1
|
Sims R, Lin B, Xue Y, Fouda R, McLelland BT, Nistor G, Keirstead HS, Browne AW, Seiler MJ. Effect of immunosuppression on hESC-derived retina organoids in vitro and in vivo. Stem Cell Res Ther 2025; 16:165. [PMID: 40188342 PMCID: PMC11972460 DOI: 10.1186/s13287-025-04271-z] [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: 11/13/2024] [Accepted: 03/11/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Photoreceptor (PR) enriched retinal organoid (RO) sheets (human embryonic stem cell [hESC]-derived ROs) resulted in restoration of visual acuity in immunocompromised retinal degenerate (RD) animal models after transplantation. Further assessment of their clinical potential requires evaluation in immunocompetent RD disease models with effective immune suppression. We characterized safety and efficacy profiles of both donor tissues and prospective immunosuppressive treatments in vitro; and in vivo in immunocompetent RD rats (strain SD-foxn1 Tg(S334ter)3Lav). METHODS Retinal identity of ROs was validated by histology, flow cytometry and gene expression profiling, and their immunogenicity to sensitized human immune cells was measured by mixed lymphocyte reactions (MLR). We measured the effect of RO exposure for 1-4 weeks to therapeutic concentrations of our immunosuppressant drugs of choice on gene expression and metabolic function using quantitative PCR (qPCR) and functional and structural fluorescence lifetime imaging (FLIM), respectively. Immunocompetent RD graft recipients were immunosuppressed by implanted tacrolimus (TAC) pellets and mycophenolate mofetil (MMF) in food. In vivo, LCMS aided assessments of drug pharmacodynamics. Flow cytometry immunophenotyping and assay of post-surgery cytokines were used to assess and monitor drug efficacy. Retinal transplants were imaged in situ using optical coherence tomography (OCT) at defined time points post-surgery. Visual function was assessed by optokinetic tests (OKT) and superior colliculus electrophysiology recording. At study endpoints, immune cell infiltration and donor photoreceptor engraftment into host retinal architecture was evaluated by immunohistochemistry. RESULTS Immunosuppressive drugs have no negative effects on RO development and metabolism in vitro; and low alloreactivity of ROs determined by MLR may be predictive to that of human graft recipients. In vivo, minimum effective dosing ranges of TAC and MMF were determined. We characterized the mechanisms and critical immune populations implicated in rejection; and subsequently demonstrated their effective suppression in our xenograft RD model. OKT measured significant visual improvement after RO transplantation. Transplants developed most retinal cell types including photoreceptors; and integrated with the host retina. However, immunosuppression induced higher sensitivity to ketamine anesthesia. CONCLUSIONS This study proves the concept that immunosuppression is likely tolerable in retinal transplantation and human stem cell therapy for retinal degeneration patients.
Collapse
Affiliation(s)
- Robert Sims
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, 845 Health Sciences Road, Irvine, CA, 92697-1705, USA
- Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA
- Physical Medicine and Rehabilitation, University of California Irvine, Irvine, CA, USA
- Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Bin Lin
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, 845 Health Sciences Road, Irvine, CA, 92697-1705, USA
- Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA
- Physical Medicine and Rehabilitation, University of California Irvine, Irvine, CA, USA
- Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Yuntian Xue
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, 845 Health Sciences Road, Irvine, CA, 92697-1705, USA
- Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Raghda Fouda
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, 845 Health Sciences Road, Irvine, CA, 92697-1705, USA
| | | | | | | | - Andrew W Browne
- Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA
- Center for Translational Vision Research, University of California Irvine, Irvine, CA, USA
- Biomedical Engineering, University of California Irvine, Irvine, CA, USA
- Institute for Clinical and Translational Science, University of California Irvine, Irvine, CA, USA
| | - Magdalene J Seiler
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, 845 Health Sciences Road, Irvine, CA, 92697-1705, USA.
- Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA.
- Physical Medicine and Rehabilitation, University of California Irvine, Irvine, CA, USA.
- Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA.
- Center for Translational Vision Research, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
2
|
Bills JD, Seifert AW, Morris AC. Retinal neuroanatomy of two emerging model organisms, the spiny mouse (Acomys dimidiatus) and the Mongolian gerbil (Meriones unguiculatus). Exp Eye Res 2024; 247:110055. [PMID: 39159803 DOI: 10.1016/j.exer.2024.110055] [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: 04/15/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Current research using animal models to investigate retinal cell biology and model retinal degenerative diseases largely utilize small mammals that are nocturnal and lack the ability to restore lost vision. In contrast, the Mongolian gerbil (Meriones) is a diurnal rodent with good photopic vision, and the spiny mouse (Acomys) is a small desert-dwelling rodent with remarkable regenerative capabilities. The goal of this study was to identify antibodies that detect retinal cell classes in Meriones and Acomys, and to describe the retinal anatomy of these two species in comparison to outbred laboratory mice (Mus musculus). Immunohistochemistry was performed on retinal sections with antibodies for various retinal cell types. Sections were imaged by light, fluorescence, and confocal microscopy. Cell density, morphology, and placement were compared between species qualitatively and quantitatively. Our analyses revealed a classic assembly of retinal cells in Meriones and Acomys, with a few deviations compared to Mus. Meriones displayed the highest density of cones and Acomys the lowest. A higher density of bipolar cell bodies in the proximal portion of the inner nuclear layer was observed in both Acomys and Meriones compared to Mus, and both species exhibited an increase in amacrine cell density compared to Mus. Our results provide a foundation for future research into the visual system adaptations of these interesting species.
Collapse
Affiliation(s)
- Jessica D Bills
- Department of Biology, University of Kentucky, Lexington, KY 40506-0225, USA
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, KY 40506-0225, USA
| | - Ann C Morris
- Department of Biology, University of Kentucky, Lexington, KY 40506-0225, USA.
| |
Collapse
|
3
|
Batabyal S, Kim S, Carlson M, Narcisse D, Tchedre K, Dibas A, Sharif NA, Mohanty S. Multi-Characteristic Opsin Therapy to Functionalize Retina, Attenuate Retinal Degeneration, and Restore Vision in Mouse Models of Retinitis Pigmentosa. Transl Vis Sci Technol 2024; 13:25. [PMID: 39412768 PMCID: PMC11486081 DOI: 10.1167/tvst.13.10.25] [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: 12/13/2023] [Accepted: 08/13/2024] [Indexed: 10/19/2024] Open
Abstract
Purpose Retinal degeneration 1 and 10 (rd1 and rd10) mice are useful animal models of retinitis pigmentosa (RP) with rapidly and slowly progressive pathologies, respectively. Our study aims were to determine the effect of adeno-associated viral vector 2 (AAV2)-delivered multi-characteristic opsin (MCO-010; under the control of a metabotropic glutamate receptor-6 promoter enhancer) on the morphological and functional characteristics of vision in both rd1 and rd10 mice. Methods Various retinal measures of MCO-010 transduction and electrophysiological, behavioral, and other routine blood analyses were performed in the rd1 and/or rd10 mice after intravitreal injection of 1 µL of MCO-010 or AAV2 vehicle. Functional tests included electroretinogram, visually evoked potential, and behavior assay (optomotor and water maze). Retinal thickness, intraocular pressure, and plasma cytokine levels were also determined. Results Following intravitreal MCO-010 injection, approximately 80% of bipolar cells were transduced in the retina, and no alterations in retinal thickness were observed at 4 months post-injection. However, retinal thickness significantly decreased in control mice. MCO-010 treatment increased head movements and induced faster navigation of mice to the platform in a water-maze test. The MCO-010 gene therapy helped preserve visually evoked electrical response in the retina and visual cortex. No ocular toxicity, immunotoxicity, or phototoxicity was observed in the MCO-010-treated mice, even under chronic intense light conditions. Conclusions Intravitreal MCO-010 was well tolerated in rd1 and rd10 mice models of RP, and it appeared to attenuate retinal photoreceptor degeneration based on retinal structure and functional outcome measures. Translational Relevance As reported here, optogenetic treatment of the inner retina attenuates further retinal degeneration in addition to photosensitizing higher order neurons, and this disease-modifying aspect should be evaluated in optogenetic clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Adnan Dibas
- Nanoscope Technologies LLC, Bedford, TX, USA
| | | | - Samarendra Mohanty
- Nanoscope Technologies LLC, Bedford, TX, USA
- Nanoscope Therapeutics, Inc., Dallas, TX, USA
| |
Collapse
|
4
|
Estay SF, Morales-Moraga C, Vielma AH, Palacios-Muñoz A, Chiu CQ, Chávez AE. Non-canonical type 1 cannabinoid receptor signaling regulates night visual processing in the inner rat retina. iScience 2024; 27:109920. [PMID: 38799553 PMCID: PMC11126983 DOI: 10.1016/j.isci.2024.109920] [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: 02/28/2024] [Revised: 04/18/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024] Open
Abstract
Type 1 cannabinoid receptors (CB1Rs) are expressed in major retinal neurons within the rod-pathway suggesting a role in regulating night visual processing, but the underlying mechanisms remain poorly understood. Using acute rat retinal slices, we show that CB1R activation reduces glutamate release from rod bipolar cell (RBC) axon terminals onto AII and A17 amacrine cells through a pathway that requires exchange proteins directly activated by cAMP (EPAC1/2) signaling. Consequently, CB1R activation abrogates reciprocal GABAergic feedback inhibition from A17 amacrine cells. Moreover, the activation of CB1Rs in vivo enhances and prolongs the time course of the dim-light rod-driven visual responses, an effect that was eliminated when both GABAA and GABAC receptors were blocked. Altogether, our findings underscore a non-canonical mechanism by which cannabinoid signaling regulates RBC dyad synapses in the inner retina to regulate dim-light visual responses to fine-tune night vision.
Collapse
Affiliation(s)
- Sebastián F. Estay
- Programa de Doctorado en Ciencias, Mención Neurociencia, Valparaíso 2340000, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Camila Morales-Moraga
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Alex H. Vielma
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Angelina Palacios-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Chiayu Q. Chiu
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| | - Andrés E. Chávez
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2340000, Chile
| |
Collapse
|
5
|
Aísa-Marín I, Rovira Q, Díaz N, Calvo-López L, Vaquerizas JM, Marfany G. Specific photoreceptor cell fate pathways are differentially altered in NR2E3-associated diseases. Neurobiol Dis 2024; 194:106463. [PMID: 38485095 DOI: 10.1016/j.nbd.2024.106463] [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: 12/27/2023] [Revised: 03/01/2024] [Accepted: 03/02/2024] [Indexed: 03/21/2024] Open
Abstract
Mutations in NR2E3, a gene encoding an orphan nuclear transcription factor, cause two retinal dystrophies with a distinct phenotype, but the precise role of NR2E3 in rod and cone transcriptional networks remains unclear. To dissect NR2E3 function, we performed scRNA-seq in the retinas of wildtype and two different Nr2e3 mouse models that show phenotypes similar to patients carrying NR2E3 mutations. Our results reveal that rod and cone populations are not homogeneous and can be separated into different sub-classes. We identify a previously unreported cone pathway that generates hybrid cones co-expressing both cone- and rod-related genes. In mutant retinas, this hybrid cone subpopulation is more abundant and includes a subpopulation of rods transitioning towards a cone cell fate. Hybrid photoreceptors with high misexpression of cone- and rod-related genes are prone to regulated necrosis. Overall, our results shed light on the role of NR2E3 in modulating photoreceptor differentiation towards cone and rod fates and explain how different mutations in NR2E3 lead to distinct visual disorders in humans.
Collapse
Affiliation(s)
- Izarbe Aísa-Marín
- Department de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Barcelona 08028, Spain; IBUB-IRSJD, Institut de Biomedicina de la Universitat de Barcelona-Institut de Recerca Sant Joan de Déu, Barcelona 08028, Spain; CIBERER, Instituto de Salud Carlos III, Barcelona 08028, Spain
| | - Quirze Rovira
- Max-Planck-Institute for Molecular Biomedicine, Münster 48149, Germany
| | - Noelia Díaz
- Max-Planck-Institute for Molecular Biomedicine, Münster 48149, Germany
| | - Laura Calvo-López
- Department de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Barcelona 08028, Spain
| | - Juan M Vaquerizas
- Max-Planck-Institute for Molecular Biomedicine, Münster 48149, Germany; MRC London Institute of Medical Sciences, Institute of Clinical Sciences, Imperial College London, London W12 0NN, UK.; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK.
| | - Gemma Marfany
- Department de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Barcelona 08028, Spain; IBUB-IRSJD, Institut de Biomedicina de la Universitat de Barcelona-Institut de Recerca Sant Joan de Déu, Barcelona 08028, Spain; CIBERER, Instituto de Salud Carlos III, Barcelona 08028, Spain; DBGen Ocular Genomics, Barcelona 08028, Spain.
| |
Collapse
|
6
|
Cheng SY, Caiazzi J, Biscans A, Alterman JF, Echeverria D, McHugh N, Hassler M, Jolly S, Giguere D, Cipi J, Khvorova A, Punzo C. Single intravitreal administration of a tetravalent siRNA exhibits robust and efficient gene silencing in mouse and pig photoreceptors. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102088. [PMID: 38192611 PMCID: PMC10772295 DOI: 10.1016/j.omtn.2023.102088] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/30/2023] [Indexed: 01/10/2024]
Abstract
Inherited retinal dystrophies caused by dominant mutations in photoreceptor (PR) cell expressed genes are a major cause of irreversible vision loss. Oligonucleotide therapy has been of interest in diseases that conventional medicine cannot target. In the early days, small interfering RNAs (siRNAs) were explored in clinical trials for retinal disorders with limited success due to a lack of stability and efficient cellular delivery. Thus, an unmet need exists to identify siRNA chemistry that targets PR cell expressed genes. Here, we evaluated 12 different fully chemically modified siRNA configurations, where the valency and conjugate structure were systematically altered. The impact on retinal distribution following intravitreal delivery was examined. We found that the increase in valency (tetravalent siRNA) supports the best PR accumulation. A single intravitreal administration induces multimonths efficacy in rodent and porcine retinas while demonstrating a good safety profile. The data suggest that this configuration can treat retinal diseases caused by PR cell expressed genes with 1-2 intravitreal injections per year.
Collapse
Affiliation(s)
- Shun-Yun Cheng
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jillian Caiazzi
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Annabelle Biscans
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Julia F. Alterman
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Dimas Echeverria
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Nicholas McHugh
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Matthew Hassler
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Samson Jolly
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Delaney Giguere
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Joris Cipi
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Claudio Punzo
- Department of Ophthalmology and Visual Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
7
|
Sturgis J, Singh R, Caron Q, Samuels IS, Shiju TM, Mukkara A, Freedman P, Bonilha VL. Modeling aging and retinal degeneration with mitochondrial DNA mutation burden. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.30.569464. [PMID: 38076962 PMCID: PMC10705408 DOI: 10.1101/2023.11.30.569464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Somatic mitochondrial DNA (mtDNA) mutation accumulation has been observed in individuals with retinal degenerative disorders. To study the effects of aging and mtDNA mutation accumulation in the retina, a Polymerase gamma (POLG) deficiency model, the POLGD257A mutator mice (PolgD257A), was used. POLG is an enzyme responsible for regulating mtDNA replication and repair. Retinas of young and older mice with this mutation were analyzed in vivo and ex vivo to provide new insights into the contribution of age-related mitochondrial dysfunction due to mtDNA damage. Optical coherence tomography (OCT) image analysis revealed a decrease in retinal and photoreceptor thickness starting at 6 months of age in mice with the POLGD257A mutation compared to wild-type (WT) mice. Electroretinography (ERG) testing showed a significant decrease in all recorded responses at 6 months of age. Sections labeled with markers of different types of retinal cells, including cones, rods, and bipolar cells, exhibited decreased labeling starting at 6 months. However, electron microscopy analysis revealed differences in retinal pigment epithelium (RPE) mitochondria morphology beginning at 3 months. Interestingly, there was no increase in oxidative stress observed in the retina or RPE of POLGD257A mice. Additionally, POLGD257A RPE exhibited an accelerated rate of autofluorescence cytoplasmic granule formation and accumulation. Mitochondrial markers displayed decreased abundance in protein lysates obtained from retina and RPE samples. These findings suggest that the accumulation of mitochondrial DNA mutations leads to impaired mitochondrial function and accelerated aging, resulting in retinal degeneration.
Collapse
Affiliation(s)
- John Sturgis
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Rupesh Singh
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Quinn Caron
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ivy S. Samuels
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH
| | - Thomas Micheal Shiju
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Aditi Mukkara
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- College of Arts and Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Paul Freedman
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Debusk College of Osteopathic Medicine, Knoxville, TN, USA
| | - Vera L. Bonilha
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
8
|
Fietz A, Schnichels S, Hurst J. Co-cultivation of primary porcine RPE cells and neuroretina induces inflammation: a potential inflammatory AMD-model. Sci Rep 2023; 13:19345. [PMID: 37935821 PMCID: PMC10630302 DOI: 10.1038/s41598-023-46029-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/26/2023] [Indexed: 11/09/2023] Open
Abstract
One common aspect in the pathology of many retinal diseases like age-related macular degeneration (AMD) is the death of retinal pigment epithelium (RPE) cells. RPE cells are essential for photoreceptor survival as they recycle and remove compounds of the visual cycle and secrete protective cytokines. Studying RPE cells is crucial to improve our understanding of retinal pathologies, yet only a few retinal ex vivo models include them or do so only indirectly. Besides the positive effects in indirect co-cultivation models, also a slight inflammation was observed. In this study we developed an ex vivo model consisting of a primary porcine RPE monolayer directly co-cultured with porcine retinal organ cultures, to investigate and simulate inflammatory retinal diseases, such as (dry) AMD. The direct co-cultivation resulted in immune reactivity (enhanced expression of pro-inflammatory cytokines e.g., IL-1β, IL-6, IL-8) and cell death. These effects were evaluated for the retinal explant as well as for the RPE-monolayer to further understand the complex interactions between these two compartments. Taken together, this ex vivo model can be used to study inflammatory retinal diseases like AMD as well as the rejection observed after RPE-transplantation.
Collapse
Affiliation(s)
- Agnes Fietz
- Centre for Ophthalmology, University Eye Hospital Tübingen, 72076, Tübingen, Germany
| | - Sven Schnichels
- Centre for Ophthalmology, University Eye Hospital Tübingen, 72076, Tübingen, Germany.
| | - José Hurst
- Centre for Ophthalmology, University Eye Hospital Tübingen, 72076, Tübingen, Germany
| |
Collapse
|
9
|
Cheng SY, Caiazzi J, Biscans A, Alterman JF, Echeverria D, McHugh N, Hassler M, Jolly S, Giguere D, Cipi J, Khvorova A, Punzo C. Single intravitreal administration of a tetravalent siRNA exhibits robust and efficient gene silencing in rodent and swine photoreceptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.20.558641. [PMID: 37790464 PMCID: PMC10542117 DOI: 10.1101/2023.09.20.558641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Inherited retinal dystrophies caused by dominant mutations in photoreceptor-expressed genes, are a major cause of irreversible vision loss. Oligonucleotide therapy has been of interest in diseases that conventional medicine cannot target. In the early days, small interfering RNAs (siRNAs) were explored in clinical trials for retinal disorders with limited success due to a lack of stability and efficient cellular delivery. Thus, an unmet need exists to identify siRNA chemistry that targets photoreceptor-expressed genes. Here we evaluated 12 different fully chemically modified siRNA configurations, where the valency and conjugate structure were systematically altered. The impact on retinal distribution following intravitreal delivery was examined. We found that the increase in valency (tetravalent siRNA) supports the best photoreceptor accumulation. A single intravitreal administration induces multi-months efficacy in rodent and porcine retinas while showing a good safety profile. The data suggest that this configuration can treat retinal diseases caused by photoreceptor-expressed genes with 1-2 intravitreal injections per year.
Collapse
|
10
|
Wang C, Fang C, Zou Y, Yang J, Sawan M. SpikeSEE: An energy-efficient dynamic scenes processing framework for retinal prostheses. Neural Netw 2023; 164:357-368. [PMID: 37167749 DOI: 10.1016/j.neunet.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/13/2023]
Abstract
Intelligent and low-power retinal prostheses are highly demanded in this era, where wearable and implantable devices are used for numerous healthcare applications. In this paper, we propose an energy-efficient dynamic scenes processing framework (SpikeSEE) that combines a spike representation encoding technique and a bio-inspired spiking recurrent neural network (SRNN) model to achieve intelligent processing and extreme low-power computation for retinal prostheses. The spike representation encoding technique could interpret dynamic scenes with sparse spike trains, decreasing the data volume. The SRNN model, inspired by the human retina's special structure and spike processing method, is adopted to predict the response of ganglion cells to dynamic scenes. Experimental results show that the Pearson correlation coefficient of the proposed SRNN model achieves 0.93, which outperforms the state-of-the-art processing framework for retinal prostheses. Thanks to the spike representation and SRNN processing, the model can extract visual features in a multiplication-free fashion. The framework achieves 8 times power reduction compared with the convolutional recurrent neural network (CRNN) processing-based framework. Our proposed SpikeSEE predicts the response of ganglion cells more accurately with lower energy consumption, which alleviates the precision and power issues of retinal prostheses and provides a potential solution for wearable or implantable prostheses.
Collapse
Affiliation(s)
- Chuanqing Wang
- Center of Excellence in Biomedical Research on Advanced Integrated-on-chips Neurotechnologies, School of Engineering, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Chaoming Fang
- Center of Excellence in Biomedical Research on Advanced Integrated-on-chips Neurotechnologies, School of Engineering, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Yong Zou
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jie Yang
- Center of Excellence in Biomedical Research on Advanced Integrated-on-chips Neurotechnologies, School of Engineering, Westlake University, Hangzhou, 310024, Zhejiang, China.
| | - Mohamad Sawan
- Center of Excellence in Biomedical Research on Advanced Integrated-on-chips Neurotechnologies, School of Engineering, Westlake University, Hangzhou, 310024, Zhejiang, China.
| |
Collapse
|
11
|
Wang C, Fang C, Zou Y, Yang J, Sawan M. Artificial intelligence techniques for retinal prostheses: a comprehensive review and future direction. J Neural Eng 2023; 20. [PMID: 36634357 DOI: 10.1088/1741-2552/acb295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/12/2023] [Indexed: 01/14/2023]
Abstract
Objective. Retinal prostheses are promising devices to restore vision for patients with severe age-related macular degeneration or retinitis pigmentosa disease. The visual processing mechanism embodied in retinal prostheses play an important role in the restoration effect. Its performance depends on our understanding of the retina's working mechanism and the evolvement of computer vision models. Recently, remarkable progress has been made in the field of processing algorithm for retinal prostheses where the new discovery of the retina's working principle and state-of-the-arts computer vision models are combined together.Approach. We investigated the related research on artificial intelligence techniques for retinal prostheses. The processing algorithm in these studies could be attributed to three types: computer vision-related methods, biophysical models, and deep learning models.Main results. In this review, we first illustrate the structure and function of the normal and degenerated retina, then demonstrate the vision rehabilitation mechanism of three representative retinal prostheses. It is necessary to summarize the computational frameworks abstracted from the normal retina. In addition, the development and feature of three types of different processing algorithms are summarized. Finally, we analyze the bottleneck in existing algorithms and propose our prospect about the future directions to improve the restoration effect.Significance. This review systematically summarizes existing processing models for predicting the response of the retina to external stimuli. What's more, the suggestions for future direction may inspire researchers in this field to design better algorithms for retinal prostheses.
Collapse
Affiliation(s)
- Chuanqing Wang
- Center of Excellence in Biomedical Research on Advanced Integrated-on-chips Neurotechnologies, School of Engineering, Westlake University, Hangzhou 310030, People's Republic of China
| | - Chaoming Fang
- Center of Excellence in Biomedical Research on Advanced Integrated-on-chips Neurotechnologies, School of Engineering, Westlake University, Hangzhou 310030, People's Republic of China
| | - Yong Zou
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Jie Yang
- Center of Excellence in Biomedical Research on Advanced Integrated-on-chips Neurotechnologies, School of Engineering, Westlake University, Hangzhou 310030, People's Republic of China
| | - Mohamad Sawan
- Center of Excellence in Biomedical Research on Advanced Integrated-on-chips Neurotechnologies, School of Engineering, Westlake University, Hangzhou 310030, People's Republic of China
| |
Collapse
|
12
|
Hu H, Long Y, Song G, Chen S, Xu Z, Li Q, Wu Z. Dysfunction of Prkcaa Links Social Behavior Defects with Disturbed Circadian Rhythm in Zebrafish. Int J Mol Sci 2023; 24:ijms24043849. [PMID: 36835261 PMCID: PMC9961154 DOI: 10.3390/ijms24043849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
Protein kinase Cα (PKCα/PRKCA) is a crucial regulator of circadian rhythm and is associated with human mental illnesses such as autism spectrum disorders and schizophrenia. However, the roles of PRKCA in modulating animal social behavior and the underlying mechanisms remain to be explored. Here we report the generation and characterization of prkcaa-deficient zebrafish (Danio rerio). The results of behavioral tests indicate that a deficiency in Prkcaa led to anxiety-like behavior and impaired social preference in zebrafish. RNA-sequencing analyses revealed the significant effects of the prkcaa mutation on the expression of the morning-preferring circadian genes. The representatives are the immediate early genes, including egr2a, egr4, fosaa, fosab and npas4a. The downregulation of these genes at night was attenuated by Prkcaa dysfunction. Consistently, the mutants demonstrated reversed day-night locomotor rhythm, which are more active at night than in the morning. Our data show the roles of PRKCA in regulating animal social interactions and link the social behavior defects with a disturbed circadian rhythm.
Collapse
Affiliation(s)
- Han Hu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Research Center of Fishery Resources and Environment, Southwest University, Chongqing 400715, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Yong Long
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- Correspondence: (Y.L.); (Z.W.); Tel.: +86-27-6878-0100 (Y.L.); +86-23-6836-6018 (Z.W.)
| | - Guili Song
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Shaoxiong Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China
| | - Zhicheng Xu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Research Center of Fishery Resources and Environment, Southwest University, Chongqing 400715, China
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Qing Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Zhengli Wu
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), College of Fisheries, Research Center of Fishery Resources and Environment, Southwest University, Chongqing 400715, China
- Correspondence: (Y.L.); (Z.W.); Tel.: +86-27-6878-0100 (Y.L.); +86-23-6836-6018 (Z.W.)
| |
Collapse
|
13
|
Križaj D, Cordeiro S, Strauß O. Retinal TRP channels: Cell-type-specific regulators of retinal homeostasis and multimodal integration. Prog Retin Eye Res 2023; 92:101114. [PMID: 36163161 PMCID: PMC9897210 DOI: 10.1016/j.preteyeres.2022.101114] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 02/05/2023]
Abstract
Transient receptor potential (TRP) channels are a widely expressed family of 28 evolutionarily conserved cationic ion channels that operate as primary detectors of chemical and physical stimuli and secondary effectors of metabotropic and ionotropic receptors. In vertebrates, the channels are grouped into six related families: TRPC, TRPV, TRPM, TRPA, TRPML, and TRPP. As sensory transducers, TRP channels are ubiquitously expressed across the body and the CNS, mediating critical functions in mechanosensation, nociception, chemosensing, thermosensing, and phototransduction. This article surveys current knowledge about the expression and function of the TRP family in vertebrate retinas, which, while dedicated to transduction and transmission of visual information, are highly susceptible to non-visual stimuli. Every retinal cell expresses multiple TRP subunits, with recent evidence establishing their critical roles in paradigmatic aspects of vertebrate vision that include TRPM1-dependent transduction of ON bipolar signaling, TRPC6/7-mediated ganglion cell phototransduction, TRP/TRPL phototransduction in Drosophila and TRPV4-dependent osmoregulation, mechanotransduction, and regulation of inner and outer blood-retina barriers. TRP channels tune light-dependent and independent functions of retinal circuits by modulating the intracellular concentration of the 2nd messenger calcium, with emerging evidence implicating specific subunits in the pathogenesis of debilitating diseases such as glaucoma, ocular trauma, diabetic retinopathy, and ischemia. Elucidation of TRP channel involvement in retinal biology will yield rewards in terms of fundamental understanding of vertebrate vision and therapeutic targeting to treat diseases caused by channel dysfunction or over-activation.
Collapse
Affiliation(s)
- David Križaj
- Departments of Ophthalmology, Neurobiology, and Bioengineering, University of Utah, Salt Lake City, USA
| | - Soenke Cordeiro
- Institute of Physiology, Faculty of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Olaf Strauß
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, a Corporate Member of Freie Universität, Humboldt-University, The Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
14
|
Gao F, Ma J, Yu YQ, Gao XF, Bai Y, Sun Y, Liu J, Liu X, Barry DM, Wilhelm S, Piccinni-Ash T, Wang N, Liu D, Ross RA, Hao Y, Huang X, Jia JJ, Yang Q, Zheng H, van Nispen J, Chen J, Li H, Zhang J, Li YQ, Chen ZF. A non-canonical retina-ipRGCs-SCN-PVT visual pathway for mediating contagious itch behavior. Cell Rep 2022; 41:111444. [PMID: 36198265 PMCID: PMC9595067 DOI: 10.1016/j.celrep.2022.111444] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 08/10/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022] Open
Abstract
Contagious itch behavior informs conspecifics of adverse environment and is crucial for the survival of social animals. Gastrin-releasing peptide (GRP) and its receptor (GRPR) in the suprachiasmatic nucleus (SCN) of the hypothalamus mediates contagious itch behavior in mice. Here, we show that intrinsically photosensitive retina ganglion cells (ipRGCs) convey visual itch information, independently of melanopsin, from the retina to GRP neurons via PACAP-PAC1R signaling. Moreover, GRPR neurons relay itch information to the paraventricular nucleus of the thalamus (PVT). Surprisingly, neither the visual cortex nor superior colliculus is involved in contagious itch. In vivo calcium imaging and extracellular recordings reveal contagious itch-specific neural dynamics of GRPR neurons. Thus, we propose that the retina-ipRGC-SCN-PVT pathway constitutes a previously unknown visual pathway that probably evolved for motion vision that encodes salient environmental cues and enables animals to imitate behaviors of conspecifics as an anticipatory mechanism to cope with adverse conditions. It has been shown that GRP-GRPR neuropeptide signaling in the SCN is important for contagious itch behavior in mice. Gao et al. find that SCN-projecting ipRGCs are sufficient to relay itch information from the retina to the SCN by releasing neuropeptide PACAP to activate the GRP-GRPR pathway.
Collapse
Affiliation(s)
- Fang Gao
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jun Ma
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yao-Qing Yu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, P. R. China
| | - Xiao-Fei Gao
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Present address: Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai 200434, P. R. China
| | - Yang Bai
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Department of Anatomy, Histology and Embryology & K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, P. R. China,Present address: Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang 110016, P. R. China
| | - Yi Sun
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Department of Anatomy, Histology and Embryology & K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, P. R. China,Present address: Binzhou Medical University, Yantai 264003, P. R. China
| | - Juan Liu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xianyu Liu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Devin M. Barry
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven Wilhelm
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tyler Piccinni-Ash
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Na Wang
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Present address: Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, P. R. China
| | - Dongyang Liu
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Department of Pain Management, the State Key Clinical Specialty in Pain Medicine, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, P.R. China
| | - Rachel A. Ross
- Department of Neuroscience, Psychiatry and Medicine, Albert Einstein College of Medicine Rose F. Kennedy Center, Bronx, NY, USA
| | - Yan Hao
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Present address: Department of Pediatrics, Tongji Hospital, Tongji Medical College, HuaZhong University of Science and Technology, Wuhan 430030, P. R. China
| | - Xu Huang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institute for Medical and Engineering Innovation, Eye & ENT Hospital, Fudan University, Shanghai 200031, P.R. China
| | - Jin-Jing Jia
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Present address: College of Life Sciences, Xinyang Normal University, Xinyang 464000, P. R. China
| | - Qianyi Yang
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hao Zheng
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institute for Medical and Engineering Innovation, Eye & ENT Hospital, Fudan University, Shanghai 200031, P.R. China
| | - Johan van Nispen
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Present address: Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Jun Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, P. R. China
| | - Hui Li
- Department of Anatomy, Histology and Embryology & K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, P. R. China
| | - Jiayi Zhang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science and Institute for Medical and Engineering Innovation, Eye & ENT Hospital, Fudan University, Shanghai 200031, P.R. China
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology & K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, P. R. China
| | - Zhou-Feng Chen
- Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA,Departments of Anesthesiology, Medicine, Psychiatry and Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA,Lead contact,Correspondence:
| |
Collapse
|
15
|
Gilhooley MJ, Lindner M, Palumaa T, Hughes S, Peirson SN, Hankins MW. A systematic comparison of optogenetic approaches to visual restoration. Mol Ther Methods Clin Dev 2022; 25:111-123. [PMID: 35402632 PMCID: PMC8956963 DOI: 10.1016/j.omtm.2022.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/04/2022] [Indexed: 02/06/2023]
Abstract
During inherited retinal degenerations (IRDs), vision is lost due to photoreceptor cell death; however, a range of optogenetic tools have been shown to restore light responses in animal models. Restored response characteristics vary between tools and the neuronal cell population to which they are delivered: the interplay between these is complex, but targeting upstream neurons (such as retinal bipolar cells) may provide functional benefit by retaining intraretinal signal processing. In this study, our aim was to compare two optogenetic tools: mammalian melanopsin (hOPN4) and microbial red-shifted channelrhodopsin (ReaChR) expressed within two subpopulations of surviving cells in a degenerate retina. Intravitreal adeno-associated viral vectors and mouse models utilising the Cre/lox system restricted expression to populations dominated by bipolar cells or retinal ganglion cells and was compared with non-targeted delivery using the chicken beta actin (CBA) promoter. In summary, we found bipolar-targeted optogenetic tools produced faster kinetics and flatter intensity-response relationships compared with non-targeted or retinal-ganglion-cell-targeted hOPN4. Hence, optogenetic tools of both mammalian and microbial origins show advantages when targeted to bipolar cells. This demonstrates the advantage of bipolar-cell-targeted optogenetics for vision restoration in IRDs. We therefore developed a bipolar-cell-specific gene delivery system employing a compressed promoter with the potential for clinical translation.
Collapse
Affiliation(s)
- Michael J. Gilhooley
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- The Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
- Moorfields Eye Hospital, 162, City Road, London EC1V 2PD, UK
| | - Moritz Lindner
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Institute of Physiology and Pathophysiology, Department of Neurophysiology, Philipps University, Deutschhausstrasse 1-2, Marburg 35037, Germany
| | - Teele Palumaa
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- East Tallinn Central Hospital Eye Clinic, Ravi 18, 10138 Tallinn, Estonia
| | - Steven Hughes
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
| | - Stuart N. Peirson
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
| | - Mark W. Hankins
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Jules Thorne SCNi, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK
- Corresponding author Mark W. Hankins, Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX1 3QU, UK.
| |
Collapse
|
16
|
Usategui-Martín R, Puertas-Neyra K, Galindo-Cabello N, Hernández-Rodríguez LA, González-Pérez F, Rodríguez-Cabello JC, González-Sarmiento R, Pastor JC, Fernandez-Bueno I. Retinal Neuroprotective Effect of Mesenchymal Stem Cells Secretome Through Modulation of Oxidative Stress, Autophagy, and Programmed Cell Death. Invest Ophthalmol Vis Sci 2022; 63:27. [PMID: 35486068 PMCID: PMC9055551 DOI: 10.1167/iovs.63.4.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Purpose Degenerative mechanisms of retinal neurodegenerative diseases (RND) share common cellular and molecular signalization pathways. Curative treatment does not exist and cell-based therapy, through the paracrine properties of mesenchymal stem cells (MSC), is a potential unspecific treatment for RND. This study aimed to evaluate the neuroprotective capability of human bone marrow (bm) MSC secretome and its potential to modulate retinal responses to neurodegeneration. Methods An in vitro model of spontaneous retinal neurodegeneration was used to compare three days of monocultured neuroretina (NR), NR cocultured with bmMSC, and NR cultured with bmMSC secretome. We evaluated retinal morphology markers (Lectin peanut agglutinin, rhodopsin, protein kinase C α isoform, neuronal-specific nuclear protein, glial fibrillary acidic protein, TdT-mediated dUTP nick-end labeling, and vimentin) and proteins involved in apoptosis (apoptosis-inductor factor, caspase-3), necroptosis (MLKL), and autophagy (p62). Besides, we analyzed the relative mRNA expression through qPCR of genes involved in apoptosis (BAX, BCL2, CASP3, CASP8, CASP9), necroptosis (MLKL, RIPK1, RIPK3), autophagy (ATG7, BCLIN1, LC3B, mTOR, SQSTM1), oxidative stress (COX2, CYBA, CYBB, GPX6, SOD1, TXN2, TXNRD1) and inflammation (IL1, IL6, IL10, TGFb1, TNFa). Results The bmMSC secretome preserves retinal morphology, limits pro-apoptotic- and pro-necroptotic-related gene and protein expression, modulates autophagy-related genes and proteins, and stimulates the activation of antioxidant-associated genes. Conclusions The neuroprotective ability of the bmMSC secretome is associated with activation of antioxidant machinery, modulation of autophagy, and inhibition of apoptosis and necroptosis during retinal degeneration. The neuroprotective effect of bmMSC secretomes in the presence/absence of MSC looks similar. Our current results reinforce the hypothesis that the human bmMSC secretome slows retinal neurodegeneration and may be a therapeutic option for treating RND.
Collapse
Affiliation(s)
- Ricardo Usategui-Martín
- Instituto Universitario de Oftalmobiología Aplicada, Retina Group, Universidad de Valladolid, Valladolid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain.,Red Temática de Investigación Cooperativa en Salud, Oftared, Instituto de Salud Carlos III, Valladolid, Spain.,RetiBrain (RED2018-102499-T), Ministerio de Ciencia, Innovación y Universidades, Valladolid, Spain
| | - Kevin Puertas-Neyra
- Instituto Universitario de Oftalmobiología Aplicada, Retina Group, Universidad de Valladolid, Valladolid, Spain
| | - Nadia Galindo-Cabello
- Instituto Universitario de Oftalmobiología Aplicada, Retina Group, Universidad de Valladolid, Valladolid, Spain.,Postgraduate Unit, Faculty of Biological Sciences, National University of San Marcos, Lima, Peru
| | | | - Fernando González-Pérez
- Group for Advanced Materials and Nanobiotechnology (GIR BIOFORGE), CIBER-BBN, Edificio LUCIA, Universidad de Valladolid, Paseo Belén 19, Valladolid, Spain
| | - José Carlos Rodríguez-Cabello
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain.,Group for Advanced Materials and Nanobiotechnology (GIR BIOFORGE), CIBER-BBN, Edificio LUCIA, Universidad de Valladolid, Paseo Belén 19, Valladolid, Spain
| | - Rogelio González-Sarmiento
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, Salamanca, Spain.,Institute of Molecular and Cellular Biology of Cancer, University of Salamanca-CSIC, Salamanca, Spain
| | - José Carlos Pastor
- Instituto Universitario de Oftalmobiología Aplicada, Retina Group, Universidad de Valladolid, Valladolid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain.,Red Temática de Investigación Cooperativa en Salud, Oftared, Instituto de Salud Carlos III, Valladolid, Spain.,RetiBrain (RED2018-102499-T), Ministerio de Ciencia, Innovación y Universidades, Valladolid, Spain
| | - Ivan Fernandez-Bueno
- Instituto Universitario de Oftalmobiología Aplicada, Retina Group, Universidad de Valladolid, Valladolid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Valladolid, Spain.,Red Temática de Investigación Cooperativa en Salud, Oftared, Instituto de Salud Carlos III, Valladolid, Spain.,RetiBrain (RED2018-102499-T), Ministerio de Ciencia, Innovación y Universidades, Valladolid, Spain
| |
Collapse
|
17
|
Excitatory Amino Acid Transporter EAAT5 Improves Temporal Resolution in the Retina. eNeuro 2021; 8:ENEURO.0406-21.2021. [PMID: 34772693 PMCID: PMC8670604 DOI: 10.1523/eneuro.0406-21.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/16/2021] [Indexed: 11/21/2022] Open
Abstract
Excitatory amino acid transporters (EAATs) remove glutamate from the synaptic cleft. In the retina, EAAT1 and EAAT2 are considered the major glutamate transporters. However, it has not yet been possible to determine how EAAT5 shapes the retinal light responses because of the lack of a selective EAAT5 blocker or EAAT5 knock-out (KO) animal model. In this study, EAAT5 was found to be expressed in a punctate manner close to release sites of glutamatergic synapses in the mouse retina. Light responses from retinae of wild-type (WT) and of a newly generated model with a targeted deletion of EAAT5 (EAAT5-/-) were recorded in vitro using multielectrode arrays (MEAs). Flicker resolution was considerably lower in EAAT5-/- retinae than in WT retinae. The close proximity to the glutamate release site makes EAAT5 an ideal tool to improve temporal information processing in the retina by controlling information transfer at glutamatergic synapses.
Collapse
|
18
|
Keilhoff G, Titze M, Ebmeyer U. Immuno-histological detection of resistant columnar units and vulnerable networks in the rat retina after asphyxia-induced transient cardiac arrest. Restor Neurol Neurosci 2021; 39:267-289. [PMID: 34334436 DOI: 10.3233/rnn-211174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Stroke-related loss of vision is one of the residual impairments, restricting the quality of life. However, studies of the ocular manifestations of asphyxia cardiac arrest/resuscitation (ACA/R) have reported very heterogeneous results. OBJECTIVE We aimed to evaluate the ACA/R-induced degeneration pattern of the different retinal cell populations in rats using different immuno-histological stainings. METHODS The staining pattern of toluidine blue and the ganglion cell markers β-III-tubulin and NeuN; the calcium-binding protein parvalbumin, indicating ganglion, amacrine, and horizontal cells; calretinin D28k, indicating ganglion and amacrine cells; calbindin, indicating horizontal cells; Chx 10, indicating cone bipolar cells; PKCα, indicating ON-type rod bipolar cells; arrestin, indicating cones; and rhodopsin, a marker of rods, as well as the glial cell markers GFAP (indicating astroglia and Müller cells) and IBA1 (indicating microglia), were evaluated after survival times of 7 and 21 days in an ACA/R rat model. Moreover, quantitative morphological analysis of the optic nerve was performed. The ACA/R specimens were compared with those from sham-operated and completely naïve rats. RESULTS ACA/R-induced effects were: (i) a significant reduction of retinal thickness after long-term survival; (ii) ganglion cell degeneration, including their fiber network in the inner plexiform layer; (iii) degeneration of amacrine and cone bipolar cells; (iv) degeneration of cone photoreceptors; (v) enhanced resistance to ACA/R by rod photoreceptors, ON-type rod bipolar and horizontal cells, possibly caused by the strong upregulation of the calcium-binding proteins calretinin, parvalbumin, and calbindin, counteracting the detrimental calcium overload; (vi) significant activation of Müller cells as further element of retinal anti-stress self-defense mechanisms; and (vii) morphological alterations of the optic nerve in form of deformed fibers. CONCLUSIONS Regardless of the many defects, the surviving neuronal structures seemed to be able to maintain retinal functionality, which can be additionally improved by regenerative processes true to the "use it or lose it" dogma.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Medical Faculty, University of Magdeburg, Magdeburg, Germany
| | - Maximilian Titze
- Institute of Biochemistry and Cell Biology, Medical Faculty, University of Magdeburg, Magdeburg, Germany
| | - Uwe Ebmeyer
- Department of Anesthesiology, Medical Faculty, University of Magdeburg, Magdeburg, Germany
| |
Collapse
|
19
|
Xu S, Zhang P, Zhang M, Wang X, Li G, Xu G, Ni Y. Synaptic changes and the response of microglia in a light-induced photoreceptor degeneration model. Mol Vis 2021; 27:206-220. [PMID: 33967574 PMCID: PMC8100860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 04/29/2021] [Indexed: 11/15/2022] Open
Abstract
Purpose To explore synaptic changes and the response of microglia in a light-induced photoreceptor degeneration model. Methods Sprague-Dawley rats were euthanized 1 h, 1 day, 3 days, 7 days, and 14 days after being exposed to intense blue light for 24 h. Hematoxylin and eosin (H&E) and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining were used to evaluate changes in the outer nuclear layer (ONL). Transmission electron microscopy (TEM) was applied to observe the ultrastructural changes in the synapses between the photoreceptors and second-order neurons. Western blotting was conducted to evaluate specific proteins, including postsynaptic density-95 (PSD-95), metabotropic glutamate receptor 6 (mGluR6), synapsin I, and synaptophysin. Immunofluorescence of CD11b and PKC-α or mGluR6 was used to explore the spatial relationships between microglial processes and synaptic elements. Immunoelectron microscopy of PSD-95 was performed to further confirm its engulfment of synaptic materials. Results H&E and TUNEL staining showed that the thickness of the ONL decreased markedly, and the number of apoptotic photoreceptors peaked at day 1. TEM revealed darkened photoreceptor terminals and that ribbons of them were floating in the cytoplasm, coinciding with the downregulation of PSD-95 and mGluR6. Downstream synaptic protein synapsin I and synaptophysin exhibited upregulation in the inner plexiform layer. Activated microglia migrated to the outer retina, and their processes were found in close proximity to synapses in the outer plexiform layer under light and electron microscopy levels. Double immunostaining of CD11b and mGluR6 showed colocalization. PSD-95-immunoreactive electron-dense materials were observed inside the microglia suggesting engulfment of synaptic components. Conclusions The study showed that there are early synaptic impairment and late compensatory changes in downstream synapses in this photic injury model. Activated microglia touched and directly engulfed synaptic materials. Microglia may play a role or a partial role in synaptic changes.
Collapse
Affiliation(s)
- Sisi Xu
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China,Key NHC Key Laboratory of Myopia (Fudan University); Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Peijun Zhang
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China,Key NHC Key Laboratory of Myopia (Fudan University); Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Meng Zhang
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China,Key NHC Key Laboratory of Myopia (Fudan University); Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xin Wang
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China,Key NHC Key Laboratory of Myopia (Fudan University); Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Gang Li
- Research Center, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Gezhi Xu
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China,Key NHC Key Laboratory of Myopia (Fudan University); Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yingqin Ni
- Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China,Key NHC Key Laboratory of Myopia (Fudan University); Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
20
|
Li L, Zhao H, Xie H, Akhtar T, Yao Y, Cai Y, Dong K, Gu Y, Bao J, Chen J, Zhang M, Zhong K, Xu W, Xue T. Electrophysiological characterization of photoreceptor-like cells in human inducible pluripotent stem cell-derived retinal organoids during in vitro maturation. STEM CELLS (DAYTON, OHIO) 2021; 39:959-974. [PMID: 33662144 DOI: 10.1002/stem.3363] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/10/2021] [Indexed: 11/10/2022]
Abstract
Retinal organoids (ROs) derived from human inducible pluripotent stem cells (hiPSCs) exhibit considerable therapeutic potential. However, current quality control of ROs during in vitro differentiation is largely limited to the detection of molecular markers, often by immunostaining, polymerase chain reaction (PCR) assays and sequencing, often without proper functional assessments. As such, in the current study, we systemically characterized the physiological maturation of photoreceptor-like cells in hiPSC-derived ROs. By performing patch-clamp recordings from photoreceptor-like cells in ROs at distinct differentiation stages (ie, Differentiation Day [D]90, D150, and D200), we determined the electrophysiological properties of the plasma membrane and several characteristic ion channels closely associated with the physiological functions of the photoreceptors. Ionic hallmarks, such as hyperpolarization-activated cyclic nucleotide-gated (HCN) channels and cyclic nucleotide-gated (CNG) channels, matured progressively during differentiation. After D200 in culture, these characteristic currents closely resembled those in macaque or human native photoreceptors. Furthermore, we demonstrated that the hyperpolarization-activated inward current/depolarization-activated outward current ratio (I-120 /I+40 ), termed as the inward-outward current (IOC) ratio hereon, accurately represented the maturity of photoreceptors and could serve as a sensitive indicator of pathological state. Thus, this study provides a comprehensive dataset describing the electrophysiological maturation of photoreceptor-like cells in hiPSC-derived ROs for precise and sensitive quality control during RO differentiation.
Collapse
Affiliation(s)
- Lingyun Li
- Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China.,CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China
| | - Huan Zhao
- School of Biology, Food, and Environment, Hefei University, Hefei, People's Republic of China
| | - Haohuan Xie
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China
| | - Tasneem Akhtar
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China
| | - Yichuan Yao
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China
| | - Yuan Cai
- Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Kai Dong
- Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Yonghao Gu
- Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Jin Bao
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, People's Republic of China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Jutao Chen
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, People's Republic of China
| | - Mei Zhang
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, People's Republic of China
| | - Kai Zhong
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, People's Republic of China.,Key Laboratory of Anhui Province for High Field Magnetic Resonance Imaging, Hefei, People's Republic of China
| | - Weiping Xu
- Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, People's Republic of China.,The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Tian Xue
- Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China.,CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei, People's Republic of China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, People's Republic of China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, People's Republic of China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
21
|
Usategui-Martín R, Puertas-Neyra K, García-Gutiérrez MT, Fuentes M, Pastor JC, Fernandez-Bueno I. Human Mesenchymal Stem Cell Secretome Exhibits a Neuroprotective Effect over In Vitro Retinal Photoreceptor Degeneration. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:1155-1166. [PMID: 32514411 PMCID: PMC7267685 DOI: 10.1016/j.omtm.2020.05.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022]
Abstract
Retinal photoreceptor degeneration occurs frequently in several neurodegenerative retinal diseases such as age-related macular degeneration, retinitis pigmentosa, or genetic retinal diseases related to the photoreceptors. Despite the impact on daily life and the social and economic consequences, there is no cure for these diseases. Considering this, cell-based therapy may be an optimal therapeutic option. This study evaluated the neuroprotective in vitro potential of a secretome of human bone marrow mesenchymal stem cells (MSCs) for retinal photoreceptors in vitro. We analyzed the photoreceptor morphologic changes and the paracrine factors secreted by human bone marrow MSCs in a physically separated co-culture with degenerated neuroretinas, using organotypic neuroretinal cultures. The results showed that the secretome of human bone marrow MSCs had a neuroprotective effect over the neuroretinal general organization and neuropreserved the photoreceptors from degeneration probably by secretion of neuroprotective proteins. The study of the expression of 1,000 proteins showed increased paracrine factors secreted by MSCs that could be crucial in the neuroprotective effect of the stem cell secretome over in vitro retinal degeneration. The current results reinforce the hypothesis that the paracrine effect of the human bone marrow MSCs may slow photoreceptor neurodegeneration and be a therapeutic option in retinal photoreceptor degenerative diseases.
Collapse
Affiliation(s)
- Ricardo Usategui-Martín
- Instituto Universitario de Oftalmobiología Aplicada (IOBA), Retina Group, Universidad de Valladolid, 47011 Valladolid, Spain
| | - Kevin Puertas-Neyra
- Instituto Universitario de Oftalmobiología Aplicada (IOBA), Retina Group, Universidad de Valladolid, 47011 Valladolid, Spain
| | - María-Teresa García-Gutiérrez
- Instituto Universitario de Oftalmobiología Aplicada (IOBA), Retina Group, Universidad de Valladolid, 47011 Valladolid, Spain
| | - Manuel Fuentes
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC), University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain.,Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC), University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José Carlos Pastor
- Instituto Universitario de Oftalmobiología Aplicada (IOBA), Retina Group, Universidad de Valladolid, 47011 Valladolid, Spain.,Department of Ophthalmology, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, 47011 Valladolid, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Oftared, Instituto de Salud Carlos III, 47011 Valladolid, Spain
| | - Ivan Fernandez-Bueno
- Instituto Universitario de Oftalmobiología Aplicada (IOBA), Retina Group, Universidad de Valladolid, 47011 Valladolid, Spain.,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, 47011 Valladolid, Spain.,Red Temática de Investigación Cooperativa en Salud (RETICS), Oftared, Instituto de Salud Carlos III, 47011 Valladolid, Spain
| |
Collapse
|
22
|
Wakeham CM, Ren G, Morgans CW. Expression and distribution of trophoblast glycoprotein in the mouse retina. J Comp Neurol 2020; 528:1660-1671. [PMID: 31891182 DOI: 10.1002/cne.24850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 02/03/2023]
Abstract
We recently identified the leucine-rich repeat (LRR) adhesion protein, trophoblast glycoprotein (TPBG), as a novel PKCα-dependent phosphoprotein in retinal rod bipolar cells (RBCs). Since TPBG has not been thoroughly examined in the retina, this study characterizes the localization and expression patterns of TPBG in the developing and adult mouse retina using two antibodies, one against the N-terminal LRR domain and the other against the C-terminal PDZ-interacting motif. Both antibodies labeled RBC dendrites in the outer plexiform layer and axon terminals in the IPL, as well as a putative amacrine cell with their cell bodies in the inner nuclear layer (INL) and a dense layer in the middle of the inner plexiform layer (IPL). In live transfected HEK293 cells, TPBG was localized to the plasma membrane with the N-terminal LRR domain facing the extracellular space. TPBG immunofluorescence in RBCs was strongly altered by the loss of TRPM1 in the adult retina, with significantly less dendritic and axon terminal labeling in TRPM1 knockout compared to wild type, despite no change in total TPBG detected by immunoblotting. During retinal development, TPBG expression increases dramatically just prior to eye opening with a time course closely correlated with that of TRPM1 expression. In the retina, LRR proteins have been implicated in the development and maintenance of functional bipolar cell synapses, and TPBG may play a similar role in RBCs.
Collapse
Affiliation(s)
- Colin M Wakeham
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Gaoying Ren
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| | - Catherine W Morgans
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
23
|
Shen Y, Luo X, Liu S, Shen Y, Nawy S, Shen Y. Rod bipolar cells dysfunction occurs before ganglion cells loss in excitotoxin-damaged mouse retina. Cell Death Dis 2019; 10:905. [PMID: 31787761 PMCID: PMC6885518 DOI: 10.1038/s41419-019-2140-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/29/2019] [Accepted: 11/07/2019] [Indexed: 11/16/2022]
Abstract
Progressive degeneration of retinal ganglion cells (RGCs) will cause a blinding disease. Most of the study is focusing on the RGCs itself. In this study, we demonstrate a decline of the presynaptic rod bipolar cells (RBCs) response precedes RGCs loss and a decrease of protein kinase Cα (PKCα) protein expression in RBCs dendrites, using whole-cell voltage-clamp, electroretinography (ERG) measurements, immunostaining and co-immunoprecipitation. We present evidence showing that N-methyl D-aspartate receptor subtype 2B (NR2B)/protein interacting with C kinase 1 (PICK1)-dependent degradation of PKCα protein in RBCs contributes to RBCs functional loss. Mechanistically, NR2B forms a complex with PKCα and PICK1 to promote the degradation of PKCα in a phosphorylation- and proteasome-dependent manner. Similar deficits in PKCα expression and response sensitivity were observed in acute ocular hypertension and optic never crush models. In conclusion, we find that three separate experimental models of neurodegeneration, often used to specifically target RGCs, disrupt RBCs function prior to the loss of RGCs. Our findings provide useful information for developing new diagnostic tools and treatments for retinal ganglion cells degeneration disease.
Collapse
Affiliation(s)
- Yumeng Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Xue Luo
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Shiliang Liu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei Province, China
| | - Ying Shen
- Medical School, Zhejiang University, Hangzhou, 310053, Zhejiang Province, China
| | - Scott Nawy
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720, CA, USA
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, Hubei Province, China.
| |
Collapse
|
24
|
Murali A, Ramlogan-Steel CA, Steel JC, Layton CJ. Characterisation and validation of the 8-fold quadrant dissected human retinal explant culture model for pre-clinical toxicology investigation. Toxicol In Vitro 2019; 63:104716. [PMID: 31706033 DOI: 10.1016/j.tiv.2019.104716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/13/2019] [Accepted: 11/04/2019] [Indexed: 11/30/2022]
Abstract
One of the major challenges in studying ocular toxicology is a lack of clinically-relevant retinal experimental models. In this study we describe the use of an in vitro human retinal explant strategy to generate a reproducible experimental model with utility in neuro-toxicity retinal studies. A retinal dissection strategy, referred to as the 8 fold quadrant dissection, was developed by dissecting human donor retinas into 4 fragments through the fovea in order to obtain 8 experimentally reproducible retinal explants from a single donor. This quadrant dissection gave rise to equivalent proportions of CD73+ photoreceptors and CD90+ ganglion cells in 8 fragments from a single donor and this remained stable for up to 3 days in culture. Major retinal cell types continued to be observed after 8 weeks in culture, despite breakdown of the retinal layers, suggesting the potential to use this model in long-term studies where observation of individual cell types is possible. The utility of this system was examined in a proof of principle neuro-toxicology study. We showed reproducible induction of toxicity in photoreceptors and retinal ganglion cells by glutamate, cobalt chloride and hydrogen peroxide insults, and observed the therapeutic positive effects of the administration of memantine, formononetin and trolox. The quadrant dissected human retinal explants have the potential to be used in toxicology studies in human ocular diseases.
Collapse
Affiliation(s)
- Aparna Murali
- LVF Ophthalmology Research Centre, Translational Research Institute, Woolloongabba, QLD, Australia; Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Australia
| | - Charmaine A Ramlogan-Steel
- LVF Ophthalmology Research Centre, Translational Research Institute, Woolloongabba, QLD, Australia; Central Queensland University, School of Health, Medical and Applied Science, Rockhampton, QLD, Australia
| | - Jason C Steel
- Central Queensland University, School of Health, Medical and Applied Science, Rockhampton, QLD, Australia.
| | - Christopher J Layton
- LVF Ophthalmology Research Centre, Translational Research Institute, Woolloongabba, QLD, Australia; Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Australia.
| |
Collapse
|
25
|
Wakeham CM, Wilmarth PA, Cunliffe JM, Klimek JE, Ren G, David LL, Morgans CW. Identification of PKCα-dependent phosphoproteins in mouse retina. J Proteomics 2019; 206:103423. [PMID: 31255707 DOI: 10.1016/j.jprot.2019.103423] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/11/2019] [Accepted: 06/19/2019] [Indexed: 12/11/2022]
Abstract
Adjusting to a wide range of light intensities is an essential feature of retinal rod bipolar cell (RBC) function. While persuasive evidence suggests this modulation involves phosphorylation by protein kinase C-alpha (PKCα), the targets of PKCα phosphorylation in the retina have not been identified. PKCα activity and phosphorylation in RBCs was examined by immunofluorescence confocal microscopy using a conformation-specific PKCα antibody and antibodies to phosphorylated PKC motifs. PKCα activity was dependent on light and expression of TRPM1, and RBC dendrites were the primary sites of light-dependent phosphorylation. PKCα-dependent retinal phosphoproteins were identified using a phosphoproteomics approach to compare total protein and phosphopeptide abundance between phorbol ester-treated wild type and PKCα knockout (PKCα-KO) mouse retinas. Phosphopeptide mass spectrometry identified over 1100 phosphopeptides in mouse retina, with 12 displaying significantly greater phosphorylation in WT compared to PKCα-KO samples. The differentially phosphorylated proteins fall into the following functional groups: cytoskeleton/trafficking (4 proteins), ECM/adhesion (2 proteins), signaling (2 proteins), transcriptional regulation (3 proteins), and homeostasis/metabolism (1 protein). Two strongly differentially expressed phosphoproteins, BORG4 and TPBG, were localized to the synaptic layers of the retina, and may play a role in PKCα-dependent modulation of RBC physiology. Data are available via ProteomeXchange with identifier PXD012906. SIGNIFICANCE: Retinal rod bipolar cells (RBCs), the second-order neurons of the mammalian rod visual pathway, are able to modulate their sensitivity to remain functional across a wide range of light intensities, from starlight to daylight. Evidence suggests that this modulation requires the serine/threonine kinase, PKCα, though the specific mechanism by which PKCα modulates RBC physiology is unknown. This study examined PKCα phosophorylation patterns in mouse rod bipolar cells and then used a phosphoproteomics approach to identify PKCα-dependent phosphoproteins in the mouse retina. A small number of retinal proteins showed significant PKCα-dependent phosphorylation, including BORG4 and TPBG, suggesting a potential contribution to PKCα-dependent modulation of RBC physiology.
Collapse
Affiliation(s)
- Colin M Wakeham
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Phillip A Wilmarth
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jennifer M Cunliffe
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA
| | - John E Klimek
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA
| | - Gaoying Ren
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Larry L David
- Proteomics Shared Resource, Oregon Health and Science University, Portland, OR 97239, USA; Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Catherine W Morgans
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
26
|
Hellmer CB, Clemons MR, Nawy S, Ichinose T. A group I metabotropic glutamate receptor controls synaptic gain between rods and rod bipolar cells in the mouse retina. Physiol Rep 2018; 6:e13885. [PMID: 30338673 PMCID: PMC6194217 DOI: 10.14814/phy2.13885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 09/10/2018] [Accepted: 09/16/2018] [Indexed: 11/24/2022] Open
Abstract
The canonical mGluR6-Trpm1 pathway that generates the sign-inverting signal between photoreceptors and ON bipolar cells has been well described. However, one type of ON bipolar cell, the rod bipolar cell (RBC), additionally is thought to express the group I mGluRs whose function is unknown. We examined the role of group I mGluRs in mouse RBCs and here provide evidence that it controls synaptic gain between rods and RBCs. In dark-adapted conditions, the mGluR1 antagonists LY367385 and (RS)-1-Aminoindan-1,5-dicarboxylic acid, but not the mGluR5 antagonist 2-Methyl-6-(phenylethynyl)pyridine hydrochloride reduced the light-evoked responses in RBCs indicating that mGluR1, but not mGluR5, serves to potentiate RBC responses. Perturbing the downstream phospholipase C (PLC)-protein kinase C (PKC) pathway by inhibiting PLC, tightly buffering intracellular Ca2+ , or preventing its release from intracellular stores reduced the synaptic potentiation by mGluR1. The effect of mGluR1 activation was dependent upon adaptation state, strongly increasing the synaptic gain in dark-, but not in light-adapted retinas, or in the presence of a moderate background light, consistent with the idea that mGluR1 activation requires light-dependent glutamate release from rods. Moreover, immunostaining revealed that protein kinase Cα (PKCα) is more strongly expressed in RBC dendrites in dark-adapted conditions, revealing an additional mechanism behind the loss of mGluR1 potentiation. In light-adapted conditions, exogenous activation of mGluR1 with the agonist 3,5-Dihydroxyphenylglycine increased the mGluR6 currents in some RBCs and decreased it in others, suggesting an additional action of mGluR1 that is unmasked in the light-adapted state. Elevating intracellular free Ca2+ , consistently resulted in a decrease in synaptic gain. Our results provide evidence that mGluR1 controls the synaptic gain in RBCs.
Collapse
Affiliation(s)
- Chase B. Hellmer
- Department of Ophthalmology, Visual and Anatomical SciencesWayne State University School of MedicineDetroitMichigan48201
| | - Melissa Rampino Clemons
- Dominic P Purpura Dept. of NeuroscienceAlbert Einstein College of Medicine BronxBronxNew York10461
| | - Scott Nawy
- Dominic P Purpura Dept. of NeuroscienceAlbert Einstein College of Medicine BronxBronxNew York10461
- Department of Ophthalmology and Visual SciencesUniversity of Nebraska Medical CenterOmahaNebraska68198
| | - Tomomi Ichinose
- Department of Ophthalmology, Visual and Anatomical SciencesWayne State University School of MedicineDetroitMichigan48201
| |
Collapse
|
27
|
Woods SM, Mountjoy E, Muir D, Ross SE, Atan D. A comparative analysis of rod bipolar cell transcriptomes identifies novel genes implicated in night vision. Sci Rep 2018; 8:5506. [PMID: 29615777 PMCID: PMC5883057 DOI: 10.1038/s41598-018-23901-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/20/2018] [Indexed: 11/09/2022] Open
Abstract
In the mammalian retina, rods and a specialised rod-driven signalling pathway mediate visual responses under scotopic (dim light) conditions. As rods primarily signal to rod bipolar cells (RBCs) under scoptic conditions, disorders that affect rod or RBC function are often associated with impaired night vision. To identify novel genes expressed by RBCs and, therefore, likely to be involved in night vision, we took advantage of the adult Bhlhe23−/− mouse retina (that lacks RBCs) to derive the RBC transcriptome. We found that genes expressed by adult RBCs are mainly involved in synaptic structure and signalling, whereas genes that influence RBC development are also involved in the cell cycle and transcription/translation. By comparing our data with other published retinal and bipolar cell transcriptomes (where we identify RBCs by the presence of Prkca and/or Pcp2 transcripts), we have derived a consensus for the adult RBC transcriptome. These findings ought to facilitate further research into physiological mechanisms underlying mammalian night vision as well as proposing candidate genes for patients with inherited causes of night blindness.
Collapse
Affiliation(s)
- Sasha M Woods
- Bristol Medical School, University of Bristol, Bristol, BS8 1TD, UK.
| | - Edward Mountjoy
- Bristol Medical School, University of Bristol, Bristol, BS8 1TD, UK.,MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
| | - Duncan Muir
- Bristol Medical School, University of Bristol, Bristol, BS8 1TD, UK
| | - Sarah E Ross
- Departments of Neurobiology and Anesthesiology and the Center for Pain Research, University of Pittsburgh, Pittsburgh, 15213-2536, USA
| | - Denize Atan
- Bristol Medical School, University of Bristol, Bristol, BS8 1TD, UK
| |
Collapse
|
28
|
Oh TW, Do HJ, Kim KY, Park KI, Ma JY. Leaves of Acer palmatum thumb. Rescues N-ethyl-N-nitrosourea (ENU)-Induced retinal degeneration in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 42:51-55. [PMID: 29655697 DOI: 10.1016/j.phymed.2018.03.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/13/2018] [Accepted: 03/12/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND In the East Asia, the genus Acer (Aceraceae) is a herbal medicine that is used to treat various diseases, including hemostasis, hepatic disorders, traumatic bleeding and poor eyesight. However, the effects of Acer palmatum thumb. on retinal degeneration are unknown. AIM In this study, we investigated whether Acer palmatum thumb.ethanol extract (KIOM-2015E) can protect eyes from retinal degeneration. Our research investigated whether KIOM-2015E could have a protective effect in the retinal degenerating mouse model induced by N-ethyl-N-nitrosourea (ENU). MATERIALS AND METHODS Retinal degeneration was induced by a single intraperitoneal injection of ENU in ICR mice. KIOM-2015E (100, 200 mg/kg) was orally administered once per day. The eyeballs were embedded and lysed after drug administration to examine the histological changed and protein expression levels. RESULTS The ENU-induced retinal degeneration model exhibited increased photoreceptor cell death and a loss of the outer nuclear layer. Additionally, the expression of PKCα and OPN1SW was reduced, and that of GFAP and Nestin was increased in ENU-treated retinal tissues. CONCLUSION KIOM-2015E treatment ameliorated the ENU-induced retinal degeneration. KIOM-2015E prevents ENU-induced retinal degeneration by modulating protein expression and the thickness of the outer nuclear layer in the retina.
Collapse
Affiliation(s)
- Tae Woo Oh
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), 70 Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea.
| | - Hyun Ju Do
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), 70 Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Kwang-Youn Kim
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), 70 Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Kwang Il Park
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), 70 Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea.
| | - Jin Yeul Ma
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), 70 Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea.
| |
Collapse
|
29
|
Haug MF, Gesemann M, Berger M, Neuhauss SCF. Phylogeny and distribution of protein kinase C variants in the zebrafish. J Comp Neurol 2018; 526:1097-1109. [DOI: 10.1002/cne.24395] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Marion F. Haug
- University of Zurich, Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology; Zurich CH-8057 Switzerland
| | - Matthias Gesemann
- University of Zurich, Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology; Zurich CH-8057 Switzerland
| | - Manuela Berger
- University of Zurich, Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology; Zurich CH-8057 Switzerland
| | - Stephan C. F. Neuhauss
- University of Zurich, Institute of Molecular Life Sciences, Neuroscience Center Zurich and Center for Integrative Human Physiology; Zurich CH-8057 Switzerland
| |
Collapse
|
30
|
Rajala A, Wang Y, Abcouwer SF, Gardner TW, Rajala RV. Developmental and light regulation of tumor suppressor protein PP2A in the retina. Oncotarget 2018; 9:1505-1523. [PMID: 29416710 PMCID: PMC5788578 DOI: 10.18632/oncotarget.23351] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 11/28/2017] [Indexed: 02/07/2023] Open
Abstract
Protein phosphatases are a group of universal enzymes that are responsible for the dephosphorylation of various proteins and enzymes in cells. Cellular signal transduction events are largely governed by the phosphorylation of key proteins. The length of cellular response depends on the activation of protein phosphatase that dephosphorylates the phosphate groups to halt a biological response, and fine-tune the defined cellular outcome. Dysregulation of these phosphatase(s) results in various disease phenotypes. The retina is a post-mitotic tissue, and oncogenic tyrosine and serine/ threonine kinase activities are important for retinal neuron survival. Aberrant activation of protein phosphatase(s) may have a negative effect on retinal neurons. In the current study, we characterized tumor suppressor protein phosphatase 2 (PP2A), a major serine/ threonine kinase with a broad substrate specificity. Our data suggest that PP2A is developmentally regulated in the retina, localized predominantly in the inner retina, and expressed in photoreceptor inner segments. Our findings indicate that PKCα and mTOR may serve as PP2A substrates. We found that light regulates PP2A activity. Our studies also suggest that rhodopsin regulates PP2A and its substrate(s) dephosphorylation. PP2A substrate phosphorylation is increased in mice lacking the A-subunit of PP2A. However, there is no accompanying effect on retina structure and function. Together, our findings suggest that controlling the activity of PP2A in the retina may be neuroprotective.
Collapse
Affiliation(s)
- Ammaji Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Yuhong Wang
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Steven F. Abcouwer
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA
- W.K. Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Thomas W. Gardner
- Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA
- W.K. Kellogg Eye Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Raju V.S. Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
31
|
Martemyanov KA, Sampath AP. The Transduction Cascade in Retinal ON-Bipolar Cells: Signal Processing and Disease. Annu Rev Vis Sci 2017; 3:25-51. [PMID: 28715957 DOI: 10.1146/annurev-vision-102016-061338] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Our robust visual experience is based on the reliable transfer of information from our photoreceptor cells, the rods and cones, to higher brain centers. At the very first synapse of the visual system, information is split into two separate pathways, ON and OFF, which encode increments and decrements in light intensity, respectively. The importance of this segregation is borne out in the fact that receptive fields in higher visual centers maintain a separation between ON and OFF regions. In the past decade, the molecular mechanisms underlying the generation of ON signals have been identified, which are unique in their use of a G-protein signaling cascade. In this review, we consider advances in our understanding of G-protein signaling in ON-bipolar cell (BC) dendrites and how insights about signaling have emerged from visual deficits, mostly night blindness. Studies of G-protein signaling in ON-BCs reveal an intricate mechanism that permits the regulation of visual sensitivity over a wide dynamic range.
Collapse
Affiliation(s)
| | - Alapakkam P Sampath
- Jules Stein Eye Institute, University of California, Los Angeles, California 90095;
| |
Collapse
|
32
|
A Naturally Occurring Canine Model of Autosomal Recessive Congenital Stationary Night Blindness. PLoS One 2015; 10:e0137072. [PMID: 26368928 PMCID: PMC4569341 DOI: 10.1371/journal.pone.0137072] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/12/2015] [Indexed: 11/20/2022] Open
Abstract
Congenital stationary night blindness (CSNB) is a non-progressive, clinically and genetically heterogeneous disease of impaired night vision. We report a naturally-occurring, stationary, autosomal recessive phenotype in beagle dogs with normal daylight vision but absent night vision. Affected dogs had normal retinas on clinical examination, but showed no detectable rod responses. They had “negative-type” mixed rod and cone responses in full-field ERGs. Their photopic long-flash ERGs had normal OFF-responses associated with severely reduced ON-responses. The phenotype is similar to the Schubert-Bornschein form of complete CSNB in humans. Homozygosity mapping ruled out most known CSNB candidates as well as CACNA2D4 and GNB3. Three remaining genes were excluded based on sequencing the open reading frame and intron-exon boundaries (RHO, NYX), causal to a different form of CSNB (RHO) or X-chromosome (NYX, CACNA1F) location. Among the genes expressed in the photoreceptors and their synaptic terminals, and mGluR6 cascade and modulators, reduced expression of GNAT1, CACNA2D4 and NYX was observed by qRT-PCR in both carrier (n = 2) and affected (n = 2) retinas whereas CACNA1F was down-regulated only in the affecteds. Retinal morphology revealed normal cellular layers and structure, and electron microscopy showed normal rod spherules and synaptic ribbons. No difference from normal was observed by immunohistochemistry (IHC) for antibodies labeling rods, cones and their presynaptic terminals. None of the retinas showed any sign of stress. Selected proteins of mGluR6 cascade and its modulators were examined by IHC and showed that PKCα weakly labeled the rod bipolar somata in the affected, but intensely labeled axonal terminals that appeared thickened and irregular. Dendritic terminals of ON-bipolar cells showed increased Goα labeling. Both PKCα and Goα labeled the more prominent bipolar dendrites that extended into the OPL in affected but not normal retinas. Interestingly, RGS11 showed no labeling in the affected retina. Our results indicate involvement of a yet unknown gene in this canine model of complete CSNB.
Collapse
|
33
|
Xiong WH, Pang JJ, Pennesi ME, Duvoisin RM, Wu SM, Morgans CW. The Effect of PKCα on the Light Response of Rod Bipolar Cells in the Mouse Retina. Invest Ophthalmol Vis Sci 2015; 56:4961-74. [PMID: 26230760 DOI: 10.1167/iovs.15-16622] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Protein kinase C α (PKCα) is abundantly expressed in rod bipolar cells (RBCs) in the retina, yet the physiological function of PKCα in these cells is not well understood. To elucidate the role of PKCα in visual processing in the eye, we examined the effect of genetic deletion of PKCα on the ERG and on RBC light responses in the mouse. METHODS Immunofluorescent labeling was performed on wild-type (WT), TRPM1 knockout, and PKCα knockout (PKC-KO) retina. Scotopic and photopic ERGs were recorded from WT and PKC-KO mice. Light responses of RBCs were measured using whole-cell recordings in retinal slices from WT and PKC-KO mice. RESULTS Protein kinase C alpha expression in RBCs is correlated with the activity state of the cell. Rod bipolar cells dendrites are a major site of PKCα phosphorylation. Electroretinogram recordings indicated that loss of PKCα affects the scotopic b-wave, including a larger peak amplitude, longer implicit time, and broader width of the b-wave. There were no differences in the ERG a- or c-wave between PKCα KO and WT mice, indicating no measurable effect of PKCα in photoreceptors or the RPE. The photopic ERG was unaffected consistent with the lack of detectable PKCα in cone bipolar cells. Whole-cell recordings from RBCs in PKC-KO retinal slices revealed that, compared with WT, RBC light responses in the PKC-KO retina are delayed and of longer duration. CONCLUSIONS Protein kinase C alpha plays an important modulatory role in RBCs, regulating both the peak amplitude and temporal properties of the RBC light response in the rod visual pathway.
Collapse
Affiliation(s)
- Wei-Hong Xiong
- Department of Physiology & Pharmacology Oregon Health & Science University, Portland, Oregon, United States
| | - Ji-Jie Pang
- Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, United States
| | - Mark E Pennesi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Robert M Duvoisin
- Department of Physiology & Pharmacology Oregon Health & Science University, Portland, Oregon, United States
| | - Samuel M Wu
- Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, United States
| | - Catherine W Morgans
- Department of Physiology & Pharmacology Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
34
|
Transcription factor PRDM8 is required for rod bipolar and type 2 OFF-cone bipolar cell survival and amacrine subtype identity. Proc Natl Acad Sci U S A 2015; 112:E3010-9. [PMID: 26023183 DOI: 10.1073/pnas.1505870112] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Retinal bipolar (BP) cells mediate the earliest steps in image processing in the visual system, but the genetic pathways that regulate their development and function are incompletely known. We identified PRDI-BF1 and RIZ homology domain containing 8 (PRDM8) as a highly conserved transcription factor that is abundantly expressed in mouse retina. During development and in maturity, PRDM8 is expressed strongly in BP cells and a fraction of amacrine and ganglion cells. To determine whether Prdm8 is essential to BP cell development or physiology, we targeted the gene in mice. Prdm8(EGFP/EGFP) mice showed nonprogressive b-wave deficits on electroretinograms, consistent with compromised BP cell function or circuitry resembling the incomplete form of human congenital stationary night blindness (CSNB). BP cell specification was normal in Prdm8(EGFP/EGFP) retina as determined by VSX2(+) cell numbers and retinal morphology at postnatal day 6. BP subtype differentiation was impaired, however, as indicated by absent or diminished expression of BP subtype-specific markers, including the putative PRDM8 regulatory target PKCα (Prkca) and its protein. By adulthood, rod bipolar (RB) and type 2 OFF-cone bipolar (CB) cells were nearly absent from Prdm8-null mice. Although no change was detected in total amacrine cell (AC) numbers, increased PRKCA(+) and cholinergic ACs and decreased GABAergic ACs were seen, suggesting an alteration in amacrine subtype identity. These findings establish that PRDM8 is required for RB and type 2 OFF-CB cell survival and amacrine subtype identity, and they present PRDM8 as a candidate gene for human CSNB.
Collapse
|
35
|
Ribic A, Liu X, Crair MC, Biederer T. Structural organization and function of mouse photoreceptor ribbon synapses involve the immunoglobulin protein synaptic cell adhesion molecule 1. J Comp Neurol 2014; 522:900-20. [PMID: 23982969 DOI: 10.1002/cne.23452] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/23/2013] [Accepted: 08/14/2013] [Indexed: 11/11/2022]
Abstract
Adhesive interactions in the retina instruct the developmental specification of inner retinal layers. However, potential roles of adhesion in the development and function of photoreceptor synapses remain incompletely understood. This contrasts with our understanding of synapse development in the CNS, which can be guided by select adhesion molecules such as the Synaptic Cell Adhesion Molecule 1 (SynCAM 1/CADM1/nectin-like 2 protein). This immunoglobulin superfamily protein modulates the development and plasticity of classical excitatory synapses. We show here by immunoelectron microscopy and immunoblotting that SynCAM 1 is expressed on mouse rod photoreceptors and their terminals in the outer nuclear and plexiform layers in a developmentally regulated manner. Expression of SynCAM 1 on rods is low in early postnatal stages (P3-P7) but increases after eye opening (P14). In support of functional roles in the photoreceptors, electroretinogram recordings demonstrate impaired responses to light stimulation in SynCAM 1 knockout (KO) mice. In addition, the structural integrity of synapses in the OPL requires SynCAM 1. Quantitative ultrastructural analysis of SynCAM 1 KO retina measured fewer fully assembled, triadic rod ribbon synapses. Furthermore, rod synapse ribbons are shortened in KO mice, and protein levels of Ribeye, a major structural component of ribbons, are reduced in SynCAM 1 KO retina. Together, our results implicate SynCAM 1 in the synaptic organization of the rod visual pathway and provide evidence for novel roles of synaptic adhesion in the structural and functional integrity of ribbon synapses.
Collapse
Affiliation(s)
- Adema Ribic
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, 06520-8024
| | | | | | | |
Collapse
|
36
|
Abstract
The b-wave is a major component of the electroretinogram that reflects the activity of depolarizing bipolar cells (DBCs). The b-wave is used diagnostically to identify patients with defects in DBC signaling or in transmission from photoreceptors to DBCs. In mouse models, an abnormal b-wave has been used to demonstrate a critical role of a particular protein in the release of glutamate from photoreceptor terminals, in establishing the structure of the photoreceptor-to-DBC synapse, in DBC signal transduction, and also in DBC development, survival, or metabolic support. The purpose of this review is to summarize these models and how they have advanced our understanding of outer retinal function.
Collapse
|
37
|
Ueno S, Nishiguchi KM, Tanioka H, Enomoto A, Yamanouchi T, Kondo M, Yasuma TR, Yasuda S, Kuno N, Takahashi M, Terasaki H. Degeneration of retinal on bipolar cells induced by serum including autoantibody against TRPM1 in mouse model of paraneoplastic retinopathy. PLoS One 2013; 8:e81507. [PMID: 24282602 PMCID: PMC3840061 DOI: 10.1371/journal.pone.0081507] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 10/14/2013] [Indexed: 11/18/2022] Open
Abstract
The paraneoplastic retinopathies (PRs) are a group of eye diseases characterized by a sudden and progressive dysfunction of the retina caused by an antibody against a protein in a neoplasm. Evidence has been obtained that the transient receptor potential melastatin 1 (TRPM1) protein was one of the antigens for the autoantibody against the ON bipolar cells in PR patients. However, it has not been determined how the autoantibody causes the dysfunction of the ON bipolar cells. We hypothesized that the antibody against TRPM1 in the serum of patients with PR causes a degeneration of retinal ON bipolar cells. To test this hypothesis, we injected the serum from the PR patient, previously shown to contain anti-TRPM1 antibodies by westerblot, intravitreally into mice and examined the effects on the retina. We found that the electroretinograms (ERGs) of the mice were altered acutely after the injection, and the shape of the ERGs resembled that of the patient with PR. Immunohistochemical analysis of the eyes injected with the serum showed immunoreactivity against bipolar cells only in wild-type animals and not in TRPM1 knockout mice,consistent with the serum containing anti-TRPM1 antibodies. Histology also showed that some of the bipolar cells were apoptotic by 5 hours after the injection in wild type mice, but no bipolar cell death was found in TRPM1 knockout mice, . At 3 months, the inner nuclear layer was thinner and the amplitudes of the ERGs were still reduced. These results indicate that the serum of a patient with PR contained an antibody against TRPM1 caused an acute death of retinal ON bipolar cells of mice.
Collapse
Affiliation(s)
- Shinji Ueno
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- * E-mail:
| | - Koji M. Nishiguchi
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidetoshi Tanioka
- Research and Development Center, Santen Pharmaceutical Co., Ltd., Ikoma, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Yamanouchi
- Research and Development Center, Santen Pharmaceutical Co., Ltd., Ikoma, Japan
| | - Mineo Kondo
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Testuhiro R. Yasuma
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shunsuke Yasuda
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Noriyuki Kuno
- Research and Development Center, Santen Pharmaceutical Co., Ltd., Ikoma, Japan
| | - Masahide Takahashi
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroko Terasaki
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
38
|
Piret G, Perez MT, Prinz CN. Neurite outgrowth and synaptophysin expression of postnatal CNS neurons on GaP nanowire arrays in long-term retinal cell culture. Biomaterials 2012; 34:875-87. [PMID: 23131535 DOI: 10.1016/j.biomaterials.2012.10.042] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 10/16/2012] [Indexed: 12/18/2022]
Abstract
We have established long-term cultures of postnatal retinal cells on arrays of gallium phosphide nanowires of different geometries. Rod and cone photoreceptors, ganglion cells and bipolar cells survived on the substrates for at least 18 days in vitro. Glial cells were also observed, but these did not overgrow the neuronal population. On nanowires, neurons extended numerous long and branched neurites that expressed the synaptic vesicle marker synaptophysin. The longest nanowires (4 μm long) allowed a greater attachment and neurite elongation and our analysis suggests that the length of the nanowire per se and/or the adsorption of biomolecules on the nanowires may have been important factors regulating the observed cell behavior. The study thus shows that CNS neurons are amenable to gallium phosphide nanowires, probably as they create conditions that more closely resemble those encountered in the in vivo environment. These findings suggest that gallium phosphide nanowires may be considered as a material of interest when improving existing or designing the next generation of implantable devices. The features of gallium phosphide nanowires can be precisely controlled, making them suitable for this purpose.
Collapse
Affiliation(s)
- Gaëlle Piret
- Division of Solid State Physics, The Nanometer Structure Consortium, Lund University, Sweden
| | | | | |
Collapse
|
39
|
Jeong C, Shin T. Immunohistochemical localization of protein kinase C (PKC) beta I in the pig retina during postnatal development. Acta Histochem 2012; 114:18-23. [PMID: 21474165 DOI: 10.1016/j.acthis.2011.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 01/23/2011] [Accepted: 01/24/2011] [Indexed: 01/07/2023]
Abstract
In order to investigate the expression of protein kinase C (PKC) beta I in the retinas of pigs during postnatal development, we analyzed retinas sampled from 3-day-old and 6-month-old pigs by Western blotting and immunohistochemistry. Western blot analysis detected the expression of PKC beta I in the retinas of 3-day-old piglets and it was increased significantly in the retinas of 6-month-old adult pigs. Immunohistochemical staining showed PKC beta I in the retinas of both groups. Immunohistochemistry of 3-day-old retinas revealed weak PKC beta I reactivity in the ganglion cell layer, inner plexiform layer, inner nuclear cell layer, outer plexiform layer and rod and cone cell layer. In the 6-month-old pig retina, the cellular localization of PKC beta I immunostaining was similar to that of the 3-day-old retina, where PKC beta I was localized in some glial fibrillary acidic protein-positive cells, glutamine synthetase-positive cells, parvalbumin-positive cells, and PKC alpha-positive cells in the retina. This is the first study to show the expression and cellular localization of PKC beta I in the retina of pigs with development, and these results suggest that PKC beta I, in accordance with PKC alpha, plays important roles in signal transduction pathways in the pig retina with development.
Collapse
|
40
|
Abstract
In the retina, light onset hyperpolarizes photoreceptors and depolarizes ON bipolar cells at the sign inverting photoreceptor-ON bipolar cell synapse. Transmission at this synapse is mediated by a signaling cascade comprised of mGluR6, a G-protein containing G(αo), and the cation channel TRP melastatin 1 (TRPM1). This system is thought to be common to both the rod- and ON-cone-driven pathways, which control vision under scotopic and photopic conditions, respectively. In this study, we present evidence that the rod pathway is uniquely susceptible to modulation by PKCα at the rod-rod bipolar cell synapse. Decreased production of DAG (an activator of PKC) by inhibition of PIP₂ (phosphatidylinositol-4,5-bisphosphate) hydrolysis caused depression of the TRPM1 current. Conversely, addition of a DAG analog, 2-acetyl-1-oleoyl-sn-glycerol (OAG), potentiated the current in rod bipolar cells but not in ON-cone bipolar cells. The potentiating effects of OAG were absent both in mutant mice that lack PKCα expression and in wild-type mice in which enzymatic activity of PKCα was pharmacologically inhibited. In addition, we found that, like other members of the TRPM subfamily, TRPM1 current is susceptible to voltage-independent inhibition by intracellular magnesium, and that modulation by PKCα relieves this inhibition, as the potentiating effects of OAG are absent in low intracellular magnesium. We conclude that activation of PKCα initiates a modulatory mechanism at the rod-rod bipolar cell synapse whose function is to reduce inhibition of the TRPM1 current by magnesium, thereby increasing the gain of transmission at this synapse.
Collapse
|