immunocytochemical evidence obtained in this study also shows that connexin 45 (cx45) is not present in BPGus-GFP cells. Taken together, our results suggest that connexins are expressed in bipolar cells in a neuronal subtype-specific manner and that cx36/cx36 gap junctions form the heterologous electrical synapses between AII amacrine cells and BPGus-GFP cells. Our findings imply that visual information can be differently processed by distinct subtypes of ON cone bipolar cells via electrical synapses.
Collapse
Affiliation(s)
- Yi Han
- Department of Ophthalmology and Visual Sciences, University of Texas Health Science Center, Houston, TX 77030, USA
| | | |
Collapse
|
22
|
Pannicke T, Biedermann B, Uckermann O, Weick M, Bringmann A, Wolf S, Wiedemann P, Habermann G, Buse E, Reichenbach A. Physiological properties of retinal Muller glial cells from the cynomolgus monkey, Macaca fascicularis--a comparison to human Muller cells. Vision Res 2005; 45:1781-91. [PMID: 15797768 DOI: 10.1016/j.visres.2005.01.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2004] [Revised: 01/11/2005] [Accepted: 01/19/2005] [Indexed: 11/16/2022]
Abstract
Retinae from rabbits and laboratory rodents are often used as 'models' of the human retina, although there are anatomical differences. To test whether monkey eyes provide a better model, a physiological study of Muller glial cells was performed comparing isolated cells and retinal wholemounts from the cynomolgus monkey, Macaca fascicularis and from man. The membrane conductance of Muller cells from both species was dominated by inward and outward K(+) currents. Cells displayed glutamate uptake currents and responded to nucleotides by intracellular Ca(2+) increases. However, there were also species differences, such as a lack of GABA(A) receptors and of Ca(2+)-dependent K(+) currents in monkey cells. Thus, the use of Muller cells from cynomolgus monkeys may be advantageous for investigating a few specific properties; in general, monkey cells are no more similar to human cells than those from standard laboratory animals.
Collapse
Affiliation(s)
- Thomas Pannicke
- Paul-Flechsig-Institut für Hirnforschung, Universität Leipzig, Jahnallee 59, D-04109 Leipzig, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Ma YP, Cui J, Pan ZH. Heterogeneous expression of voltage-dependent Na+ and
K+ channels in mammalian retinal bipolar cells. Vis Neurosci 2005; 22:119-33. [PMID: 15935105 DOI: 10.1017/s0952523805222010] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2004] [Indexed: 11/05/2022]
Abstract
Retinal bipolar cells show heterogeneous expression of
voltage-dependent Na+ and K+ currents. We used
whole-cell patch-clamp recordings to investigate the possible roles of
these currents in the response properties of bipolar cells in rats.
Isolated bipolar cells showed robust spontaneous regenerative activity,
but the regenerative potential of rod bipolar cells reached a more
depolarized level than that of cone bipolar cells. In both isolated cells
and cells in retinal slices, the membrane depolarization evoked by current
injection was apparently capped. The evoked membrane potential was again
more depolarized in rod bipolar cells than in cone bipolar cells.
Application of tetraethylammonium and 4-aminopyridine shifted the
spontaneous regenerative potential as well as the evoked potential to a
more depolarized level. In addition, a subclass of cone bipolar cells
showed a prominent spike in the initial phase of the voltage response when
the cells were depolarized from a relatively negative membrane potential.
The spike was mediated mainly by tetrodotoxin-sensitive Na+
current. The presence of the spike sped up the response kinetics and
enhanced the peak membrane potential. Results of this study raise the
possibility that voltage-dependent K+ currents may play a role
in defining different membrane operating ranges of rod and cone bipolar
cells and that voltage-dependent Na+ currents may enhance the
response kinetics and amplitude of certain cone bipolar cells.
Collapse
Affiliation(s)
- Yu-Ping Ma
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | |
Collapse
|
24
|
Cia D, Bordais A, Varela C, Forster V, Sahel JA, Rendon A, Picaud S. Voltage-Gated Channels and Calcium Homeostasis in Mammalian Rod Photoreceptors. J Neurophysiol 2005; 93:1468-75. [PMID: 15483058 DOI: 10.1152/jn.00874.2004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent reports on rod photoreceptor neuroprotection by Ca2+channel blockers have pointed out the need to assess the effect of these blockers on mammalian rods. However, in mammals, rod electrophysiological characterization has been hampered by the small size of these photoreceptors, which were instead extensively studied in nonmammalian vertebrates. To further characterize ionic conductances and to assess the pharmacology of Ca2+channels in mammalian rods, freshly dissociated pig rod photoreceptors were recorded with the whole cell patch-clamp technique. Rod cells expressed 1) a hyperpolarization-activated inward-rectifying conductance ( Ih) sensitive to external Cs+; 2) a sustained outward K+current ( IK) sensitive to tetraethylammonium; 3) a sustained voltage-gated Ca2+current ( ICa) sensitive to benzothiazepine (diltiazem) and phenylalkylamine (verapamil) derivatives; 4) a Ca2+-activated Cl−current ( ICl(Ca)); and 5) a plasma membrane Ca2+-ATPase. The Ca2+current showed a range of activation from positive potentials to –60 mV with a maximum between –30 and –20 mV. In contrast to other L-type Ca2+channels, rod Ca2+channels were blocked at similar and relatively high concentrations by the diltiazem isomers and verapamil. The biphasic dose-response for d-diltiazem confirmed the low sensitivity of Ca2+channels for the molecule. The ATPase, which was localized at the axon terminal, was found to contribute to Ca2+extrusion. These results suggest that the electrophysiological features of rod photoreceptors had been preserved during evolution from nonmammalian vertebrates to mammals. This work indicates further that mammalian rods express nonclassic L-type Ca2+channels, showing a low sensitivity to the diltiazem isomers used in neuroprotective studies.
Collapse
Affiliation(s)
- David Cia
- Laboratoire de Physiopathologie Cellulaire et Moléculaire de la Rétine, Institut National de la Santé et de la Recherche Médicale U-592, Université Pierre et Marie Curie
| | | | | | | | | | | | | |
Collapse
|
25
|
Xiao J, Cai Y, Yen J, Steffen M, Baxter DA, Feigenspan A, Marshak D. Voltage-clamp analysis and computational model of dopaminergic
neurons from mouse retina. Vis Neurosci 2005; 21:835-49. [PMID: 15733339 DOI: 10.1017/s0952523804216042] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2004] [Indexed: 11/05/2022]
Abstract
Isolated dopaminergic amacrine (DA) cells in mouse retina fire
rhythmic, spontaneous action potentials and respond to depolarizing
current with trains of low-frequency action potentials. To investigate
the roles of voltage-gated ion channels in these processes, the
transient A-type K+ current (IK,A) and
Ca2+ current (ICa) in isolated mouse DA cells
were analyzed by voltage clamp. The IK,A activated at
−60 mV and inactivated rapidly. ICa activated at
around −30 mV and reached a peak at 10 mV without apparent
inactivation. We also extended our previous computational model of the
mouse DA cell to include the new electrophysiological data. The model
consisted of a membrane capacitance in parallel with eight currents:
Na+ transient (INa,T), Na+ persistent
(INa,P), delayed rectifier potassium (IKdr),
IK,A, calcium-dependent potassium (IK,Ca), L-type
Ca2+ ICa, hyperpolarization-activated cation
current (Ih), and a leak current (IL).
Hodgkin-Huxley type equations were used to define the voltage- and
time-dependent activation and inactivation. The simulations were
implemented using the neurosimulator SNNAP. The model DA cell was
spontaneously active from a wide range of initial membrane potentials.
The spontaneous action potentials reached 35 mV at the peak and
hyperpolarized to −76 mV between spikes. The spontaneous firing
frequency in the model was 6 Hz. The model DA cell responded to
prolonged depolarizing current injection by increasing its spiking
frequency and eventually reaching a depolarization block at membrane
potentials greater than −10 mV. The most important current for
determining the firing rate was IK,A. When the amplitude of
IK,A was decreased, the firing rate increased.
ICa and IK,Ca also affected the width of action
potentials but had only minor effects on the firing rate. Ih
affected the firing rate slightly but did not change the waveform of
the action potentials.
Collapse
Affiliation(s)
- Jianguo Xiao
- Department of Neurobiology and Anatomy, The University of Texas-Houston Medical School, Houston, TX 77225, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Shen W, Finnegan S, Lein P, Sullivan S, Slaughter M, Higgins D. Bone morphogenetic proteins regulate ionotropic glutamate receptors in human retina. Eur J Neurosci 2004; 20:2031-7. [PMID: 15450082 DOI: 10.1111/j.1460-9568.2004.03681.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Bone morphogenetic proteins (BMPs) are required for the development of retina, but their role in the mature eye is unknown. We therefore examined the expression of BMP-7 in adult human retina and assessed its effects on horizontal cells cultured from adult human retina. BMP-7 expression was detected in all retinal layers, with high levels of expression being present in the inner and outer nuclear layers. Human horizontal cells, found in the inner nuclear layer, possess both AMPA and kainate receptors, and glutamatergic agonists that activate these receptors induce prominent inward currents. Exposure to BMP-7 suppresses the kainate receptor current but enhances the AMPA receptor current. BMP-6, activin, and cartilage-derived morphogenic protein-2 (CDMP-2) have similar effects to BMP-7 and act just as rapidly (< 1 s). In contrast BMP-2 and transforming growth factor-beta2 are inactive. The actions of BMP-7 on both AMPA and kainate receptors were blocked by the nonselective kinase inhibitor, staurosporine. In contrast, the serine/threonine kinase inhibitors blocked only the effects of BMP-7 on the AMPA current. Thus, BMPs rapidly and differentially regulate two ionotropic glutamate receptors through distinct pathways, neither of which involves nuclear regulatory activity. These observations suggest that BMPs might modify synaptic function in the mature nervous system.
Collapse
Affiliation(s)
- Wen Shen
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Glutamate receptor currents were examined in horizontal cells from cultured human retina using whole-cell recording procedures. Horizontal cells possess both AMPA and kainate receptors and both produce significant sustained currents. The kainate-induced current did not show significant desensitization and was not enhanced by concanavalin A. The sustained AMPA current was smaller than the kainate current, but the difference was almost entirely due to pronounced desensitization. The horizontal cell AMPA current was enhanced by cyclothiazide but not by PEPA, indicating the presence of the flip receptor variant. GYKI-52466 blocked the AMPA response (IC50= 5 μM against 100 μM AMPA) but also blocked the kainate response (IC50= 45 μM against 100 μM kainate). The diversity of glutamate receptors in human horizontal cells suggests that synaptic input to these neurons may be multiplexed through both kainate and AMPA channels.
Collapse
MESH Headings
- Benzodiazepines/pharmacology
- Chlorothiazide/pharmacology
- Concanavalin A/pharmacology
- Culture Techniques
- Electric Conductivity
- Excitatory Amino Acid Agonists/pharmacology
- Excitatory Amino Acid Antagonists/pharmacology
- Humans
- Kainic Acid/pharmacology
- Neurons/drug effects
- Neurons/metabolism
- Patch-Clamp Techniques
- Receptors, AMPA/antagonists & inhibitors
- Receptors, AMPA/metabolism
- Receptors, AMPA/physiology
- Receptors, GABA/metabolism
- Receptors, GABA/physiology
- Receptors, Glutamate/metabolism
- Receptors, Glutamate/physiology
- Receptors, Kainic Acid/antagonists & inhibitors
- Receptors, Kainic Acid/metabolism
- Receptors, Kainic Acid/physiology
- Retina/cytology
- Retina/metabolism
- alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
Collapse
Affiliation(s)
- Wen Shen
- Department of Physiology and Biophysics, University at Buffalo, Buffalo, NY 14214, USA
| | | | | |
Collapse
|