1
|
Barta CL, Thoreson WB. Retinal inputs that drive optomotor responses of mice under mesopic conditions. IBRO Neurosci Rep 2024; 17:138-144. [PMID: 39170059 PMCID: PMC11338136 DOI: 10.1016/j.ibneur.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/11/2024] [Accepted: 07/20/2024] [Indexed: 08/23/2024] Open
Abstract
Optomotor responses are a popular way to assess sub-cortical visual responses in mice. We studied photoreceptor inputs into optomotor circuits using genetically-modified mice lacking the exocytotic calcium sensors synaptotagmin 1 (Syt1) and 7 (Syt7) in rods or cones. We also tested mice that in which cone transducin, GNAT2, had been eliminated. We studied spatial frequency sensitivity under mesopic conditions by varying the spatial frequency of a grating rotating at 12 deg/s and contrast sensitivity by varying luminance contrast of 0.2c/deg gratings. We found that eliminating Syt1 from rods reduced responses to a low spatial frequency grating (0.05c/deg) consistent with low resolution in this pathway. Conversely, eliminating the ability of cones to respond to light (by eliminating GNAT2) or transmit light responses (by selectively eliminating Syt1) showed weaker responses to a high spatial frequency grating (3c/deg). Eliminating Syt7 from the entire optomotor pathway in a global knockout had no significant effect on optomotor responses. We isolated the secondary rod pathway involving transmission of rod responses to cones via gap junctions by simultaneously eliminating Syt1 from rods and GNAT2 from cones. We found that the secondary rod pathway is sufficient to drive robust optomotor responses under mesopic conditions. Finally, eliminating Syt1 from both rods and cones almost completely abolished optomotor responses, but we detected weak responses to large, bright rotating gratings that are likely driven by input from intrinsically photosensitive retinal ganglion cells.
Collapse
Affiliation(s)
- CL Barta
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, USA
| | - WB Thoreson
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, USA
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
2
|
Mesnard CS, Hays CL, Townsend LE, Barta CL, Gurumurthy CB, Thoreson WB. Synaptotagmin-9 in mouse retina. Vis Neurosci 2024; 41:E003. [PMID: 39291699 PMCID: PMC11417998 DOI: 10.1017/s0952523824000026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/18/2024] [Accepted: 05/14/2024] [Indexed: 09/19/2024]
Abstract
Synaptotagmin-9 (Syt9) is a Ca2+ sensor mediating fast synaptic release expressed in various parts of the brain. The presence and role of Syt9 in retina is unknown. We found evidence for Syt9 expression throughout the retina and created mice to conditionally eliminate Syt9 in a cre-dependent manner. We crossed Syt9fl/fl mice with Rho-iCre, HRGP-Cre, and CMV-Cre mice to generate mice in which Syt9 was eliminated from rods (rodSyt9CKO), cones (coneSyt9CKO), or whole animals (CMVSyt9). CMVSyt9 mice showed an increase in scotopic electroretinogram (ERG) b-waves evoked by bright flashes with no change in a-waves. Cone-driven photopic ERG b-waves were not significantly different in CMVSyt9 knockout mice and selective elimination of Syt9 from cones had no effect on ERGs. However, selective elimination from rods decreased scotopic and photopic b-waves as well as oscillatory potentials. These changes occurred only with bright flashes where cone responses contribute. Synaptic release was measured in individual rods by recording anion currents activated by glutamate binding to presynaptic glutamate transporters. Loss of Syt9 from rods had no effect on spontaneous or depolarization-evoked release. Our data show that Syt9 acts at multiple sites in the retina and suggest that it may play a role in regulating transmission of cone signals by rods.
Collapse
Affiliation(s)
- Chris S. Mesnard
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cassandra L. Hays
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Medical Education, Creighton University, Omaha, NE, USA
| | - Lou E. Townsend
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cody L. Barta
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Wallace B. Thoreson
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
3
|
Kumar D, Khan B, Okcay Y, Sis ÇÖ, Abdallah A, Murray F, Sharma A, Uemura M, Taliyan R, Heinbockel T, Rahman S, Goyal R. Dynamic endocannabinoid-mediated neuromodulation of retinal circadian circuitry. Ageing Res Rev 2024; 99:102401. [PMID: 38964508 DOI: 10.1016/j.arr.2024.102401] [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: 10/04/2023] [Revised: 06/05/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Circadian rhythms are biological rhythms that originate from the "master circadian clock," called the suprachiasmatic nucleus (SCN). SCN orchestrates the circadian rhythms using light as a chief zeitgeber, enabling humans to synchronize their daily physio-behavioral activities with the Earth's light-dark cycle. However, chronic/ irregular photic disturbances from the retina via the retinohypothalamic tract (RHT) can disrupt the amplitude and the expression of clock genes, such as the period circadian clock 2, causing circadian rhythm disruption (CRd) and associated neuropathologies. The present review discusses neuromodulation across the RHT originating from retinal photic inputs and modulation offered by endocannabinoids as a function of mitigation of the CRd and associated neuro-dysfunction. Literature indicates that cannabinoid agonists alleviate the SCN's ability to get entrained to light by modulating the activity of its chief neurotransmitter, i.e., γ-aminobutyric acid, thus preventing light-induced disruption of activity rhythms in laboratory animals. In the retina, endocannabinoid signaling modulates the overall gain of the retinal ganglion cells by regulating the membrane currents (Ca2+, K+, and Cl- channels) and glutamatergic neurotransmission of photoreceptors and bipolar cells. Additionally, endocannabinoids signalling also regulate the high-voltage-activated Ca2+ channels to mitigate the retinal ganglion cells and intrinsically photosensitive retinal ganglion cells-mediated glutamate release in the SCN, thus regulating the RHT-mediated light stimulation of SCN neurons to prevent excitotoxicity. As per the literature, cannabinoid receptors 1 and 2 are becoming newer targets in drug discovery paradigms, and the involvement of endocannabinoids in light-induced CRd through the RHT may possibly mitigate severe neuropathologies.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, HP 173229, India.
| | - Bareera Khan
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, HP 173229, India
| | - Yagmur Okcay
- University of Health Sciences Gulhane Faculty of Pharmacy Department of Pharmacology, Turkey.
| | - Çağıl Önal Sis
- University of Health Sciences Gulhane Faculty of Pharmacy Department of Pharmacology, Turkey.
| | - Aya Abdallah
- Institute of Medical Science, University of Aberdeen, Aberdeen, Scotland.
| | - Fiona Murray
- Institute of Medical Science, University of Aberdeen, Aberdeen, Scotland.
| | - Ashish Sharma
- School of Medicine, Washington University, St. Louis, USA
| | - Maiko Uemura
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology Science, Pilani, Rajasthan 333301, India.
| | - Thomas Heinbockel
- Howard University College of Medicine, Department of Anatomy, Washington, DC 20059, USA
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy South Dakota State University, Brookings, SD, USA.
| | - Rohit Goyal
- Department of Neuropharmacology, School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, HP 173229, India.
| |
Collapse
|
4
|
Wagner N, Tsai T, Reinehr S, Theile J, Dick HB, Joachim SC. Retinal debris triggers cytotoxic damage in cocultivated primary porcine RPE cells. Front Neurosci 2024; 18:1401571. [PMID: 39114482 PMCID: PMC11303199 DOI: 10.3389/fnins.2024.1401571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/12/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction One of the most common causes of vision loss in the elderly population worldwide is age-related macular degeneration (AMD). Subsequently, the number of people affected by AMD is estimated to reach approximately 288 million by the year 2040. The aim of this study was to develop an ex vivo model that simulates various aspects of the complex AMD pathogenesis. Methods For this purpose, primary porcine retinal pigment epithelial cells (ppRPE) were isolated and cultured. One group was exposed to medium containing sodium iodate (NaIO3) to induce degeneration. The others were exposed to different supplemented media, such as bovine serum albumin (BSA), homogenized porcine retinas (HPR), or rod outer segments (ROOS) for eight days to promote retinal deposits. Then, these ppRPE cells were cocultured with porcine neuroretina explants for another eight days. To assess the viability of ppRPE cells, live/dead assay was performed at the end of the study. The positive RPE65 and ZO1 area was evaluated by immunocytochemistry and the expression of RLBP1, RPE65, and TJP1 was analyzed by RT-qPCR. Additionally, drusen (APOE), inflammation (ITGAM, IL6, IL8, NLRP3, TNF), oxidative stress (NFE2L2, SOD1, SOD2), and hypoxia (HIF1A) markers were investigated. The concentration of the inflammatory cytokines IL-6 and IL-8 was determined in medium supernatants from day 16 and 24 via ELISA. Results Live/dead assay suggests that especially exposure to NaIO3 and HPR induced damage to ppRPE cells, leading in a significant ppRPE cell loss. All supplemented media resulted in decreased RPE-characteristic markers (RPE65; ZO-1) and gene expression like RLBP1 and RPE65 in the cultured ppRPE cells. Besides, some inflammatory, oxidative as well as hypoxic stress markers were altered in ppRPE cells cultivated with NaIO3. The application of HPR induced an enhanced APOE expression. Pre-exposure of the ppRPE cells led to a diminished number of cones in all supplemented media groups compared to controls. Discussion Overall, this novel coculture model represents an interesting initial approach to incorporating deposits into coculture to mimic AMD pathogenesis. Nevertheless, the effects of the media used need to be investigated in further studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Stephanie C. Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
5
|
Stoddart PR, Begeng JM, Tong W, Ibbotson MR, Kameneva T. Nanoparticle-based optical interfaces for retinal neuromodulation: a review. Front Cell Neurosci 2024; 18:1360870. [PMID: 38572073 PMCID: PMC10987880 DOI: 10.3389/fncel.2024.1360870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 03/04/2024] [Indexed: 04/05/2024] Open
Abstract
Degeneration of photoreceptors in the retina is a leading cause of blindness, but commonly leaves the retinal ganglion cells (RGCs) and/or bipolar cells extant. Consequently, these cells are an attractive target for the invasive electrical implants colloquially known as "bionic eyes." However, after more than two decades of concerted effort, interfaces based on conventional electrical stimulation approaches have delivered limited efficacy, primarily due to the current spread in retinal tissue, which precludes high-acuity vision. The ideal prosthetic solution would be less invasive, provide single-cell resolution and an ability to differentiate between different cell types. Nanoparticle-mediated approaches can address some of these requirements, with particular attention being directed at light-sensitive nanoparticles that can be accessed via the intrinsic optics of the eye. Here we survey the available known nanoparticle-based optical transduction mechanisms that can be exploited for neuromodulation. We review the rapid progress in the field, together with outstanding challenges that must be addressed to translate these techniques to clinical practice. In particular, successful translation will likely require efficient delivery of nanoparticles to stable and precisely defined locations in the retinal tissues. Therefore, we also emphasize the current literature relating to the pharmacokinetics of nanoparticles in the eye. While considerable challenges remain to be overcome, progress to date shows great potential for nanoparticle-based interfaces to revolutionize the field of visual prostheses.
Collapse
Affiliation(s)
- Paul R. Stoddart
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - James M. Begeng
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC, Australia
- Department of Biomedical Engineering, Faculty of Engineering & Information Technology, The University of Melbourne, Melbourne, VIC, Australia
| | - Wei Tong
- Department of Biomedical Engineering, Faculty of Engineering & Information Technology, The University of Melbourne, Melbourne, VIC, Australia
- School of Physics, The University of Melbourne, Melbourne, VIC, Australia
| | - Michael R. Ibbotson
- Department of Biomedical Engineering, Faculty of Engineering & Information Technology, The University of Melbourne, Melbourne, VIC, Australia
| | - Tatiana Kameneva
- School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Hawthorn, VIC, Australia
| |
Collapse
|
6
|
Suiwal S, Wartenberg P, Boehm U, Schmitz F, Schwarz K. A Novel Cre Recombinase Mouse Strain for Cell-Specific Deletion of Floxed Genes in Ribbon Synapse-Forming Retinal Neurons. Int J Mol Sci 2024; 25:1916. [PMID: 38339191 PMCID: PMC10856425 DOI: 10.3390/ijms25031916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/28/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024] Open
Abstract
We generated a novel Cre mouse strain for cell-specific deletion of floxed genes in ribbon synapse-forming retinal neurons. Previous studies have shown that the RIBEYE promotor targets the expression of recombinant proteins such as fluorescently tagged RIBEYE to photoreceptors and retinal bipolar cells and generates fluorescent synaptic ribbons in situ in these neurons. Here, we used the same promotor to generate a novel transgenic mouse strain in which the RIBEYE promotor controls the expression of a Cre-ER(T2) recombinase (RIBEYE-Cre). To visualize Cre expression, the RIBEYE-Cre animals were crossed with ROSA26 tau-GFP (R26-τGFP) reporter mice. In the resulting RIBEYE-Cre/R26 τGFP animals, Cre-mediated removal of a transcriptional STOP cassette results in the expression of green fluorescent tau protein (tau-GFP) that binds to cellular microtubules. We detected robust tau-GFP expression in retinal bipolar cells. Surprisingly, we did not find fluorescent tau-GFP expression in mouse photoreceptors. The lack of tau-GFP reporter protein in these cells could be based on the previously reported absence of tau protein in mouse photoreceptors which could lead to the degradation of the recombinant tau protein. Consistent with this, we detected Cre and tau-GFP mRNA in mouse photoreceptor slices by RT-PCR. The transgenic RIBEYE-Cre mouse strain provides a new tool to study the deletion of floxed genes in ribbon synapse-forming neurons of the retina and will also allow for analyzing gene deletions that are lethal if globally deleted in neurons.
Collapse
Affiliation(s)
- Shweta Suiwal
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, 66421 Homburg, Germany;
| | - Philipp Wartenberg
- Institute of Clinical and Experimental Pharmacology, Center for Molecular Signaling (PZMS) and Center for Gender-Specific Biology and Medicine (CGBM), Medical School, Saarland University, 66421 Homburg, Germany; (P.W.); (U.B.)
| | - Ulrich Boehm
- Institute of Clinical and Experimental Pharmacology, Center for Molecular Signaling (PZMS) and Center for Gender-Specific Biology and Medicine (CGBM), Medical School, Saarland University, 66421 Homburg, Germany; (P.W.); (U.B.)
| | - Frank Schmitz
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, 66421 Homburg, Germany;
| | - Karin Schwarz
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, 66421 Homburg, Germany;
| |
Collapse
|
7
|
Liu Y, Mahony BW, Wang X, Daye PM, Wang W, Cavanagh P, Pouget P, Andolina IM. Assessing perceptual chromatic equiluminance using a reflexive pupillary response. Sci Rep 2024; 14:2420. [PMID: 38286801 PMCID: PMC10825167 DOI: 10.1038/s41598-024-51982-z] [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: 10/17/2023] [Accepted: 01/11/2024] [Indexed: 01/31/2024] Open
Abstract
Equiluminant stimuli help assess the integrity of colour perception and the relationship of colour to other visual features. As a result of individual variation, it is necessary to calibrate experimental visual stimuli to suit each individual's unique equiluminant ratio. Most traditional methods rely on training observers to report their subjective equiluminance point. Such paradigms cannot easily be implemented on pre-verbal or non-verbal observers. Here, we present a novel Pupil Frequency-Tagging Method (PFTM) for detecting a participant's unique equiluminance point without verbal instruction and with minimal training. PFTM analyses reflexive pupil oscillations induced by slow (< 2 Hz) temporal alternations between coloured stimuli. Two equiluminant stimuli will induce a similar pupil dilation response regardless of colour; therefore, an observer's equiluminant point can be identified as the luminance ratio between two colours for which the oscillatory amplitude of the pupil at the tagged frequency is minimal. We compared pupillometry-based equiluminance ratios to those obtained with two established techniques in humans: minimum flicker and minimum motion. In addition, we estimated the equiluminance point in non-human primates, demonstrating that this new technique can be successfully employed in non-verbal subjects.
Collapse
Affiliation(s)
- Ye Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | | | - Xiaochun Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Pierre M Daye
- Sorbonne Université, Inserm, CNRS, ICM, Paris, France
| | - Wei Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Patrick Cavanagh
- Glendon College and Centre for Vision Research, York University, Toronto, Canada
| | - Pierre Pouget
- Sorbonne Université, Inserm, CNRS, ICM, Paris, France.
| | - Ian Max Andolina
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
8
|
Kramer AC, Carthage J, Berry Y, Gurdziel K, Cook TA, Thummel R. A comparative analysis of gene and protein expression in chronic and acute models of photoreceptor degeneration in adult zebrafish. Front Cell Dev Biol 2023; 11:1233269. [PMID: 37745292 PMCID: PMC10512720 DOI: 10.3389/fcell.2023.1233269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Background: Adult zebrafish are capable of photoreceptor (PR) regeneration following acute phototoxic lesion (AL). We developed a chronic low light (CLL) exposure model that more accurately reflects chronic PR degeneration observed in many human retinal diseases. Methods: Here, we characterize the morphological and transcriptomic changes associated with acute and chronic models of PR degeneration at 8 time-points over a 28-day window using immunohistochemistry and 3'mRNA-seq. Results: We first observed a differential sensitivity of rod and cone PRs to CLL. Next, we found no evidence for Müller glia (MG) gliosis or regenerative cell-cycle re-entry in the CLL model, which is in contrast to the robust gliosis and proliferative response from resident MG in the AL model. Differential responses of microglia between the models was also observed. Transcriptomic comparisons between the models revealed gene-specific networks of PR regeneration and degeneration, including genes that are activated under conditions of chronic PR stress. Finally, we showed that CLL is at least partially reversible, allowing for rod and cone outer segment outgrowth and replacement of rod cell nuclei via an apparent upregulation of the existing rod neurogenesis mechanism. Discussion: Collectively, these data provide a direct comparison of the morphological and transcriptomic PR degeneration and regeneration models in zebrafish.
Collapse
Affiliation(s)
- Ashley C. Kramer
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Justin Carthage
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Yasmeen Berry
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Katherine Gurdziel
- Genomic Sciences Core, Wayne State University, Detroit, MI, United States
| | - Tiffany A. Cook
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Ryan Thummel
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
9
|
Haq W, Zrenner E, Ueffing M, Paquet-Durand F. Using Micro-Electrode-Array Recordings and Retinal Disease Models to Elucidate Visual Functions: Simultaneous Recording of Local Electroretinograms and Ganglion Cell Action Potentials Reveals the Origin of Retinal Oscillatory Potentials. Bioengineering (Basel) 2023; 10:725. [PMID: 37370656 DOI: 10.3390/bioengineering10060725] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The electroretinogram (ERG) is an essential diagnostic tool for visual function, both in clinical and research settings. Here, we establish an advanced in vitro approach to assess cell-type-specific ERG signal components. METHODS Retinal explant cultures, maintained under entirely controlled conditions, were derived from wild-type mice and rd10 rod- and cpfl1 cone-degeneration mouse models. Local micro-ERG (µERG) and simultaneous ganglion cell (GC) recordings were obtained from the retinal explants using multi-electrode arrays. Band-pass filtering was employed to distinguish photoreceptor, bipolar cell, amacrine cell (AC), and GC responses. RESULTS Scotopic and photopic stimulation discriminated between rod and cone responses in wild-type and mutant retina. The 25 kHz sampling rate allowed the visualization of oscillatory potentials (OPs) in extraordinary detail, revealing temporal correlations between OPs and GC responses. Pharmacological isolation of different retinal circuits found that OPs are generated by inner retinal AC electrical synapses. Importantly, this AC activity helped synchronise GC activity. CONCLUSION Our µERG protocol simultaneously records the light-dependent activities of the first-, second-, and third-order neurons within the native neuronal circuitry, providing unprecedented insights into retinal physiology and pathophysiology. This method now also enables complete in vitro retinal function testing of therapeutic interventions, providing critical guidance for later in vivo investigations.
Collapse
Affiliation(s)
- Wadood Haq
- Centre for Ophthalmology, Institute for Ophthalmic Research, Elfriede-Aulhorn-Straße 7, 72076 Tuebingen, Germany
| | - Eberhart Zrenner
- Centre for Ophthalmology, Institute for Ophthalmic Research, Elfriede-Aulhorn-Straße 7, 72076 Tuebingen, Germany
| | - Marius Ueffing
- Centre for Ophthalmology, Institute for Ophthalmic Research, Elfriede-Aulhorn-Straße 7, 72076 Tuebingen, Germany
| | - François Paquet-Durand
- Centre for Ophthalmology, Institute for Ophthalmic Research, Elfriede-Aulhorn-Straße 7, 72076 Tuebingen, Germany
| |
Collapse
|
10
|
Griffis KG, Fehlhaber KE, Rieke F, Sampath AP. Light Adaptation of Retinal Rod Bipolar Cells. J Neurosci 2023; 43:4379-4389. [PMID: 37208176 PMCID: PMC10278674 DOI: 10.1523/jneurosci.0444-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/07/2023] [Accepted: 05/02/2023] [Indexed: 05/21/2023] Open
Abstract
The sensitivity of retinal cells is altered in background light to optimize the detection of contrast. For scotopic (rod) vision, substantial adaptation occurs in the first two cells, the rods and rod bipolar cells (RBCs), through sensitivity adjustments in rods and postsynaptic modulation of the transduction cascade in RBCs. To study the mechanisms mediating these components of adaptation, we made whole-cell, voltage-clamp recordings from retinal slices of mice from both sexes. Adaptation was assessed by fitting the Hill equation to response-intensity relationships with the parameters of half-maximal response (I1/2 ), Hill coefficient (n), and maximum response amplitude (Rmax ). We show that rod sensitivity decreases in backgrounds according to the Weber-Fechner relation with an I1/2 of ∼50 R* s-1 The sensitivity of RBCs follows a near-identical function, indicating that changes in RBC sensitivity in backgrounds bright enough to adapt the rods are mostly derived from the rods themselves. Backgrounds too dim to adapt the rods can however alter n, relieving a synaptic nonlinearity likely through entry of Ca2+ into the RBCs. There is also a surprising decrease of Rmax , indicating that a step in RBC synaptic transduction is desensitized or that the transduction channels became reluctant to open. This effect is greatly reduced after dialysis of BAPTA at a membrane potential of +50 mV to impede Ca2+ entry. Thus the effects of background illumination in RBCs are in part the result of processes intrinsic to the photoreceptors and in part derive from additional Ca2+-dependent processes at the first synapse of vision.SIGNIFICANCE STATEMENT Light adaptation adjusts the sensitivity of vision as ambient illumination changes. Adaptation for scotopic (rod) vision is known to occur partly in the rods and partly in the rest of the retina from presynaptic and postsynaptic mechanisms. We recorded light responses of rods and rod bipolar cells to identify different components of adaptation and study their mechanisms. We show that bipolar-cell sensitivity largely follows adaptation of the rods but that light too dim to adapt the rods produces a linearization of the bipolar-cell response and a surprising decrease in maximum response amplitude, both mediated by a change in intracellular Ca2+ These findings provide a new understanding of how the retina responds to changing illumination.
Collapse
Affiliation(s)
- Khris G Griffis
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095
| | - Katherine E Fehlhaber
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington 98195
| | - Alapakkam P Sampath
- Department of Ophthalmology and Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| |
Collapse
|
11
|
Sladek AL, Thoreson WB. Using optogenetics to dissect rod inputs to OFF ganglion cells in the mouse retina. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1146785. [PMID: 37426783 PMCID: PMC10327572 DOI: 10.3389/fopht.2023.1146785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Introduction Light responses of rod photoreceptor cells traverse the retina through three pathways. The primary pathway involves synapses from rods to ON-type rod bipolar cells with OFF signals reaching retinal ganglion cells (RGCs) via sign-inverting glycinergic synapses. Secondly, rod signals can enter cones through gap junctions. Finally, rods can synapse directly onto cone OFF bipolar cells. Methods To analyze these pathways, we obtained whole cell recordings from OFF-type α RGCs in mouse retinas while expressing channelrhodopsin-2 in rods and/or cones. Results Optogenetic stimulation of rods or cones evoked large fast currents in OFF RGCs. Blocking the primary rod pathway with L-AP4 and/or strychnine reduced rod-driven optogenetic currents in OFF RGCs by ~1/3. Blocking kainate receptors of OFF cone bipolar cells suppressed both rod- and cone-driven optogenetic currents in OFF RGCs. Inhibiting gap junctions between rods and cones with mecloflenamic acid or quinpirole reduced rod-driven responses in OFF RGCs. Eliminating the exocytotic Ca2+ sensor, synaptotagmin 1 (Syt1), from cones abolished cone-driven optogenetic responses in RGCs. Rod-driven currents were not significantly reduced after isolating the secondary pathway by eliminating Syt1 and synaptotagmin 7 (Syt7) to block synaptic release from rods. Eliminating Syt1 from both rods and cones abolished responses to optogenetic stimulation. In Cx36 KO retinas lacking rod-cone gap junctions, optogenetic activation of rods evoked small and slow responses in most OFF RGCs suggesting rod signals reached them through an indirect pathway. Two OFF cells showed faster responses consistent with more direct input from cone OFF bipolar cells. Discussion These data show that the secondary rod pathway supports robust inputs into OFF α RGCs and suggests the tertiary pathway recruits both direct and indirect inputs.
Collapse
Affiliation(s)
- Asia L. Sladek
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Wallace B. Thoreson
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, United States
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
12
|
Wilmet B, Callebert J, Duvoisin R, Goulet R, Tourain C, Michiels C, Frederiksen H, Schaeffel F, Marre O, Sahel JA, Audo I, Picaud S, Zeitz C. Mice Lacking Gpr179 with Complete Congenital Stationary Night Blindness Are a Good Model for Myopia. Int J Mol Sci 2022; 24:ijms24010219. [PMID: 36613663 PMCID: PMC9820543 DOI: 10.3390/ijms24010219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
Mutations in GPR179 are one of the most common causes of autosomal recessive complete congenital stationary night blindness (cCSNB). This retinal disease is characterized in patients by impaired dim and night vision, associated with other ocular symptoms, including high myopia. cCSNB is caused by a complete loss of signal transmission from photoreceptors to ON-bipolar cells. In this study, we hypothesized that the lack of Gpr179 and the subsequent impaired ON-pathway could lead to myopic features in a mouse model of cCSNB. Using ultra performance liquid chromatography, we show that adult Gpr179-/- mice have a significant decrease in both retinal dopamine and 3,4-dihydroxyphenylacetic acid, compared to Gpr179+/+ mice. This alteration of the dopaminergic system is thought to be correlated with an increased susceptibility to lens-induced myopia but does not affect the natural refractive development. Altogether, our data added a novel myopia model, which could be used to identify therapeutic interventions.
Collapse
Affiliation(s)
- Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Correspondence: (B.W.); (C.Z.); Tel.: +33-1-53-46-25-26 (B.W.); +33-1-53-46-25-40 (C.Z.)
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, AP-HP, 75010 Paris, France
| | - Robert Duvoisin
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ruben Goulet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Christophe Tourain
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Wavefront-Engineering Microscopy Group, Neurophotonics Laboratory, CNRS UMR8250, Paris Descartes University, 75270 Paris, France
| | - Christelle Michiels
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Helen Frederiksen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Frank Schaeffel
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4056 Basel, Switzerland
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
- Zeiss Vision Lab, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
| | - Olivier Marre
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - José Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
- Fondation Ophtalmologique Adolphe de Rothschild, 75019 Paris, France
- Académie des Sciences, Institut de France, 75006 Paris, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Correspondence: (B.W.); (C.Z.); Tel.: +33-1-53-46-25-26 (B.W.); +33-1-53-46-25-40 (C.Z.)
| |
Collapse
|
13
|
Gupta SK, Chakraborty R, Verkicharla PK. Electroretinogram responses in myopia: a review. Doc Ophthalmol 2022; 145:77-95. [PMID: 34787722 PMCID: PMC9470726 DOI: 10.1007/s10633-021-09857-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/11/2021] [Indexed: 11/02/2022]
Abstract
The stretching of a myopic eye is associated with several structural and functional changes in the retina and posterior segment of the eye. Recent research highlights the role of retinal signaling in ocular growth. Evidence from studies conducted on animal models and humans suggests that visual mechanisms regulating refractive development are primarily localized at the retina and that the visual signals from the retinal periphery are also critical for visually guided eye growth. Therefore, it is important to study the structural and functional changes in the retina in relation to refractive errors. This review will specifically focus on electroretinogram (ERG) changes in myopia and their implications in understanding the nature of retinal functioning in myopic eyes. Based on the available literature, we will discuss the fundamentals of retinal neurophysiology in the regulation of vision-dependent ocular growth, findings from various studies that investigated global and localized retinal functions in myopia using various types of ERGs.
Collapse
Affiliation(s)
- Satish Kumar Gupta
- Myopia Research Lab, Prof. Brien Holden Eye Research Centre, Brien Holden Institute of Optometry and Vision Sciences, Kallam Anji Reddy Campus, L V Prasad Eye Institute, Hyderabad, India
| | - Ranjay Chakraborty
- Caring Futures Institute, College of Nursing and Health Sciences, Optometry and Vision Science, Flinders University, Adelaide, South Australia, Australia
| | - Pavan Kumar Verkicharla
- Myopia Research Lab, Prof. Brien Holden Eye Research Centre, Brien Holden Institute of Optometry and Vision Sciences, Kallam Anji Reddy Campus, L V Prasad Eye Institute, Hyderabad, India.
| |
Collapse
|
14
|
Retinal Toxicity Induced by Chemical Agents. Int J Mol Sci 2022; 23:ijms23158182. [PMID: 35897758 PMCID: PMC9331776 DOI: 10.3390/ijms23158182] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Vision is an important sense for humans, and visual impairment/blindness has a huge impact in daily life. The retina is a nervous tissue that is essential for visual processing since it possesses light sensors (photoreceptors) and performs a pre-processing of visual information. Thus, retinal cell dysfunction or degeneration affects visual ability and several general aspects of the day-to-day of a person's lives. The retina has a blood-retinal barrier, which protects the tissue from a wide range of molecules or microorganisms. However, several agents, coming from systemic pathways, reach the retina and influence its function and survival. Pesticides are still used worldwide for agriculture, contaminating food with substances that could reach the retina. Natural products have also been used for therapeutic purposes and are another group of substances that can get to the retina. Finally, a wide number of medicines administered for different diseases can also affect the retina. The present review aimed to gather recent information about the hazard of these products to the retina, which could be used to encourage the search for more healthy, suitable, or less risky agents.
Collapse
|
15
|
Cha S, Ahn J, Jeong Y, Lee YH, Kim HK, Lee D, Yoo Y, Goo YS. Stage-Dependent Changes of Visual Function and Electrical Response of the Retina in the rd10 Mouse Model. Front Cell Neurosci 2022; 16:926096. [PMID: 35936494 PMCID: PMC9345760 DOI: 10.3389/fncel.2022.926096] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/23/2022] [Indexed: 12/28/2022] Open
Abstract
One of the critical prerequisites for the successful development of retinal prostheses is understanding the physiological features of retinal ganglion cells (RGCs) in the different stages of retinal degeneration (RD). This study used our custom-made rd10 mice, C57BL/6-Pde6bem1(R560C)Dkl/Korl mutated on the Pde6b gene in C57BL/6J mouse with the CRISPR/Cas9-based gene-editing method. We selected the postnatal day (P) 45, P70, P140, and P238 as representative ages for RD stages. The optomotor response measured the visual acuity across degeneration stages. At P45, the rd10 mice exhibited lower visual acuity than wild-type (WT) mice. At P140 and older, no optomotor response was observed. We classified RGC responses to the flashed light into ON, OFF, and ON/OFF RGCs via in vitro multichannel recording. With degeneration, the number of RGCs responding to the light stimulation decreased in all three types of RGCs. The OFF response disappeared faster than the ON response with older postnatal ages. We elicited RGC spikes with electrical stimulation and analyzed the network-mediated RGC response in the rd10 mice. Across all postnatal ages, the spikes of rd10 RGCs were less elicited by pulse amplitude modulation than in WT RGCs. The ratio of RGCs showing multiple peaks of spike burst increased in older ages. The electrically evoked RGC spikes by the pulse amplitude modulation differ across postnatal ages. Therefore, degeneration stage-dependent stimulation strategies should be considered for developing retinal prosthesis and successful vision restoration.
Collapse
Affiliation(s)
- Seongkwang Cha
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Jungryul Ahn
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Yurim Jeong
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Yong Hee Lee
- Department of Biochemistry, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Hyong Kyu Kim
- Department of Microbiology, Chungbuk National University School of Medicine, Cheongju, South Korea
| | - Daekee Lee
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Yongseok Yoo
- Department of Electronics Engineering, Incheon National University, Incheon, South Korea
- *Correspondence: Yongseok Yoo,
| | - Yong Sook Goo
- Department of Physiology, Chungbuk National University School of Medicine, Cheongju, South Korea
- Yong Sook Goo,
| |
Collapse
|
16
|
Jung R, Kelbsch C, Wilhelm H, Wilhelm B, Strasser T, Peters T, Kempf M, Kortüm F, Pohl L, Stingl K, Stingl K. Cell-specific electrical stimulation of human retinal neurons assessed by pupillary response dynamics in vivo. Exp Eye Res 2022; 222:109185. [PMID: 35850172 DOI: 10.1016/j.exer.2022.109185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 06/12/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022]
Abstract
Studies on the electrical excitability of retinal neurons show that photoreceptors and other cell types can be selectively activated by distinct stimulation frequencies in vitro. Yet, this principle still needs to be validated in humans in vivo. As a first step, this study explored the frequency preferences of human rods by means of transcorneal electrostimulation (TES), using the electrically-elicited pupillary responses (EEPRs) as an objective readout. The stimulation paradigm contained a 1.2 Hz sinusoidal envelope, which was superimposed on variable carrier frequencies (4-30 Hz). These currents were delivered to one of the participant's eyes via a corneal electrode and consensual pupillary reactions were recorded from the contralateral eye. The responsiveness of the retina at each frequency was assessed based on the EEPR dynamics. Differences between healthy participants and patients with retinitis pigmentosa were evaluated to identify the preferred frequency range of rods. The responsiveness of healthy individuals revealed a clear peak around 6-8 Hz. In contrast, the pupillary responses of patients were significantly reduced in the lower frequency range. These findings suggest that the responses in this frequency bin were selectively mediated by rods. This work provides evidence that different retinal cell types can be selectively activated via TES in vivo, and that this effect can be captured noninvasively using EEPRs. This knowledge may be exploited for the diagnostics and therapy of retinal diseases, e.g., to design cell-specific functional tests for the degenerating retina, or to optimize stimulation paradigms which are currently used by retinal prostheses.
Collapse
Affiliation(s)
- Ronja Jung
- University Eye Hospital, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany.
| | - Carina Kelbsch
- University Eye Hospital, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany; Pupil Research Group, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany
| | - Helmut Wilhelm
- Pupil Research Group, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany
| | - Barbara Wilhelm
- Pupil Research Group, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany
| | - Torsten Strasser
- University Eye Hospital, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany; Pupil Research Group, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany; Institute for Ophthalmic Research, University of Tübingen, 72076, Tübingen, Germany
| | - Tobias Peters
- Pupil Research Group, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany
| | - Melanie Kempf
- University Eye Hospital, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany; Center for Rare Diseases, University of Tübingen, 72076, Tübingen, Germany
| | - Friederike Kortüm
- University Eye Hospital, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany
| | - Lisa Pohl
- University Eye Hospital, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany
| | - Krunoslav Stingl
- University Eye Hospital, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany; Pupil Research Group, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany; Center for Rare Diseases, University of Tübingen, 72076, Tübingen, Germany
| | - Katarina Stingl
- University Eye Hospital, Center for Ophthalmology, University of Tuebingen, 72076, Tuebingen, Germany; Center for Rare Diseases, University of Tübingen, 72076, Tübingen, Germany
| |
Collapse
|
17
|
Fain GL. Vision: Life on the dark side. Curr Biol 2022; 32:R741-R743. [PMID: 35820384 DOI: 10.1016/j.cub.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Mice detect decreases in illumination in dim light near the visual threshold with OFF retinal ganglion cells.
Collapse
Affiliation(s)
- Gordon L Fain
- Departments of Integrative Biology/Physiology and Ophthalmology, Stein Eye Institute, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
18
|
Lankford CK, Umino Y, Poria D, Kefalov V, Solessio E, Baker SA. Cone-Driven Retinal Responses Are Shaped by Rod But Not Cone HCN1. J Neurosci 2022; 42:4231-4249. [PMID: 35437278 PMCID: PMC9145265 DOI: 10.1523/jneurosci.2271-21.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 11/21/2022] Open
Abstract
Signal integration of converging neural circuits is poorly understood. One example is in the retina where the integration of rod and cone signaling is responsible for the large dynamic range of vision. The relative contribution of rods versus cones is dictated by a complex function involving background light intensity and stimulus temporal frequency. One understudied mechanism involved in coordinating rod and cone signaling onto the shared retinal circuit is the hyperpolarization activated current (Ih) mediated by hyperpolarization-activated cyclic nucleotide-gated 1 (HCN1) channels expressed in rods and cones. Ih opposes membrane hyperpolarization driven by activation of the phototransduction cascade and modulates the strength and kinetics of the photoreceptor voltage response. We examined conditional knock-out (KO) of HCN1 from mouse rods using electroretinography (ERG). In the absence of HCN1, rod responses are prolonged in dim light which altered the response to slow modulation of light intensity both at the level of retinal signaling and behavior. Under brighter intensities, cone-driven signaling was suppressed. To our surprise, conditional KO of HCN1 from mouse cones had no effect on cone-mediated signaling. We propose that Ih is dispensable in cones because of the high level of temporal control of cone phototransduction. Thus, HCN1 is required for cone-driven retinal signaling only indirectly by modulating the voltage response of rods to limit their output.SIGNIFICANCE STATEMENT Hyperpolarization gated hyperpolarization-activated cyclic nucleotide-gated 1 (HCN1) channels carry a feedback current that helps to reset light-activated photoreceptors. Using conditional HCN1 knock-out (KO) mice we show that ablating HCN1 from rods allows rods to signal in bright light when they are normally shut down. Instead of enhancing vision this results in suppressing cone signaling. Conversely, ablating HCN1 from cones was of no consequence. This work provides novel insights into the integration of rod and cone signaling in the retina and challenges our assumptions about the role of HCN1 in cones.
Collapse
Affiliation(s)
- Colten K Lankford
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, Iowa 52242
| | - Yumiko Umino
- Center for Vision Research, Department of Ophthalmology and Visual Sciences, State University of New York Upstate Medical University, Syracuse, New York 13210
| | - Deepak Poria
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, California 92697
| | - Vladimir Kefalov
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, California 92697
- Department of Physiology and Biophysics, University of California, Irvine, California 92697
| | - Eduardo Solessio
- Center for Vision Research, Department of Ophthalmology and Visual Sciences, State University of New York Upstate Medical University, Syracuse, New York 13210
| | - Sheila A Baker
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, Iowa 52242
- Department of Ophthalmology and Visual Sciences and Institute for Vision Research, University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
19
|
Frederiksen R, Fain GL, Sampath AP. A hyperpolarizing rod bipolar cell in the sea lamprey, Petromyzon marinus. J Exp Biol 2022; 225:jeb243949. [PMID: 35319772 PMCID: PMC10658897 DOI: 10.1242/jeb.243949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/15/2022] [Indexed: 11/20/2022]
Abstract
Retinal bipolar cells receive direct input from rod and cone photoreceptors and send axons into the inner retina, synapsing onto amacrine and ganglion cells. Bipolar cell responses can be either depolarizing (ON) or hyperpolarizing (OFF); in lower vertebrates, bipolar cells receive mixed rod and cone input, whereas in mammals, input is mostly segregated into 14 classes of cone ON and OFF cells and a single rod ON bipolar cell. We show that lamprey, like mammals, have rod bipolar cells with little or no cone input, but these cells are OFF rather than ON. They have a characteristic morphology and a spectral sensitivity nearly indistinguishable from that of rod photoreceptors. In background light known to saturate rods, rod bipolar cells are also saturated and cannot respond to increment flashes. Our results suggest that early vertebrate progenitors of both agnathans and gnathostomes may have had a more fluid retinal organization than previously thought.
Collapse
Affiliation(s)
- Rikard Frederiksen
- Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095-7000, USA
| | - Gordon L. Fain
- Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095-7000, USA
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA 90095-7239, USA
| | - Alapakkam P. Sampath
- Stein Eye Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095-7000, USA
| |
Collapse
|
20
|
Wagner N, Safaei A, Vogt PA, Gammel MR, Dick HB, Schnichels S, Joachim SC. Coculture of ARPE-19 Cells and Porcine Neural Retina as an Ex Vivo Retinal Model. Altern Lab Anim 2022; 50:27-44. [PMID: 35302924 DOI: 10.1177/02611929221082662] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Neural retinal organ cultures are used to investigate ocular pathomechanisms. However, these cultures lack the essential retinal pigment epithelium (RPE) cells, which are part of the actual in vivo retina. To simulate a more realistic ex vivo model, porcine neural retina explants were cocultured with ARPE-19 cells (ARPE-19 group), which are derived from human RPE. To identify whether the entire cells or just the cell factors are necessary, in a second experimental group, porcine neural retina explants were cultured with medium derived from ARPE-19 cells (medium group). Individually cultured neural retina explants served as controls (control group). After 8 days, all neural retinas were analysed to evaluate retinal thickness, photoreceptors, microglia, complement factors and synapses (n = 6-8 per group). The neural retina thickness in the ARPE-19 group was significantly better preserved than in the control group (p = 0.031). Also, the number of L-cones was higher in the ARPE-19 group, as compared to the control group (p < 0.001). Furthermore, the ARPE-19 group displayed an increased presynaptic glutamate uptake (determined via vGluT1 labelling) and enhanced post-synaptic density (determined via PSD-95 labelling). Combined Iba1 and iNOS detection revealed only minor effects of ARPE-19 cells on microglial activity, with a slight downregulation of total microglia activity apparent in the medium group. Likewise, only minor beneficial effects on photoreceptors and synaptic structure were found in the medium group. This novel system offers the opportunity to investigate interactions between the neural retina and RPE cells, and suggests that the inclusion of a RPE feeder layer has beneficial effects on the ex vivo maintenance of neural retina. By modifying the culture conditions, this coculture model allows a better understanding of photoreceptor death and photoreceptor-RPE cell interactions in retinal diseases.
Collapse
Affiliation(s)
- Natalie Wagner
- Experimental Eye Research Institute, University Eye Hospital, 9142Ruhr-University Bochum, Germany
| | - Armin Safaei
- Experimental Eye Research Institute, University Eye Hospital, 9142Ruhr-University Bochum, Germany
| | - Pia A Vogt
- Experimental Eye Research Institute, University Eye Hospital, 9142Ruhr-University Bochum, Germany
| | - Maurice R Gammel
- Experimental Eye Research Institute, University Eye Hospital, 9142Ruhr-University Bochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, 9142Ruhr-University Bochum, Germany
| | - Sven Schnichels
- Centre for Ophthalmology Tübingen, University Eye Hospital Tübingen, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, 9142Ruhr-University Bochum, Germany
| |
Collapse
|
21
|
Heisterkamp P, Borsch O, Lezama ND, Gasparini S, Fathima A, Carvalho LS, Wagner F, Karl MO, Schlierf M, Ader M. Evidence for endogenous exchange of cytoplasmic material between a subset of cone and rod photoreceptors within the adult mammalian retina via direct cell-cell connections. Exp Eye Res 2022; 219:109033. [DOI: 10.1016/j.exer.2022.109033] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022]
|
22
|
Goodenow D, Greer AJ, Cone SJ, Gaddameedhi S. Circadian effects on UV-induced damage and mutations. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2022; 789:108413. [PMID: 35690416 PMCID: PMC9188652 DOI: 10.1016/j.mrrev.2022.108413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/09/2022] [Accepted: 02/14/2022] [Indexed: 10/19/2022]
Abstract
Skin cancer is the most diagnosed type of cancer in the United States, and while most of these malignancies are highly treatable, treatment costs still exceed $8 billion annually. Over the last 50 years, the annual incidence of skin cancer has steadily grown; therefore, understanding the environmental factors driving these types of cancer is a prominent research-focus. A causality between ultraviolet radiation (UVR) exposure and skin cancer is well-established, but exposure to UVR alone is not necessarily sufficient to induce carcinogenesis. The emerging field of circadian biology intersects strongly with the physiological systems of the mammalian body and introduces a unique opportunity for analyzing mechanisms of homeostatic disruption. The circadian clock refers to the approximate 24-hour cycle, in which protein levels of specific clock-controlled genes (CCGs) fluctuate based on the time of day. Though these CCGs are tissue specific, the skin has been observed to have a robust circadian clock that plays a role in its response to UVR exposure. This in-depth review will detail the mechanisms of the circadian clock and its role in cellular homeostasis. Next, the skin's response to UVR exposure and its induction of DNA damage and mutations will be covered - with an additional focus placed on how the circadian clock influences this response through nucleotide excision repair. Lastly, this review will discuss current models for studying UVR-induced skin lesions and perturbations of the circadian clock, as well as the impact of these factors on human health.
Collapse
Affiliation(s)
- Donna Goodenow
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27606, USA
| | - Adam J Greer
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27606, USA
| | - Sean J Cone
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27606, USA
| | - Shobhan Gaddameedhi
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27606, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27606, USA.
| |
Collapse
|
23
|
Cortical Visual Mapping following Ocular Gene Augmentation Therapy for Achromatopsia. J Neurosci 2021; 41:7363-7371. [PMID: 34349002 DOI: 10.1523/jneurosci.3222-20.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 07/07/2021] [Accepted: 07/15/2021] [Indexed: 11/21/2022] Open
Abstract
The ability of the adult human brain to develop function following correction of congenital deafferentation is controversial. Specifically, cases of recovery from congenital visual deficits are rare. CNGA3-achromatopsia is a congenital hereditary disease caused by cone-photoreceptor dysfunction, leading to impaired acuity, photoaversion, and complete color blindness. Essentially, these patients have rod-driven vision only, seeing the world in blurry shades of gray. We use the uniqueness of this rare disease, in which the cone-photoreceptors and afferent fibers are preserved but do not function, as a model to study cortical visual plasticity. We had the opportunity to study two CNGA3-achromatopsia adults (one female) before and after ocular gene augmentation therapy. Alongside behavioral visual tests, we used novel fMRI-based measurements to assess participants' early visual population receptive-field sizes and color regions. Behaviorally, minor improvements were observed, including reduction in photoaversion, marginal improvement in acuity, and a new ability to detect red color. No improvement was observed in color arrangement tests. Cortically, pretreatment, patients' population-receptive field sizes of early visual areas were untypically large, but were decreased following treatment specifically in the treated eye. We suggest that this demonstrates cortical ability to encode new input, even at adulthood. On the other hand, no activation of color-specific cortical regions was demonstrated in these patients either before or up to 1 year post-treatment. The source of this deficiency might be attributed either to insufficient recovery of cone function at the retinal level or to challenges that the adult cortex faces when computing new cone-derived input to achieve color perception.SIGNIFICANCE STATEMENT The possibility that the adult human brain may regain or develop function following correction of congenital deafferentation has fired the imagination of scientists over the years. In the visual domain, cases of recovery from congenital deficits are rare. Gene therapy visual restoration for congenital CNGA3-achromatopsia, a disease caused by cone photoreceptor dysfunction, gave us the opportunity to examine cortical function, to the best of our knowledge for the first time, both before and after restorative treatment. While behaviorally only minor improvements were observed post-treatment, fMRI analysis, including size algorithms of population-receptive fields, revealed cortical changes, specifically receptive field size decrease in the treated eyes. This suggests that, at least to some degree, the adult cortex is able to encode new input.
Collapse
|
24
|
Cangiano L, Asteriti S. Interphotoreceptor coupling: an evolutionary perspective. Pflugers Arch 2021; 473:1539-1554. [PMID: 33988778 PMCID: PMC8370920 DOI: 10.1007/s00424-021-02572-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/13/2021] [Accepted: 04/23/2021] [Indexed: 12/16/2022]
Abstract
In the vertebrate retina, signals generated by cones of different spectral preference and by highly sensitive rod photoreceptors interact at various levels to extract salient visual information. The first opportunity for such interaction is offered by electrical coupling of the photoreceptors themselves, which is mediated by gap junctions located at the contact points of specialised cellular processes: synaptic terminals, telodendria and radial fins. Here, we examine the evolutionary pressures for and against interphotoreceptor coupling, which are likely to have shaped how coupling is deployed in different species. The impact of coupling on signal to noise ratio, spatial acuity, contrast sensitivity, absolute and increment threshold, retinal signal flow and colour discrimination is discussed while emphasising available data from a variety of vertebrate models spanning from lampreys to primates. We highlight the many gaps in our knowledge, persisting discrepancies in the literature, as well as some major unanswered questions on the actual extent and physiological role of cone-cone, rod-cone and rod-rod communication. Lastly, we point toward limited but intriguing evidence suggestive of the ancestral form of coupling among ciliary photoreceptors.
Collapse
Affiliation(s)
- Lorenzo Cangiano
- Dept. of Translational Research, University of Pisa, Via San Zeno 31, 56123, Pisa, Italy.
| | - Sabrina Asteriti
- Dept. of Translational Research, University of Pisa, Via San Zeno 31, 56123, Pisa, Italy
| |
Collapse
|
25
|
Khani MH, Gollisch T. Linear and nonlinear chromatic integration in the mouse retina. Nat Commun 2021; 12:1900. [PMID: 33772000 PMCID: PMC7997992 DOI: 10.1038/s41467-021-22042-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 02/23/2021] [Indexed: 11/09/2022] Open
Abstract
The computations performed by a neural circuit depend on how it integrates its input signals into an output of its own. In the retina, ganglion cells integrate visual information over time, space, and chromatic channels. Unlike the former two, chromatic integration is largely unexplored. Analogous to classical studies of spatial integration, we here study chromatic integration in mouse retina by identifying chromatic stimuli for which activation from the green or UV color channel is maximally balanced by deactivation through the other color channel. This reveals nonlinear chromatic integration in subsets of On, Off, and On-Off ganglion cells. Unlike the latter two, nonlinear On cells display response suppression rather than activation under balanced chromatic stimulation. Furthermore, nonlinear chromatic integration occurs independently of nonlinear spatial integration, depends on contributions from the rod pathway and on surround inhibition, and may provide information about chromatic boundaries, such as the skyline in natural scenes.
Collapse
Affiliation(s)
- Mohammad Hossein Khani
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany.
- Bernstein Center for Computational Neuroscience, Göttingen, Germany.
- International Max Planck Research School for Neuroscience, Göttingen, Germany.
| | - Tim Gollisch
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany.
- Bernstein Center for Computational Neuroscience, Göttingen, Germany.
| |
Collapse
|
26
|
Light responses of mammalian cones. Pflugers Arch 2021; 473:1555-1568. [PMID: 33742309 DOI: 10.1007/s00424-021-02551-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 12/24/2022]
Abstract
Cone photoreceptors provide the foundation of most of human visual experience, but because they are smaller and less numerous than rods in most mammalian retinas, much less is known about their physiology. We describe new techniques and approaches which are helping to provide a better understanding of cone function. We focus on several outstanding issues, including the identification of the features of the phototransduction cascade that are responsible for the more rapid kinetics and decreased sensitivity of the cone response, the roles of inner-segment voltage-gated and Ca2+-activated channels, the means by which cones remain responsive even in the brightest illumination, mechanisms of cone visual pigment regeneration in constant light, and energy consumption of cones in comparison to that of rods.
Collapse
|
27
|
Striebel JF, Race B, Leung JM, Schwartz C, Chesebro B. Prion-induced photoreceptor degeneration begins with misfolded prion protein accumulation in cones at two distinct sites: cilia and ribbon synapses. Acta Neuropathol Commun 2021; 9:17. [PMID: 33509294 PMCID: PMC7845122 DOI: 10.1186/s40478-021-01120-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/09/2021] [Indexed: 12/15/2022] Open
Abstract
Accumulation of misfolded host proteins is central to neuropathogenesis of numerous human brain diseases including prion and prion-like diseases. Neurons of retina are also affected by these diseases. Previously, our group and others found that prion-induced retinal damage to photoreceptor cells in mice and humans resembled pathology of human retinitis pigmentosa caused by mutations in retinal proteins. Here, using confocal, epifluorescent and electron microscopy we followed deposition of disease-associated prion protein (PrPSc) and its association with damage to critical retinal structures following intracerebral prion inoculation. The earliest time and place of retinal PrPSc deposition was 67 days post-inoculation (dpi) on the inner segment (IS) of cone photoreceptors. At 104 and 118 dpi, PrPSc was associated with the base of cilia and swollen cone inner segments, suggesting ciliopathy as a pathogenic mechanism. By 118 dpi, PrPSc was deposited in both rods and cones which showed rootlet damage in the IS, and photoreceptor cell death was indicated by thinning of the outer nuclear layer. In the outer plexiform layer (OPL) in uninfected mice, normal host PrP (PrPC) was mainly associated with cone bipolar cell processes, but in infected mice, at 118 dpi, PrPSc was detected on cone and rod bipolar cell dendrites extending into ribbon synapses. Loss of ribbon synapses in cone pedicles and rod spherules in the OPL was observed to precede destruction of most rods and cones over the next 2–3 weeks. However, bipolar cells and horizontal cells were less damaged, indicating high selectivity among neurons for injury by prions. PrPSc deposition in cone and rod inner segments and on the bipolar cell processes participating in ribbon synapses appear to be critical early events leading to damage and death of photoreceptors after prion infection. These mechanisms may also occur in human retinitis pigmentosa and prion-like diseases, such as AD.
Collapse
|
28
|
Stingl K, Kempf M, Bartz-Schmidt KU, Dimopoulos S, Reichel F, Jung R, Kelbsch C, Kohl S, Kortüm FC, Nasser F, Peters T, Wilhelm B, Wissinger B, Wozar F, Zrenner E, Fischer MD, Stingl K. Spatial and temporal resolution of the photoreceptors rescue dynamics after treatment with voretigene neparvovec. Br J Ophthalmol 2021; 106:831-838. [PMID: 33472769 PMCID: PMC9132865 DOI: 10.1136/bjophthalmol-2020-318286] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022]
Abstract
Background Voretigene neparvovec is a gene therapeutic agent for treatment of retinal dystrophies caused by bi-allelic RPE65 mutations. In this study, we report on a novel and objective evaluation of a retinotopic photoreceptor rescue. Methods Seven eyes of five patients (14, 21, 23, 24, 36 years, 1 male, 4 females) with bi-allelic RPE65 mutations have been treated with voretigene neparvovec. The clinical examinations included visual acuity testing, dark-adapted full-field stimulus threshold (FST), dark-adapted chromatic perimeter (DAC) with a 30-degree grid, and a 30 degrees grid scotopic and photopic chromatic pupil campimetry (CPC). All evaluations and spectral domain optical coherence tomography were performed at baseline, 1 month and 3 months. Results All except the oldest patient had a measurable improvement of the rod function assessed via FST, DAC or scotopic CPC at 1 month. The visual acuity improved slightly or remained stable in all eyes. A cone function improvement as measured by photopic CPC was observed in three eyes. The gain of the dark-adapted threshold with blue FST and the DAC stimuli (cyan) average correlated strongly with age (R2>0.7). The pupil response improvement in the scotopic CPC correlated with the baseline local retinal volume (R2=0.5). Conclusions The presented protocols allow evaluating the individual spatial and temporal effects of gene therapy effects. Additionally, we explored parameters that correlated with the success of the therapy. CPC and DAC present new and fast ways to assess functional changes in retinotopic maps of rod and cone function, measuring complementary aspects of retinal function.
Collapse
Affiliation(s)
- Krunoslav Stingl
- University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Center for Rare Eye Diseases, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Pupil Research Group at the University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Melanie Kempf
- University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Center for Rare Eye Diseases, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Karl U Bartz-Schmidt
- University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Spyridon Dimopoulos
- University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Felix Reichel
- University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Ronja Jung
- University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Carina Kelbsch
- University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Pupil Research Group at the University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Susanne Kohl
- Molecular Genetics Laboratory, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Friederike Charlotte Kortüm
- University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Fadi Nasser
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Tobias Peters
- Pupil Research Group at the University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- STZ eyetrial at the Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Barbara Wilhelm
- Pupil Research Group at the University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- STZ eyetrial at the Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Bernd Wissinger
- Molecular Genetics Laboratory, Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Fabian Wozar
- University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Eberhart Zrenner
- Center for Rare Eye Diseases, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Werner Reichardt Centre for Integrative Neuroscience (CIN), University of Tübingen, Tübingen, Germany
| | - M Dominik Fischer
- University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, Oxfordshire, UK
- Nuffield Laboratory of Ophthalmology, Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Katarina Stingl
- University Eye Hospital, Center for Ophthalmology, University of Tübingen, Tübingen, Baden-Württemberg, Germany
- Center for Rare Eye Diseases, University of Tübingen, Tübingen, Baden-Württemberg, Germany
| |
Collapse
|
29
|
Li Y, Cohen ED, Qian H. Rod and Cone Coupling Modulates Photopic ERG Responses in the Mouse Retina. Front Cell Neurosci 2020; 14:566712. [PMID: 33100974 PMCID: PMC7546330 DOI: 10.3389/fncel.2020.566712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/31/2020] [Indexed: 11/13/2022] Open
Abstract
Light adaptation changes both the sensitivity and maximum amplitude (Rmax) of the mouse photopic electroretinogram (ERG) b-wave. Using the ERG, we examined how modulation of gap junctional coupling between rod and cones alters the light-adapted ERG. To measure changes, a b-wave light adaptation enhancement factor (LAEF), was defined as the ratio of Rmax after 15 min light adaptation to Rmax recorded at the onset of an adapting light. For wild-type mice (WT), the LAEF averaged 2.64 ± 0.29, however, it was significantly reduced (1.06 ± 0.04) for connexin 36 knock out (Cx36KO) mice, which lack electrical coupling between photoreceptors. Wild type mice intraocularly injected with meclofenamic acid (MFA), a gap junction blocker, also showed a significantly reduced LAEF. Degeneration of rod photoreceptors significantly alters the effects of light adaptation on the photopic ERG response. Rd10 mice at P21, with large portions of their rod photoreceptors present in the retina, exhibited a similar b-wave enhancement as wildtype controls, with a LAEF of 2.55 ± 0.19. However, by P31 with most of their rod photoreceptors degenerated, rd10 mice had a much reduced b-wave enhancement during light-adaptation (LAEF of 1.54 ± 0.12). Flicker ERG responses showed a higher temporal amplitude in mesopic conditions for WT than those of Cx36KO mice, suggesting rod-cone coupling help high-frequency signals to pass from rods to cone pathways in the retina. In conclusion, our study provides a novel method to noninvasively measure the dynamics and modulation by the light adaptation for rod-cone gap junctional coupling in intact eyes.
Collapse
Affiliation(s)
- Yichao Li
- Visual Function Core, National Eye Institute (NEI), National Institutes of Health, Bethesda, MD, United States
| | - Ethan D Cohen
- Division of Biomedical Physics, Office of Science and Engineering Labs, Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, MD, United States
| | - Haohua Qian
- Visual Function Core, National Eye Institute (NEI), National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
30
|
Bonezzi PJ, Tarchick MJ, Renna JM. Ex vivo electroretinograms made easy: performing ERGs using 3D printed components. J Physiol 2020; 598:4821-4842. [PMID: 32886799 DOI: 10.1113/jp280014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 09/02/2020] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Rod and cone photoreceptors convert light into electrochemical signals that are transferred to second order cells, initiating image-forming visual processing. Electroretinograms (ERGs) can detect the associated light-induced extracellular transretinal events, allowing for physiological assessment of cellular activity from morphologically intact retinas. We outline a method for economically configuring a traditional patch-clamp rig for performing high signal-to-noise ex vivo ERGs. We accomplish this by incorporating various 3D printed components and by modifying existing light pathways in a typical patch-clamp rig. This methodology provides an additional set of tools to labs interested in studying the physiological function of neuronal populations in isolated retinal tissue. ABSTRACT Rod and cone photoreceptors of the retina are responsible for the initial stages in vision and convey sensory information regarding our visual world across a wide range of lighting conditions. These photoreceptors hyperpolarize in the presence of light and subsequently transmit signals to second-order bipolar and horizontal cells. The electrical components of these events are experimentally detectable, and in conjunction with pharmacological agents, can be further separated into their respective cellular contributions using electroretinograms (ERGs). Extracellular activity from populations of rods and cones generate the negative-going a-wave, while ON-bipolar cells generate positive-going b-waves. ERGs can be performed in vivo or alternatively using an ex vivo configuration, where retinas are isolated and transretinal photovoltages are recorded at high signal-to-noise ratios. However, most ERG set-ups require their own unique set of tools. We demonstrate how, at low cost, to reconfigure a typical patch-clamp rig for ERG recordings. The bulk of these modifications require implementation of various 3D printed components, which can alternatively aid in generating a stand-alone ERG set-up without a patch-rig. Further, we discuss how to configure an ERG system without a patch-clamp rig. Compared to in vivo ERGs, these are superior when measuring small responses, such as those that are cone-evoked or those from immature mouse retinae. This recording configuration provides high signal-to-noise detection of a-waves (300-600 µV) and b-waves (1-3 mV), and is ultimately capable of discerning small (1-2 µV) photovoltages from noise. These quick and economical modifications allow researchers to equip their technical arsenal with an interchangeable patch-clamp/ERG system.
Collapse
|
31
|
Reingruber J, Ingram NT, Griffis KG, Fain GL. A kinetic analysis of mouse rod and cone photoreceptor responses. J Physiol 2020; 598:3747-3763. [PMID: 32557629 PMCID: PMC7484371 DOI: 10.1113/jp279524] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 06/08/2020] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Most vertebrate eyes have rods for dim-light vision and cones for brighter light and higher temporal sensitivity. Rods evolved from cone-like precursors through expression of different transduction genes or the same genes at different expression levels, but we do not know which molecular differences were most important. We approached this problem by analysing rod and cone responses with the same model but with different values for model parameters. We showed that, in addition to outer-segment volume, the most important differences between rods and cones are: (1) decreased transduction gain, reflecting smaller amplification in the G-protein cascade; (2) a faster rate of turnover of the second messenger cGMP in darkness; and (3) an accelerated rate of decay of the effector enzyme phosphodiesterase and perhaps also of activated visual pigment. We believe our analysis has identified the principal alterations during evolution responsible for the duplex retina. ABSTRACT Most vertebrates have rod and cone photoreceptors, which differ in their sensitivity and response kinetics. We know that rods evolved from cone-like precursors through the expression of different transduction genes or the same genes at different levels, but we do not know which molecular differences were most important. We have approached this problem in mouse retina by analysing the kinetic differences between rod flash responses and recent voltage-clamp recordings of cone flash responses, using a model incorporating the principal features of photoreceptor transduction. We apply a novel method of analysis using the log-transform of the current, and we ask which of the model's dynamic parameters need be changed to transform the flash response of a rod into that of a cone. The most important changes are a decrease in the gain of the response, reflecting a reduction in amplification of the transduction cascade; an increase in the rate of turnover of cGMP in darkness; and an increase in the rate of decay of activated phosphodiesterase, with perhaps also an increase in the rate of decay of light-activated visual pigment. Although we cannot exclude other differences, and in particular alterations in the Ca2+ economy of the photoreceptors, we believe that we have identified the kinetic parameters principally responsible for the differences in the flash responses of the two kinds of photoreceptors, which were likely during evolution to have resulted in the duplex retina.
Collapse
Affiliation(s)
- Jürgen Reingruber
- Institut de Biologie de l’École Normale Supérieure, 46 rue d’Ulm, 75005 Paris, France
| | - Norianne T. Ingram
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095–7239, USA
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA 90095–7000, USA
| | - Khris G. Griffis
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA 90095–7000, USA
| | - Gordon L. Fain
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095–7239, USA
- Department of Ophthalmology and Jules Stein Eye Institute, University of California, Los Angeles, CA 90095–7000, USA
| |
Collapse
|
32
|
Abstract
We have used recent measurements of mammalian cone light responses and voltage-gated currents to calculate cone ATP utilization and compare it to that of rods. The largest expenditure of ATP results from ion transport, particularly from removal of Na+ entering outer segment light-dependent channels and inner segment hyperpolarization-activated cyclic nucleotide-gated channels, and from ATP-dependent pumping of Ca2+ entering voltage-gated channels at the synaptic terminal. Single cones expend nearly twice as much energy as single rods in darkness, largely because they make more synapses with second-order retinal cells and thus must extrude more Ca2+ In daylight, cone ATP utilization per cell remains high because cones never remain saturated and must continue to export Na+ and synaptic Ca2+ even in bright illumination. In mouse and human retina, rods greatly outnumber cones and consume more energy overall even in background light. In primates, however, the high density of cones in the fovea produces a pronounced peak of ATP utilization, which becomes particularly prominent in daylight and may make this part of the retina especially sensitive to changes in energy availability.
Collapse
|
33
|
Chen NS, Ingram NT, Frederiksen R, Sampath AP, Chen J, Fain GL. Diminished Cone Sensitivity in cpfl3 Mice Is Caused by Defective Transducin Signaling. Invest Ophthalmol Vis Sci 2020; 61:26. [PMID: 32315379 PMCID: PMC7401474 DOI: 10.1167/iovs.61.4.26] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Purpose Cone photoreceptor function loss 3 (Gnat2cpfl3/cpfl3 or cpfl3) is a mouse model commonly used as a functional cones null from a naturally occurring mutation in the α-subunit of cone transducin (Gnat2). We nevertheless detected robust cone-mediated light responses from cpfl3 animals, which we now explore. Methods Recordings were made from whole retina and from identified cones with whole-cell patch clamp in retinal slices. Relative levels of GNAT2 protein and numbers of cones in isolated retinas were compared between cpfl3, rod transducin knockout (Gnat1-/-), cpfl3/Gnat1-/- double mutants, and control C57Bl/6J age-matched mice at 4, 9, and 14 weeks of age. Results Cones from cpfl3 and cpfl3/Gnat1-/- mice 2 to 3 months of age displayed normal dark currents but greatly reduced sensitivity and amplification constants. Responses decayed more slowly than in control (C57Bl/6J) mice, indicating an altered mechanism of inactivation. At dim light intensities rod responses could be recorded from cpfl3 cones, indicating intact rod/cone gap junctions. The cpfl3 and cpfl3/Gnat1-/- mice express two-fold less GNAT2 protein compared with C57 at 4 weeks, and a four-fold decrease by 14 weeks. This is accompanied by a small decrease in the number of cones. Conclusions Cplf3 cones can respond to light with currents of normal amplitude and cannot be assumed to be a Gnat2 null. The decreased sensitivity and amplification rate of cones is not explained by a reduction in GNAT2 protein level, but instead by abnormal interactions of the mutant transducin with rhodopsin and the effector molecule, cGMP phosphodiesterase.
Collapse
Affiliation(s)
- Natalie S. Chen
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Norianne T. Ingram
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine, Los Angeles, California, United States,Department of Integrative Biology and Physiology, University of California, Los Angeles, California,United States
| | - Rikard Frederiksen
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine, Los Angeles, California, United States
| | - Alapakkam P. Sampath
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine, Los Angeles, California, United States
| | - Jeannie Chen
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, United States
| | - Gordon L. Fain
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine, Los Angeles, California, United States,Department of Integrative Biology and Physiology, University of California, Los Angeles, California,United States
| |
Collapse
|
34
|
Elsner AE, Papay JA, Johnston KD, Sawides L, de Castro A, King BJ, Jones DW, Clark CA, Gast TJ, Burns SA. Cones in ageing and harsh environments: the neural economy hypothesis. Ophthalmic Physiol Opt 2020; 40:88-116. [PMID: 32017191 PMCID: PMC7155023 DOI: 10.1111/opo.12670] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/31/2019] [Indexed: 01/22/2023]
Abstract
PURPOSE Cones are at great risk in a wide variety of retinal diseases, especially when there is a harsh microenvironment and retinal pigment epithelium is damaged. We provide established and new methods for assessing cones and retinal pigment epithelium, together with new results. We investigated conditions under which cones can be imaged and could guide light, despite the proximity of less than ideal retinal pigment epithelium. RECENT FINDINGS We used a variety of imaging methods to detect and localise damage to the retinal pigment epithelium. As age-related macular degeneration is a particularly widespread disease, we imaged clinical hallmarks: drusen and hyperpigmentation. Using near infrared light provided improved imaging of the deeper fundus layers. We compared confocal and multiply scattered light images, using both the variation of detection apertures and polarisation analysis. We used optical coherence tomography to examine distances between structures and thickness of retinal layers, as well as identifying damage to the retinal pigment epithelium. We counted cones using adaptive optics scanning laser ophthalmoscopy. We compared the results of five subjects with geographic atrophy to data from a previous normative ageing study. Using near infrared imaging and layer analysis of optical coherence tomography, the widespread aspect of drusen became evident. Both multiply scattered light imaging and analysis of the volume in the retinal pigment epithelial layer from the optical coherence tomography were effective in localising drusen and hyperpigmentation beneath the photoreceptors. Cone photoreceptors in normal older eyes were shorter than in younger eyes. Cone photoreceptors survived in regions of atrophy, but with greatly reduced and highly variable density. Regular arrays of cones were found in some locations, despite abnormal retinal pigment epithelium. For some subjects, the cone density was significantly greater than normative values in some retinal locations outside the atrophy. SUMMARY The survival of cones within atrophy is remarkable. The unusually dense packing of cones at some retinal locations outside the atrophy indicates more fluidity in cone distribution than typically thought. Together these findings suggest strategies for therapy that includes preserving cones.
Collapse
|
35
|
Light stress affects cones and horizontal cells via rhodopsin-mediated mechanisms. Exp Eye Res 2019; 186:107719. [PMID: 31291592 DOI: 10.1016/j.exer.2019.107719] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/04/2019] [Accepted: 07/04/2019] [Indexed: 01/20/2023]
Abstract
Retinal degenerations are a major cause of blindness in human patients. The identification of endogenous mechanisms involved in neurodegeneration or neuroprotection helps to understand the response of the retina to stress and provides essential information not only for basic retinal physiology but also for defining molecular targets for neuroprotective strategies. Here we used excessive light exposure as a model system to study mechanisms of photoreceptor degeneration in mice. Using one wild type and four genetically modified mouse strains, we demonstrate that light exposure resulted not only in the degeneration of rods but also in an early but transient repression of several cone-specific genes, in a reversible hyperreflectivity of the outer retina including the outer plexiform layer, and in the loss of horizontal cells. The effects on cones, horizontal cells and the inner retina depended on light absorption by rhodopsin and, at least partially, on leukemia inhibitory factor. This demonstrates the existence of intercellular communication routes that transduce rod stress to other cells, likely to provide support for photoreceptors and increase cell survival in the injured retina.
Collapse
|
36
|
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: 72] [Impact Index Per Article: 14.4] [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
|
37
|
Roy S, Field GD. Dopaminergic modulation of retinal processing from starlight to sunlight. J Pharmacol Sci 2019; 140:86-93. [PMID: 31109761 DOI: 10.1016/j.jphs.2019.03.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/13/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022] Open
Abstract
Neuromodulators such as dopamine, enable context-dependent plasticity of neural circuit function throughout the central nervous system. For example, in the retina, dopamine tunes visual processing for daylight and nightlight conditions. Specifically, high levels of dopamine release in the retina tune vision for daylight (photopic) conditions, while low levels tune it for nightlight (scotopic) conditions. This review covers the cellular and circuit-level mechanisms within the retina that are altered by dopamine. These mechanisms include changes in gap junction coupling and ionic conductances, both of which are altered by the activation of diverse types of dopamine receptors across diverse types of retinal neurons. We contextualize the modulatory actions of dopamine in terms of alterations and optimizations to visual processing under photopic and scotopic conditions, with particular attention to how they differentially impact distinct cell types. Finally, we discuss how transgenic mice and disease models have shaped our understanding of dopaminergic signaling and its role in visual processing. Cumulatively, this review illustrates some of the diverse and potent mechanisms through which neuromodulation can shape brain function.
Collapse
Affiliation(s)
- Suva Roy
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA
| | - Greg D Field
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
38
|
Increase in Mutual Information During Interaction with the Environment Contributes to Perception. ENTROPY 2019; 21:e21040365. [PMID: 33267079 PMCID: PMC7514849 DOI: 10.3390/e21040365] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/22/2019] [Accepted: 04/02/2019] [Indexed: 02/04/2023]
Abstract
Perception and motor interaction with physical surroundings can be analyzed by the changes in probability laws governing two possible outcomes of neuronal activity, namely the presence or absence of spikes (binary states). Perception and motor interaction with the physical environment are partly accounted for by a reduction in entropy within the probability distributions of binary states of neurons in distributed neural circuits, given the knowledge about the characteristics of stimuli in physical surroundings. This reduction in the total entropy of multiple pairs of circuits in networks, by an amount equal to the increase of mutual information, occurs as sensory information is processed successively from lower to higher cortical areas or between different areas at the same hierarchical level, but belonging to different networks. The increase in mutual information is partly accounted for by temporal coupling as well as synaptic connections as proposed by Bahmer and Gupta (Front. Neurosci. 2018). We propose that robust increases in mutual information, measuring the association between the characteristics of sensory inputs' and neural circuits' connectivity patterns, are partly responsible for perception and successful motor interactions with physical surroundings. The increase in mutual information, given the knowledge about environmental sensory stimuli and the type of motor response produced, is responsible for the coupling between action and perception. In addition, the processing of sensory inputs within neural circuits, with no prior knowledge of the occurrence of a sensory stimulus, increases Shannon information. Consequently, the increase in surprise serves to increase the evidence of the sensory model of physical surroundings.
Collapse
|
39
|
On the origin of proteins in human drusen: The meet, greet and stick hypothesis. Prog Retin Eye Res 2018; 70:55-84. [PMID: 30572124 DOI: 10.1016/j.preteyeres.2018.12.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022]
Abstract
Retinal drusen formation is not only a clinical hallmark for the development of age-related macular degeneration (AMD) but also for other disorders, such as Alzheimer's disease and renal diseases. The initiation and growth of drusen is poorly understood. Attention has focused on lipids and minerals, but relatively little is known about the origin of drusen-associated proteins and how they are retained in the space between the basal lamina of the retinal pigment epithelium and the inner collagenous layer space (sub-RPE-BL space). While some authors suggested that drusen proteins are mainly derived from cellular debris from processed photoreceptor outer segments and the RPE, others suggest a choroidal cell or blood origin. Here, we reviewed and supplemented the existing literature on the molecular composition of the retina/choroid complex, to gain a more complete understanding of the sources of proteins in drusen. These "drusenomics" studies showed that a considerable proportion of currently identified drusen proteins is uniquely originating from the blood. A smaller, but still large fraction of drusen proteins comes from both blood and/or RPE. Only a small proportion of drusen proteins is uniquely derived from the photoreceptors or choroid. We next evaluated how drusen components may "meet, greet and stick" to each other and/or to structures like hydroxyapatite spherules to form macroscopic deposits in the sub-RPE-BL space. Finally, we discuss implications of our findings with respect to the previously proposed homology between drusenogenesis in AMD and plaque formation in atherosclerosis.
Collapse
|