1
|
Ling Y, Wang Y, Ye J, Luan C, Bi A, Gu Y, Shi X. Changes in Intrinsically Photosensitive Retinal Ganglion Cells, Dopaminergic Amacrine Cells, and Their Connectivity in the Retinas of Lid Suture Myopia. Invest Ophthalmol Vis Sci 2024; 65:8. [PMID: 39230992 PMCID: PMC11379095 DOI: 10.1167/iovs.65.11.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
Purpose This study investigates alterations in intrinsically photosensitive retinal ganglion cells (ipRGCs) and dopaminergic amacrine cells (DACs) in lid suture myopia (LSM) rats. Methods LSM was induced in rats by suturing the right eyes for 4 weeks. Double immunofluorescence staining of ipRGCs and DACs in whole-mount retinas was performed to analyze changes in the density and morphology of control, LSM, and fellow eyes. Real-time quantitative PCR and Western blotting were used to detect related genes and protein expression levels. Results Significant myopia was induced in the lid-sutured eye, but the fellow eye was not different to control. Decreased ipRGC density with paradoxically increased overall melanopsin expression and enlarged dendritic beads was observed in both the LSM and fellow eyes of the LSM rat retinas. In contrast, DAC changes occurred only in the LSM eyes, with reduced DAC density and tyrosine hydroxylase (TH) expression, sparser dendritic processes, and fewer varicosities. Interestingly, contacts between ipRGCs and DACs in the inner plexiform layer (IPL) and the expression of pituitary adenylate cyclase-activating polypeptide (PACAP) and vesicular monoamine transporter protein 2 (VMAT2) mRNA were decreased in the LSM eyes. Conclusions The ipRGCs and DACs in LSM rat retinas undergo multiple alterations in density, morphology, and related molecule expressions. However, the ipRGC changes alone appear not to be required for the development of myopia, given that myopia is only induced in the lid-sutured eye, and they are unlikely alone to drive the DAC changes. Reduced contacts between ipRGCs and DACs in the LSM eyes may be the structural foundation for the impaired signaling between them. PACAP and VMAT2, strongly associated with ipRGCs and DACs, may play important roles in LSM through complex mechanisms.
Collapse
Affiliation(s)
- Ying Ling
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yao Wang
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Jingjing Ye
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changlin Luan
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Ailing Bi
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Gu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xuefeng Shi
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Institute of Ophthalmology, Nankai University, Tianjin, China
| |
Collapse
|
2
|
Romano EJ, Zhang DQ. Dopaminergic amacrine cells express HCN channels in the developing and adult mouse retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.20.604440. [PMID: 39091772 PMCID: PMC11291019 DOI: 10.1101/2024.07.20.604440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Purpose To determine the molecular and functional expression of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in developing and mature dopaminergic amacrine cells (DACs), the sole source of ocular dopamine that plays a vital role in visual function and eye development. Methods HCN channels are encoded by isoforms 1-4. HCN1, HCN2, and HCN4 were immunostained in retinal slices obtained from mice at postnatal day 4 (P4), P8, and P12 as well as in adults. Each HCN channel isoform was also immunostained with tyrosine hydroxylase, a marker for DACs, at P12 and adult retinas. Genetically-marked DACs were recorded in flat-mount retina preparation using a whole-cell current-clamp technique. Results HCN1 was expressed in rods/cones, amacrine cells, and retinal ganglion cells (RGCs) at P4, along with bipolar cells by P12. Different from HCN1, HCN2 and HCN4 were each expressed in amacrine cells and RGCs at P4, along with bipolar cells by P8, and in rods/cones by P12. Double immunostaining shows that each of the three isoforms was expressed in approximately half of DACs at P12 but in almost all DACs in adults. Electrophysiology results demonstrate that HCN channel isoforms form functional HCN channels, and the pharmacological blockade of HCN channels reduced the spontaneous firing frequency in most DACs. Conclusions Each class of retinal neurons may use different isoforms of HCN channels to function during development. HCN1, HCN2, and HCN4 form functional HCN channels in DACs, which appears to modulate their spontaneous firing activity.
Collapse
Affiliation(s)
- Emilio J Romano
- Eye Research Institute, Oakland University, Rochester, Michigan
| | - Dao-Qi Zhang
- Eye Research Institute, Oakland University, Rochester, Michigan
- Eye Research Center, Oakland University William Beaumont School of Medicine, Rochester, Michigan
| |
Collapse
|
3
|
Lau MYH, Gadiwalla S, Jones S, Galliano E. Different electrophysiological profiles of genetically labelled dopaminergic neurons in the mouse midbrain and olfactory bulb. Eur J Neurosci 2024; 59:1480-1499. [PMID: 38169095 DOI: 10.1111/ejn.16239] [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: 08/29/2023] [Revised: 12/06/2023] [Accepted: 12/09/2023] [Indexed: 01/05/2024]
Abstract
Dopaminergic (DA) neurons play pivotal roles in diverse brain functions, spanning movement, reward processing and sensory perception. DA neurons are most abundant in the midbrain (Substantia Nigra pars compacta [SNC] and Ventral Tegmental Area [VTA]) and the olfactory bulb (OB) in the forebrain. Interestingly, a subtype of OB DA neurons is capable of regenerating throughout life, while a second class is exclusively born during embryonic development. Compelling evidence in SNC and VTA also indicates substantial heterogeneity in terms of morphology, connectivity and function. To further investigate this heterogeneity and directly compare form and function of midbrain and forebrain bulbar DA neurons, we performed immunohistochemistry and whole-cell patch-clamp recordings in ex vivo brain slices from juvenile DAT-tdTomato mice. After confirming the penetrance and specificity of the dopamine transporter (DAT) Cre line, we compared soma shape, passive membrane properties, voltage sags and action potential (AP) firing across midbrain and forebrain bulbar DA subtypes. We found that each DA subgroup within midbrain and OB was highly heterogeneous, and that DA neurons across the two brain areas are also substantially different. These findings complement previous work in rats as well as gene expression and in vivo datasets, further questioning the existence of a single "dopaminergic" neuronal phenotype.
Collapse
Affiliation(s)
- Maggy Yu Hei Lau
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Sana Gadiwalla
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Susan Jones
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| | - Elisa Galliano
- Department of Physiology, Development and Neuroscience, University of Cambridge, UK
| |
Collapse
|
4
|
Felder-Schmittbuhl MP, Hicks D, Ribelayga CP, Tosini G. Melatonin in the mammalian retina: Synthesis, mechanisms of action and neuroprotection. J Pineal Res 2024; 76:e12951. [PMID: 38572848 DOI: 10.1111/jpi.12951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/09/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
Melatonin is an important player in the regulation of many physiological functions within the body and in the retina. Melatonin synthesis in the retina primarily occurs during the night and its levels are low during the day. Retinal melatonin is primarily synthesized by the photoreceptors, but whether the synthesis occurs in the rods and/or cones is still unclear. Melatonin exerts its influence by binding to G protein-coupled receptors named melatonin receptor type 1 (MT1) and type 2 (MT2). MT1 and MT2 receptors activate a wide variety of signaling pathways and both receptors are present in the vertebrate photoreceptors where they may form MT1/MT2 heteromers (MT1/2h). Studies in rodents have shown that melatonin signaling plays an important role in the regulation of retinal dopamine levels, rod/cone coupling as well as the photopic and scotopic electroretinogram. In addition, melatonin may play an important role in protecting photoreceptors from oxidative stress and can protect photoreceptors from apoptosis. Critically, melatonin signaling is involved in the modulation of photoreceptor viability during aging and other studies have implicated melatonin in the pathogenesis of age-related macular degeneration. Hence melatonin may represent a useful tool in the fight to protect photoreceptors-and other retinal cells-against degeneration due to aging or diseases.
Collapse
Affiliation(s)
- Marie Paule Felder-Schmittbuhl
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives (UPR 3212), Université de Strasbourg, Strasbourg, France
| | - David Hicks
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives (UPR 3212), Université de Strasbourg, Strasbourg, France
| | - Christophe P Ribelayga
- Department of Vision Sciences, College of Optometry, University of Houston, Houston, Texas, USA
| | - Gianluca Tosini
- Department of Pharmacology & Toxicology, Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
5
|
Jo A, Deniz S, Xu J, Duvoisin RM, DeVries SH, Zhu Y. A sign-inverted receptive field of inhibitory interneurons provides a pathway for ON-OFF interactions in the retina. Nat Commun 2023; 14:5937. [PMID: 37741839 PMCID: PMC10517963 DOI: 10.1038/s41467-023-41638-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 09/11/2023] [Indexed: 09/25/2023] Open
Abstract
A fundamental organizing plan of the retina is that visual information is divided into ON and OFF streams that are processed in separate layers. This functional dichotomy originates in the ON and OFF bipolar cells, which then make excitatory glutamatergic synapses onto amacrine and ganglion cells in the inner plexiform layer. We have identified an amacrine cell (AC), the sign-inverting (SI) AC, that challenges this fundamental plan. The glycinergic, ON-stratifying SI-AC has OFF light responses. In opposition to the classical wiring diagrams, it receives inhibitory inputs from glutamatergic ON bipolar cells at mGluR8 synapses, and excitatory inputs from an OFF wide-field AC at electrical synapses. This "inhibitory ON center - excitatory OFF surround" receptive-field of the SI-AC allows it to use monostratified dendrites to conduct crossover inhibition and push-pull activation to enhance light detection by ACs and RGCs in the dark and feature discrimination in the light.
Collapse
Affiliation(s)
- Andrew Jo
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sercan Deniz
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jian Xu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Robert M Duvoisin
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Steven H DeVries
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| | - Yongling Zhu
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
6
|
Kinane C, Calligaro H, Jandot A, Coutanson C, Haddjeri N, Bennis M, Dkhissi-Benyahya O. Dopamine modulates the retinal clock through melanopsin-dependent regulation of cholinergic waves during development. BMC Biol 2023; 21:146. [PMID: 37365544 DOI: 10.1186/s12915-023-01647-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 06/08/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND The mammalian retina contains an autonomous circadian clock that controls various aspects of retinal physiology and function, including dopamine (DA) release by amacrine cells. This neurotransmitter plays a critical role in retina development, visual signalling, and phase resetting of the retinal clock in adulthood. Interestingly, bidirectional regulation between dopaminergic cells and melanopsin-expressing retinal ganglion cells has been demonstrated in the adult and during development. Additionally, the adult melanopsin knockout mouse (Opn4 -/-) exhibits a shortening of the endogenous period of the retinal clock. However, whether DA and / or melanopsin influence the retinal clock mechanism during its maturation is still unknown. RESULTS Using wild-type Per2 Luc and melanopsin knockout (Opn4 -/-::Per2 Luc) mice at different postnatal stages, we found that the retina generates self-sustained circadian rhythms from postnatal day 5 in both genotypes and that the ability to express these rhythms emerges in the absence of external time cues. Intriguingly, only in wild-type explants, DA supplementation lengthened the endogenous period of the clock during the first week of postnatal development through both D1- and D2-like dopaminergic receptors. Furthermore, the blockade of spontaneous cholinergic retinal waves, which drive DA release in the early developmental stages, shortened the period and reduced the light-induced phase shift of the retinal clock only in wild-type retinas. CONCLUSIONS These data suggest that DA modulates the molecular core of the clock through melanopsin-dependent regulation of acetylcholine retinal waves, thus offering an unprecedented role of DA and melanopsin in the endogenous functioning and the light response of the retinal clock during development.
Collapse
Affiliation(s)
- Chaimaa Kinane
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, University Cadi Ayyad, Marrakech, Morocco
| | - Hugo Calligaro
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France
- Salk Institute for Biological Studies, La Lolla, CA, USA
| | - Antonin Jandot
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France
| | - Christine Coutanson
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France
| | - Nasser Haddjeri
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France
| | - Mohamed Bennis
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, University Cadi Ayyad, Marrakech, Morocco
| | - Ouria Dkhissi-Benyahya
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France.
| |
Collapse
|
7
|
Poudel S, Rahimi-Nasrabadi H, Jin J, Najafian S, Alonso JM. Differences in visual stimulation between reading and walking and implications for myopia development. J Vis 2023; 23:3. [PMID: 37014657 PMCID: PMC10080958 DOI: 10.1167/jov.23.4.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 02/11/2023] [Indexed: 04/05/2023] Open
Abstract
Visual input plays an important role in the development of myopia (nearsightedness), a visual disorder that blurs vision at far distances. The risk of myopia progression increases with the time spent reading and decreases with outdoor activity for reasons that remain poorly understood. To investigate the stimulus parameters driving this disorder, we compared the visual input to the retina of humans performing two tasks associated with different risks of myopia progression, reading and walking. Human subjects performed the two tasks while wearing glasses with cameras and sensors that recorded visual scenes and visuomotor activity. When compared with walking, reading black text in white background reduced spatiotemporal contrast in central vision and increased it in peripheral vision, leading to a pronounced reduction in the ratio of central/peripheral strength of visual stimulation. It also made the luminance distribution heavily skewed toward negative dark contrast in central vision and positive light contrast in peripheral vision, decreasing the central/peripheral stimulation ratio of ON visual pathways. It also decreased fixation distance, blink rate, pupil size, and head-eye coordination reflexes dominated by ON pathways. Taken together with previous work, these results support the hypothesis that reading drives myopia progression by understimulating ON visual pathways.
Collapse
Affiliation(s)
- Sabina Poudel
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Hamed Rahimi-Nasrabadi
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Jianzhong Jin
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Sohrab Najafian
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Jose-Manuel Alonso
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| |
Collapse
|
8
|
Seilheimer RL, McClard CK, Sabharwal J, Wu SM. Modulation of narrow-field amacrine cells on light-evoked spike responses and receptive fields of retinal ganglion cells. Vision Res 2023; 205:108186. [PMID: 36764009 PMCID: PMC11339979 DOI: 10.1016/j.visres.2023.108186] [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: 11/21/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 02/10/2023]
Abstract
By using multi-electrode array (MEA) recording technique in conjunction with white-noise checkerboard stimuli and reverse correlation methods, we studied modulatory actions of glycinergic narrow-field amacrine cells (NFACs) on spatiotemporal profiles of five functional groups of ganglion cells (GCs) in dark-adapted mouse retinas. We found that application of 2 µM strychnine significantly altered light-evoked spike rates of three groups of GCs. It also decreased receptive field center radii of all five groups of GC by a mean value of 11%, and shifted the GC receptive field (RF) centers of all GCs and the mean shift distances for the sustained GCs are significantly longer than the transient GCs. On the other hand, strychnine did not affect temporal profiles of the GC center responses, as it did not alter the time-to-peak or the biphasic index of the spike triggered average (STA) functions of GC RF centers. Strychnine also exerts limited actions on RF surrounds of most GCs, except that it moderately weakens the antagonistic surround of sustained OFF GCs and strengthens the antagonistic surround of the ON/OFF GCs, possibly through serial connections between NFACs and GABAergic wide-field amacrine cells (WFACs). Using the Sum of Separable Subfilter (SoSS) model and singular value decomposition method, we decomposed GCs' STAs into five space-time separable subfilters, studied the observation rates of each subfilter in the five functional groups of GCs and determined NFAC-dependent and -independent synaptic circuitries that mediate center and surround responses of various groups of mouse retina retinal ganglion cells.
Collapse
Affiliation(s)
- R L Seilheimer
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States
| | - C K McClard
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States
| | - J Sabharwal
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States
| | - S M Wu
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States.
| |
Collapse
|
9
|
Raja S, Milosavljevic N, Allen AE, Cameron MA. Burning the candle at both ends: Intraretinal signaling of intrinsically photosensitive retinal ganglion cells. Front Cell Neurosci 2023; 16:1095787. [PMID: 36687522 PMCID: PMC9853061 DOI: 10.3389/fncel.2022.1095787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/13/2022] [Indexed: 01/09/2023] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) are photoreceptors located in the ganglion cell layer. They project to brain regions involved in predominately non-image-forming functions including entrainment of circadian rhythms, control of the pupil light reflex, and modulation of mood and behavior. In addition to possessing intrinsic photosensitivity via the photopigment melanopsin, these cells receive inputs originating in rods and cones. While most research in the last two decades has focused on the downstream influence of ipRGC signaling, recent studies have shown that ipRGCs also act retrogradely within the retina itself as intraretinal signaling neurons. In this article, we review studies examining intraretinal and, in addition, intraocular signaling pathways of ipRGCs. Through these pathways, ipRGCs regulate inner and outer retinal circuitry through both chemical and electrical synapses, modulate the outputs of ganglion cells (both ipRGCs and non-ipRGCs), and influence arrangement of the correct retinal circuitry and vasculature during development. These data suggest that ipRGC function plays a significant role in the processing of image-forming vision at its earliest stage, positioning these photoreceptors to exert a vital role in perceptual vision. This research will have important implications for lighting design to optimize the best chromatic lighting environments for humans, both in adults and potentially even during fetal and postnatal development. Further studies into these unique ipRGC signaling pathways could also lead to a better understanding of the development of ocular dysfunctions such as myopia.
Collapse
Affiliation(s)
- Sushmitha Raja
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Nina Milosavljevic
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Annette E. Allen
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Morven A. Cameron
- School of Medicine, Western Sydney University, Sydney, NSW, Australia,*Correspondence: Morven A. Cameron,
| |
Collapse
|
10
|
Sharma P, Ramachandran R. Retina regeneration: lessons from vertebrates. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac012. [PMID: 38596712 PMCID: PMC10913848 DOI: 10.1093/oons/kvac012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/24/2022] [Accepted: 06/25/2022] [Indexed: 04/11/2024]
Abstract
Unlike mammals, vertebrates such as fishes and frogs exhibit remarkable tissue regeneration including the central nervous system. Retina being part of the central nervous system has attracted the interest of several research groups to explore its regenerative ability in different vertebrate models including mice. Fishes and frogs completely restore the size, shape and tissue structure of an injured retina. Several studies have unraveled molecular mechanisms underlying retina regeneration. In teleosts, soon after injury, the Müller glial cells of the retina reprogram to form a proliferating population of Müller glia-derived progenitor cells capable of differentiating into various neural cell types and Müller glia. In amphibians, the transdifferentiation of retinal pigment epithelium and differentiation of ciliary marginal zone cells contribute to retina regeneration. In chicks and mice, supplementation with external growth factors or genetic modifications cause a partial regenerative response in the damaged retina. The initiation of retina regeneration is achieved through sequential orchestration of gene expression through controlled modulations in the genetic and epigenetic landscape of the progenitor cells. Several developmental biology pathways are turned on during the Müller glia reprogramming, retinal pigment epithelium transdifferentiation and ciliary marginal zone differentiation. Further, several tumorigenic pathways and gene expression events also contribute to the complete regeneration cascade of events. In this review, we address the various retinal injury paradigms and subsequent gene expression events governed in different vertebrate species. Further, we compared how vertebrates such as teleost fishes and amphibians can achieve excellent regenerative responses in the retina compared with their mammalian counterparts.
Collapse
Affiliation(s)
- Poonam Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Knowledge City, SAS Nagar, Sector 81, Manauli PO, 140306 Mohali, Punjab, India
| | - Rajesh Ramachandran
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Knowledge City, SAS Nagar, Sector 81, Manauli PO, 140306 Mohali, Punjab, India
| |
Collapse
|
11
|
Huang W, Xu Q, Liu F, Su J, Xiao D, Tang L, Hao ZZ, Liu R, Xiang K, Bi Y, Miao Z, Liu X, Liu Y, Liu S. Identification of TPBG-Expressing Amacrine Cells in DAT-tdTomato Mouse. Invest Ophthalmol Vis Sci 2022; 63:13. [PMID: 35551574 PMCID: PMC9123489 DOI: 10.1167/iovs.63.5.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
Purpose Neurons are the bricks of the neuronal system and experimental access to certain neuron subtypes will be of great help to decipher neuronal circuits. Here, we identified trophoblast glycoprotein (TPBG)-expressing GABAergic amacrine cells (ACs) that were selectively labeled in DAT-tdTomato transgenic mice. Methods Retina and brain sections were prepared for immunostaining with antibodies against various biomarkers. Patch-sequencing was performed to obtain the transcriptomes of tdTomato-positive cells in DAT-tdTomato mice. Whole-cell recordings were conducted to identify responses to light stimulation. Results Tyrosine hydroxylase immunoreactive cells were colocalized with tdTomato-positive cells in substantia nigra pars compacta, but not in the retina. Transcriptomes collected from tdTomato-positive cells in retinas via Patch-sequencing exhibited the expression of marker genes of ACs (Pax6 and Slc32a1) and marker genes of GABAergic neurons (Gad1, Gad2, and Slc6a1). Immunostaining with antibodies against relevant proteins (GAD67, GAD65, and GABA) also confirmed transcriptomic results. Furthermore, tdTomato-positive cells in retinas selectively expressed Tpbg, a marker gene for distinct clusters molecularly defined, which was proved with TPBG immunoreactivity in fluorescently labeled cells. Finally, tdTomato-positive cells recorded showed ON-OFF responses to light stimulation. Conclusions Ectopic expression occurs in the retina but not in the substantia nigra pars compacta in the DAT-tdTomato mouse, and fluorescently labeled cells in the retina are TPBG-expressing GABAergic ACs. This type of transgenic mice has been proved as an ideal tool to achieve efficient labeling of a distinct subset of ACs that selectively express Tpbg.
Collapse
Affiliation(s)
- Wanjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Qiang Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Feng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jing Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Dongchang Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lei Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Zhao-Zhe Hao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Ruifeng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Kangjian Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yalan Bi
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge CB10 1SD, United Kingdom
| | - Zhichao Miao
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge CB10 1SD, United Kingdom
- Translational Research Institute of Brain and Brain-Like Intelligence and Department of Anesthesiology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
- Research Unit of Ocular Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China
| |
Collapse
|
12
|
Dendro-somatic synaptic inputs to ganglion cells contradict receptive field and connectivity conventions in the mammalian retina. Curr Biol 2022; 32:315-328.e4. [PMID: 34822767 PMCID: PMC8792273 DOI: 10.1016/j.cub.2021.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/08/2021] [Accepted: 11/02/2021] [Indexed: 01/26/2023]
Abstract
The morphology of retinal neurons strongly influences their physiological function. Ganglion cell (GC) dendrites ramify in distinct strata of the inner plexiform layer (IPL) so that GCs responding to light increments (ON) or decrements (OFF) receive appropriate excitatory inputs. This vertical stratification prescribes response polarity and ensures consistent connectivity between cell types, whereas the lateral extent of GC dendritic arbors typically dictates receptive field (RF) size. Here, we identify circuitry in mouse retina that contradicts these conventions. AII amacrine cells are interneurons understood to mediate "crossover" inhibition by relaying excitatory input from the ON layer to inhibitory outputs in the OFF layer. Ultrastructural and physiological analyses show, however, that some AIIs deliver powerful inhibition to OFF GC somas and proximal dendrites in the ON layer, rendering the inhibitory RFs of these GCs smaller than their dendritic arbors. This OFF pathway, avoiding entirely the OFF region of the IPL, challenges several tenets of retinal circuitry. These results also indicate that subcellular synaptic organization can vary within a single population of neurons according to their proximity to potential postsynaptic targets.
Collapse
|
13
|
Lee YS, Choi SE, Hahm J, Kim MJ, Bae HS, Yi K, Lim HT, Hyon JY. Digital Therapeutics: Exploring the Possibilities of Digital Intervention for Myopia. Front Digit Health 2021; 3:710644. [PMID: 34713181 PMCID: PMC8521975 DOI: 10.3389/fdgth.2021.710644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/03/2021] [Indexed: 11/23/2022] Open
Abstract
Pediatric myopia is increasing globally and has become a major public health issue. However, the mechanism of pediatric myopia is still poorly understood, and there is no effective treatment to prevent its progression. Based on results from animal and clinical studies, certain neuronal–humoral factors (NHFs), such as IGF-1, dopamine, and cortisol may be involved in the progression of pediatric myopia. Digital therapeutics uses evidence-based software as therapeutic interventions and it has the potential to offer innovative treatment strategies for pediatric myopia beyond conventional treatment methods. In this perspective article, we introduce digital therapeutics SAT-001, a software algorithm that modulates the level of NHFs to reduce the progression of pediatric myopia. The proposed mechanism is based on a theoretical hypothesis derived from scientific research and clinical studies and will be further confirmed by evidence generated from clinical studies involving pediatric myopia.
Collapse
Affiliation(s)
| | | | - Jarang Hahm
- S-Alpha Therapeutics, Inc., Seoul, South Korea
| | - Myoung Joon Kim
- S-Alpha Therapeutics, Inc., Seoul, South Korea.,Renew Seoul Eye Center, Seoul, South Korea
| | | | - Kayoung Yi
- Department of Ophthalmology, Hallym University Kangnam Sacred Heart Hospital, Seoul, South Korea
| | - Hyun Taek Lim
- Department of Ophthalmology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, South Korea
| | - Joon Young Hyon
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul University Bundang Hospital, Seongnam, South Korea
| |
Collapse
|
14
|
Goel M, Mangel SC. Dopamine-Mediated Circadian and Light/Dark-Adaptive Modulation of Chemical and Electrical Synapses in the Outer Retina. Front Cell Neurosci 2021; 15:647541. [PMID: 34025356 PMCID: PMC8131545 DOI: 10.3389/fncel.2021.647541] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/06/2021] [Indexed: 12/17/2022] Open
Abstract
The vertebrate retina, like most other brain regions, undergoes relatively slow alterations in neural signaling in response to gradual changes in physiological conditions (e.g., activity changes to rest), or in response to gradual changes in environmental conditions (e.g., day changes into night). As occurs elsewhere in the brain, the modulatory processes that mediate slow adaptation in the retina are driven by extrinsic signals (e.g., changes in ambient light level) and/or by intrinsic signals such as those of the circadian (24-h) clock in the retina. This review article describes and discusses the extrinsic and intrinsic modulatory processes that enable neural circuits in the retina to optimize their visual performance throughout day and night as the ambient light level changes by ~10 billion-fold. In the first synaptic layer of the retina, cone photoreceptor cells form gap junctions with rods and signal cone-bipolar and horizontal cells (HCs). Distinct extrinsic and intrinsic modulatory processes in this synaptic layer are mediated by long-range feedback of the neuromodulator dopamine. Dopamine is released by dopaminergic cells, interneurons whose cell bodies are located in the second synaptic layer of the retina. Distinct actions of dopamine modulate chemical and electrical synapses in day and night. The retinal circadian clock increases dopamine release in the day compared to night, activating high-affinity dopamine D4 receptors on cones. This clock effect controls electrical synapses between rods and cones so that rod-cone electrical coupling is minimal in the day and robust at night. The increase in rod-cone coupling at night improves the signal-to-noise ratio and the reliability of very dim multi-photon light responses, thereby enhancing detection of large dim objects on moonless nights.Conversely, maintained (30 min) bright illumination in the day compared to maintained darkness releases sufficient dopamine to activate low-affinity dopamine D1 receptors on cone-bipolar cell dendrites. This non-circadian light/dark adaptive process regulates the function of GABAA receptors on ON-cone-bipolar cell dendrites so that the receptive field (RF) surround of the cells is strong following maintained bright illumination but minimal following maintained darkness. The increase in surround strength in the day following maintained bright illumination enhances the detection of edges and fine spatial details.
Collapse
Affiliation(s)
- Manvi Goel
- Department of Neuroscience, Ohio State University College of Medicine, Columbus, OH, United States
| | - Stuart C Mangel
- Department of Neuroscience, Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|
15
|
Amorim-de-Sousa A, Schilling T, Fernandes P, Seshadri Y, Bahmani H, González-Méijome JM. Blue light blind-spot stimulation upregulates b-wave and pattern ERG activity in myopes. Sci Rep 2021; 11:9273. [PMID: 33927248 PMCID: PMC8085027 DOI: 10.1038/s41598-021-88459-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 04/05/2021] [Indexed: 01/03/2023] Open
Abstract
Upregulation of retinal dopaminergic activity may be a target treatment for myopia progression. This study aimed to explore the viability of inducing changes in retinal electrical activity with short-wavelength light targeting melanopsin-expressing retinal ganglion cells (ipRGCs) passing through the optic nerve head. Fifteen healthy non-myopic or myopic young adults were recruited and underwent stimulation with blue light using a virtual reality headset device. Amplitudes and implicit times from photopic 3.0 b-wave and pattern electroretinogram (PERG) were measured at baseline and 10 and 20 min after stimulation. Relative changes were compared between non-myopes and myopes. The ERG b-wave amplitude was significantly larger 20 min after blind-spot stimulation compared to baseline (p < 0.001) and 10 min (p < 0.001) post-stimulation. PERG amplitude P50-N95 also showed a significant main effect for ‘Time after stimulation’ (p < 0.050). Implicit times showed no differences following blind-spot stimulation. PERG and b-wave changes after blind-spot stimulation were stronger in myopes than non-myopes. It is possible to induce significant changes in retinal electrical activity by stimulating ipRGCs axons at the optic nerve head with blue light. The results suggest that the changes in retinal electrical activity are located at the inner plexiform layer and are likely to involve the dopaminergic system.
Collapse
Affiliation(s)
- Ana Amorim-de-Sousa
- Clinical & Experimental Optometry Research Lab (CEORLab), Center of Physics (Optometry), School of Sciences, University of Minho, Gualtar, 4710-057, Braga, Portugal
| | | | - Paulo Fernandes
- Clinical & Experimental Optometry Research Lab (CEORLab), Center of Physics (Optometry), School of Sciences, University of Minho, Gualtar, 4710-057, Braga, Portugal
| | | | - Hamed Bahmani
- Dopavision GmbH, Berlin, Germany.,Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Bernstein Center for Computational Neuroscience, Tübingen, Germany
| | - José Manuel González-Méijome
- Clinical & Experimental Optometry Research Lab (CEORLab), Center of Physics (Optometry), School of Sciences, University of Minho, Gualtar, 4710-057, Braga, Portugal.
| |
Collapse
|
16
|
Galliano E, Hahn C, Browne LP, R Villamayor P, Tufo C, Crespo A, Grubb MS. Brief Sensory Deprivation Triggers Cell Type-Specific Structural and Functional Plasticity in Olfactory Bulb Neurons. J Neurosci 2021; 41:2135-2151. [PMID: 33483429 PMCID: PMC8018761 DOI: 10.1523/jneurosci.1606-20.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/11/2020] [Accepted: 11/17/2020] [Indexed: 02/03/2023] Open
Abstract
Can alterations in experience trigger different plastic modifications in neuronal structure and function, and if so, how do they integrate at the cellular level? To address this question, we interrogated circuitry in the mouse olfactory bulb responsible for the earliest steps in odor processing. We induced experience-dependent plasticity in mice of either sex by blocking one nostril for one day, a minimally invasive manipulation that leaves the sensory organ undamaged and is akin to the natural transient blockage suffered during common mild rhinal infections. We found that such brief sensory deprivation produced structural and functional plasticity in one highly specialized bulbar cell type: axon-bearing dopaminergic neurons in the glomerular layer. After 24 h naris occlusion, the axon initial segment (AIS) in bulbar dopaminergic neurons became significantly shorter, a structural modification that was also associated with a decrease in intrinsic excitability. These effects were specific to the AIS-positive dopaminergic subpopulation because no experience-dependent alterations in intrinsic excitability were observed in AIS-negative dopaminergic cells. Moreover, 24 h naris occlusion produced no structural changes at the AIS of bulbar excitatory neurons, mitral/tufted and external tufted cells, nor did it alter their intrinsic excitability. By targeting excitability in one specialized dopaminergic subpopulation, experience-dependent plasticity in early olfactory networks might act to fine-tune sensory processing in the face of continually fluctuating inputs.SIGNIFICANCE STATEMENT Sensory networks need to be plastic so they can adapt to changes in incoming stimuli. To see how cells in mouse olfactory circuits can change in response to sensory challenges, we blocked a nostril for just one day, a naturally relevant manipulation akin to the deprivation that occurs with a mild cold. We found that this brief deprivation induces forms of axonal and intrinsic functional plasticity in one specific olfactory bulb cell subtype: axon-bearing dopaminergic interneurons. In contrast, intrinsic properties of axon-lacking bulbar dopaminergic neurons and neighboring excitatory neurons remained unchanged. Within the same sensory circuits, specific cell types can therefore make distinct plastic changes in response to an ever-changing external landscape.
Collapse
Affiliation(s)
- Elisa Galliano
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3DY, United Kingdom
| | - Christiane Hahn
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Lorcan P Browne
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Paula R Villamayor
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Candida Tufo
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Andres Crespo
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Matthew S Grubb
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| |
Collapse
|
17
|
Abstract
Myopia is a globally emerging issue, with multiple medical and socio-economic burdens and no well-established causal treatment thus far. A better insight into altered biochemical pathways and underlying pathogenesis might facilitate early diagnosis and treatment of myopia, ultimately leading to the development of more effective preventive and therapeutic measures. In this review, we summarize current data about the metabolomics and proteomics of myopia in humans and present various experimental approaches and animal models, along with their strengths and weaknesses. We also discuss the potential applicability of these findings to medical practice and suggest directions for future research.
Collapse
|
18
|
Korshunov KS, Blakemore LJ, Trombley PQ. Illuminating and Sniffing Out the Neuromodulatory Roles of Dopamine in the Retina and Olfactory Bulb. Front Cell Neurosci 2020; 14:275. [PMID: 33110404 PMCID: PMC7488387 DOI: 10.3389/fncel.2020.00275] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/04/2020] [Indexed: 01/28/2023] Open
Abstract
In the central nervous system, dopamine is well-known as the neuromodulator that is involved with regulating reward, addiction, motivation, and fine motor control. Yet, decades of findings are revealing another crucial function of dopamine: modulating sensory systems. Dopamine is endogenous to subsets of neurons in the retina and olfactory bulb (OB), where it sharpens sensory processing of visual and olfactory information. For example, dopamine modulation allows the neural circuity in the retina to transition from processing dim light to daylight and the neural circuity in the OB to regulate odor discrimination and detection. Dopamine accomplishes these tasks through numerous, complex mechanisms in both neural structures. In this review, we provide an overview of the established and emerging research on these mechanisms and describe similarities and differences in dopamine expression and modulation of synaptic transmission in the retinas and OBs of various vertebrate organisms. This includes discussion of dopamine neurons’ morphologies, potential identities, and biophysical properties along with their contributions to circadian rhythms and stimulus-driven synthesis, activation, and release of dopamine. As dysregulation of some of these mechanisms may occur in patients with Parkinson’s disease, these symptoms are also discussed. The exploration and comparison of these two separate dopamine populations shows just how remarkably similar the retina and OB are, even though they are functionally distinct. It also shows that the modulatory properties of dopamine neurons are just as important to vision and olfaction as they are to motor coordination and neuropsychiatric/neurodegenerative conditions, thus, we hope this review encourages further research to elucidate these mechanisms.
Collapse
Affiliation(s)
- Kirill S Korshunov
- Department of Biological Science, Florida State University, Tallahassee, FL, United States.,Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Laura J Blakemore
- Department of Biological Science, Florida State University, Tallahassee, FL, United States.,Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Paul Q Trombley
- Department of Biological Science, Florida State University, Tallahassee, FL, United States.,Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
19
|
Hellmer CB, Bohl JM, Hall LM, Koehler CC, Ichinose T. Dopaminergic Modulation of Signal Processing in a Subset of Retinal Bipolar Cells. Front Cell Neurosci 2020; 14:253. [PMID: 32922266 PMCID: PMC7456991 DOI: 10.3389/fncel.2020.00253] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/23/2020] [Indexed: 11/13/2022] Open
Abstract
The retina and the olfactory bulb are the gateways to the visual and olfactory systems, respectively, similarly using neural networks to initiate sensory signal processing. Sensory receptors receive signals that are transmitted to neural networks before projecting to primary cortices. These networks filter sensory signals based on their unique features and adjust their sensitivities by gain control systems. Interestingly, dopamine modulates sensory signal transduction in both systems. In the retina, dopamine adjusts the retinal network for daylight conditions (“light adaptation”). In the olfactory system, dopamine mediates lateral inhibition between the glomeruli, resulting in odorant signal decorrelation and discrimination. While dopamine is essential for signal discrimination in the olfactory system, it is not understood whether dopamine has similar roles in visual signal processing in the retina. To elucidate dopaminergic effects on visual processing, we conducted patch-clamp recording from second-order retinal bipolar cells, which exhibit multiple types that can convey different temporal features of light. We recorded excitatory postsynaptic potentials (EPSPs) evoked by various frequencies of sinusoidal light in the absence and presence of a dopamine receptor 1 (D1R) agonist or antagonist. Application of a D1R agonist, SKF-38393, shifted the peak temporal responses toward higher frequencies in a subset of bipolar cells. In contrast, a D1R antagonist, SCH-23390, reversed the effects of SKF on these types of bipolar cells. To examine the mechanism of dopaminergic modulation, we recorded voltage-gated currents, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, and low-voltage activated (LVA) Ca2+ channels. SKF modulated HCN and LVA currents, suggesting that these channels are the target of D1R signaling to modulate visual signaling in these bipolar cells. Taken together, we found that dopamine modulates the temporal tuning of a subset of retinal bipolar cells. Consequently, we determined that dopamine plays a role in visual signal processing, which is similar to its role in signal decorrelation in the olfactory bulb.
Collapse
Affiliation(s)
- Chase B Hellmer
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Jeremy M Bohl
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Leo M Hall
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Christina C Koehler
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Tomomi Ichinose
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
20
|
Cameron MA, Morley JW, Pérez-Fernández V. Seeing the light: different photoreceptor classes work together to drive adaptation in the mammalian retina. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
21
|
Yan W, Laboulaye MA, Tran NM, Whitney IE, Benhar I, Sanes JR. Mouse Retinal Cell Atlas: Molecular Identification of over Sixty Amacrine Cell Types. J Neurosci 2020; 40:5177-5195. [PMID: 32457074 PMCID: PMC7329304 DOI: 10.1523/jneurosci.0471-20.2020] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/07/2020] [Accepted: 05/13/2020] [Indexed: 02/01/2023] Open
Abstract
Amacrine cells (ACs) are a diverse class of interneurons that modulate input from photoreceptors to retinal ganglion cells (RGCs), rendering each RGC type selectively sensitive to particular visual features, which are then relayed to the brain. While many AC types have been identified morphologically and physiologically, they have not been comprehensively classified or molecularly characterized. We used high-throughput single-cell RNA sequencing to profile >32,000 ACs from mice of both sexes and applied computational methods to identify 63 AC types. We identified molecular markers for each type and used them to characterize the morphology of multiple types. We show that they include nearly all previously known AC types as well as many that had not been described. Consistent with previous studies, most of the AC types expressed markers for the canonical inhibitory neurotransmitters GABA or glycine, but several expressed neither or both. In addition, many expressed one or more neuropeptides, and two expressed glutamatergic markers. We also explored transcriptomic relationships among AC types and identified transcription factors expressed by individual or multiple closely related types. Noteworthy among these were Meis2 and Tcf4, expressed by most GABAergic and most glycinergic types, respectively. Together, these results provide a foundation for developmental and functional studies of ACs, as well as means for genetically accessing them. Along with previous molecular, physiological, and morphologic analyses, they establish the existence of at least 130 neuronal types and nearly 140 cell types in the mouse retina.SIGNIFICANCE STATEMENT The mouse retina is a leading model for analyzing the development, structure, function, and pathology of neural circuits. A complete molecular atlas of retinal cell types provides an important foundation for these studies. We used high-throughput single-cell RNA sequencing to characterize the most heterogeneous class of retinal interneurons, amacrine cells, identifying 63 distinct types. The atlas includes types identified previously as well as many novel types. We provide evidence for the use of multiple neurotransmitters and neuropeptides, and identify transcription factors expressed by groups of closely related types. Combining these results with those obtained previously, we proposed that the mouse retina contains ∼130 neuronal types and is therefore comparable in complexity to other regions of the brain.
Collapse
Affiliation(s)
- Wenjun Yan
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Mallory A Laboulaye
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Nicholas M Tran
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Irene E Whitney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138
| | - Inbal Benhar
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138
| |
Collapse
|
22
|
Retinal biomarkers and pharmacological targets for Hermansky-Pudlak syndrome 7. Sci Rep 2020; 10:3972. [PMID: 32132582 PMCID: PMC7055265 DOI: 10.1038/s41598-020-60931-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 02/19/2020] [Indexed: 01/24/2023] Open
Abstract
Deletion of dystrobrevin binding protein 1 has been linked to Hermansky-Pudlak syndrome type 7 (HPS-7), a rare disease characterized by oculocutaneous albinism and retinal dysfunction. We studied dysbindin-1 null mutant mice (Dys−/−) to shed light on retinal neurodevelopment defects in HPS-7. We analyzed the expression of a focused set of miRNAs in retina of wild type (WT), Dys+/− and Dys−/− mice. We also investigated the retinal function of these mice through electroretinography (ERG). We found that miR-101-3p, miR-137, miR-186-5p, miR-326, miR-382-5p and miR-876-5p were up-regulated in Dys−/−mice retina. Dys−/− mice showed significant increased b-wave in ERG, compared to WT mice. Bioinformatic analysis highlighted that dysregulated miRNAs target synaptic plasticity and dopaminergic signaling pathways, affecting retinal functions of Dys−/− mice. Overall, the data indicate potential mechanisms in retinal neurodevelopment of Dys−/− mice, which may have translational significance in HSP-7 patients, both in terms of diagnostic/prognostic biomarkers and novel pharmacological targets.
Collapse
|
23
|
Ko GYP. Circadian regulation in the retina: From molecules to network. Eur J Neurosci 2020; 51:194-216. [PMID: 30270466 PMCID: PMC6441387 DOI: 10.1111/ejn.14185] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/16/2018] [Accepted: 08/20/2018] [Indexed: 12/14/2022]
Abstract
The mammalian retina is the most unique tissue among those that display robust circadian/diurnal oscillations. The retina is not only a light sensing tissue that relays light information to the brain, it has its own circadian "system" independent from any influence from other circadian oscillators. While all retinal cells and retinal pigment epithelium (RPE) possess circadian oscillators, these oscillators integrate by means of neural synapses, electrical coupling (gap junctions), and released neurochemicals (such as dopamine, melatonin, adenosine, and ATP), so the whole retina functions as an integrated circadian system. Dysregulation of retinal clocks not only causes retinal or ocular diseases, it also impacts the circadian rhythm of the whole body, as the light information transmitted from the retina entrains the brain clock that governs the body circadian rhythms. In this review, how circadian oscillations in various retinal cells are integrated, and how retinal diseases affect daily rhythms.
Collapse
Affiliation(s)
- Gladys Y-P Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, Texas
| |
Collapse
|
24
|
Wang M, Aleman AC, Schaeffel F. Probing the Potency of Artificial Dynamic ON or OFF Stimuli to Inhibit Myopia Development. Invest Ophthalmol Vis Sci 2019; 60:2599-2611. [PMID: 31219534 DOI: 10.1167/iovs.18-26471] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To determine whether equiluminant artificial dynamic ON or OFF stimuli on a computer screen can induce bidirectional changes in choroidal thickness (ChTh) in both humans and chickens, and whether such changes are associated with bidirectional changes in retinal dopamine release in chickens. Methods Experiment 1: Before and after ON or OFF stimulation for 1 hour, ChTh was measured with optical coherence tomography (OCT). Experiment 2: chicks (n = 14) were raised under ON or OFF stimulation for 3 hours. ChTh was determined by OCT. Experiment 3: chicks were raised for 7 days either under room light (500 lux, n = 11), dynamic ON stimulus (700 lux, n = 15), or dynamic OFF stimulus (700 lux, n = 7). In addition, negative lenses were attached to their right eyes. After experiments 2 and 3, retinal and vitreal dopamine (DA), and its metabolites, were measured by HPLC-electrochemical detection. Results Experiment 1: Dynamic ON stimuli caused thicker choroids (+5.3 ± 2.0 μm), whereas OFF stimuli caused choroidal thinning (-4.7 ± 0.5 μm) (right eye data only, P < 0.001). Experiment 2: After 3 hours, chickens developed thicker choroids with ON stimuli (+37.4 ± 12.4 μm) and thinner choroids with OFF stimuli (-11.3 ± 3.6 μm, difference P < 0.01). Vitreal DA, 3-methoxytyramine, and homovanillic acid levels were elevated after ON stimulation, compared with the OFF (P < 0.05). Experiment 3: After 7 days, chickens with lenses developed more myopia both with ON and OFF stimulation, compared with room light. ON stimulation increased vitreal DA compared with OFF. Conclusions Artificial dynamic ON or OFF stimuli had similar effects on ChTh in humans and chickens, but more work will be necessary to determine whether such stimuli can be used as novel interventions of myopia.
Collapse
Affiliation(s)
- Min Wang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Hunan Province, China.,Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Germany
| | - Andrea C Aleman
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Germany
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Germany
| |
Collapse
|
25
|
Liu LL, Alessio EJ, Spix NJ, Zhang DQ. Expression of GluA2-containing calcium-impermeable AMPA receptors on dopaminergic amacrine cells in the mouse retina. Mol Vis 2019; 25:780-790. [PMID: 31819340 PMCID: PMC6882663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/17/2019] [Indexed: 11/18/2022] Open
Abstract
Purpose The neuromodulator dopamine plays an important role in light adaptation for the visual system. Light can stimulate dopamine release from dopaminergic amacrine cells (DACs) by activating three classes of photosensitive retinal cells: rods, cones, and melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs). However, the synaptic mechanisms by which these photoreceptors excite DACs remain poorly understood. Our previous work demonstrated that α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptors contribute to light regulation of DAC activity. AMPA receptors are classified into Ca2+-permeable and Ca2+-impermeable subtypes. We sought to identify which subtype of AMPA receptors is involved in light regulation of DAC activity. Methods AMPA receptor-mediated light responses and miniature excitatory postsynaptic currents were recorded from genetically labeled DACs in mouse retinas with the whole-cell voltage-clamp mode. Immunostaining with antibodies against tyrosine hydroxylase, GluA2 (GluR2), and PSD-95 was performed in vertical retinal slices. Results The biophysical and pharmacological data showed that only Ca2+-impermeable AMPA receptors contribute to DAC light responses driven by ipRGCs or cones (via depolarizing bipolar cells). We further found that the same subtype of AMPA receptors mediates miniature excitatory postsynaptic currents of DACs. These findings are supported by the immunohistochemical results demonstrating that DACs express the PSD-95 with GluA2, a subunit that is essential for determining the impermeability of AMPA receptors to calcium. Conclusions The results indicated that GluA2-containing Ca2+-impermeable AMPA receptors contribute to signal transmission from photosensitive retinal cells to DACs.
Collapse
Affiliation(s)
- Lei-Lei Liu
- Eye Research Institute, Oakland University, Rochester, MI
| | | | - Nathan J Spix
- Eye Research Institute, Oakland University, Rochester, MI
| | - Dao-Qi Zhang
- Eye Research Institute, Oakland University, Rochester, MI
| |
Collapse
|
26
|
Van Hook MJ, Nawy S, Thoreson WB. Voltage- and calcium-gated ion channels of neurons in the vertebrate retina. Prog Retin Eye Res 2019; 72:100760. [PMID: 31078724 PMCID: PMC6739185 DOI: 10.1016/j.preteyeres.2019.05.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/25/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
In this review, we summarize studies investigating the types and distribution of voltage- and calcium-gated ion channels in the different classes of retinal neurons: rods, cones, horizontal cells, bipolar cells, amacrine cells, interplexiform cells, and ganglion cells. We discuss differences among cell subtypes within these major cell classes, as well as differences among species, and consider how different ion channels shape the responses of different neurons. For example, even though second-order bipolar and horizontal cells do not typically generate fast sodium-dependent action potentials, many of these cells nevertheless possess fast sodium currents that can enhance their kinetic response capabilities. Ca2+ channel activity can also shape response kinetics as well as regulating synaptic release. The L-type Ca2+ channel subtype, CaV1.4, expressed in photoreceptor cells exhibits specific properties matching the particular needs of these cells such as limited inactivation which allows sustained channel activity and maintained synaptic release in darkness. The particular properties of K+ and Cl- channels in different retinal neurons shape resting membrane potentials, response kinetics and spiking behavior. A remaining challenge is to characterize the specific distributions of ion channels in the more than 100 individual cell types that have been identified in the retina and to describe how these particular ion channels sculpt neuronal responses to assist in the processing of visual information by the retina.
Collapse
Affiliation(s)
- Matthew J Van Hook
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Scott Nawy
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department Pharmacology & Experimental Neuroscience(2), University of Nebraska Medical Center, Omaha, NE, USA
| | - Wallace B Thoreson
- Truhlsen Eye Institute, Department of Ophthalmology & Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department Pharmacology & Experimental Neuroscience(2), University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
27
|
Molecular Classification and Comparative Taxonomics of Foveal and Peripheral Cells in Primate Retina. Cell 2019; 176:1222-1237.e22. [PMID: 30712875 DOI: 10.1016/j.cell.2019.01.004] [Citation(s) in RCA: 292] [Impact Index Per Article: 58.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/08/2018] [Accepted: 12/31/2018] [Indexed: 01/03/2023]
Abstract
High-acuity vision in primates, including humans, is mediated by a small central retinal region called the fovea. As more accessible organisms lack a fovea, its specialized function and its dysfunction in ocular diseases remain poorly understood. We used 165,000 single-cell RNA-seq profiles to generate comprehensive cellular taxonomies of macaque fovea and peripheral retina. More than 80% of >60 cell types match between the two regions but exhibit substantial differences in proportions and gene expression, some of which we relate to functional differences. Comparison of macaque retinal types with those of mice reveals that interneuron types are tightly conserved. In contrast, projection neuron types and programs diverge, despite exhibiting conserved transcription factor codes. Key macaque types are conserved in humans, allowing mapping of cell-type and region-specific expression of >190 genes associated with 7 human retinal diseases. Our work provides a framework for comparative single-cell analysis across tissue regions and species.
Collapse
|
28
|
Jacoby J, Nath A, Jessen ZF, Schwartz GW. A Self-Regulating Gap Junction Network of Amacrine Cells Controls Nitric Oxide Release in the Retina. Neuron 2018; 100:1149-1162.e5. [PMID: 30482690 PMCID: PMC6317889 DOI: 10.1016/j.neuron.2018.09.047] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/28/2018] [Accepted: 09/25/2018] [Indexed: 01/31/2023]
Abstract
Neuromodulators regulate circuits throughout the nervous system, and revealing the cell types and stimulus conditions controlling their release is vital to understanding their function. The effects of the neuromodulator nitric oxide (NO) have been studied in many circuits, including in the vertebrate retina, where it regulates synaptic release, gap junction coupling, and blood vessel dilation, but little is known about the cells that release NO. We show that a single type of amacrine cell (AC) controls NO release in the inner retina, and we report its light responses, electrical properties, and calcium dynamics. We discover that this AC forms a dense gap junction network and that the strength of electrical coupling in the network is regulated by light through NO. A model of the network offers insights into the biophysical specializations leading to auto-regulation of NO release within the network.
Collapse
Affiliation(s)
- Jason Jacoby
- Department of Ophthalmology, Northwestern University, Chicago, IL, USA
| | - Amurta Nath
- Interdepartmental Neuroscience Program, Northwestern University, Chicago, IL, USA; Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, USA
| | - Zachary F Jessen
- Medical Scientist Training Program, Northwestern University, Chicago, IL, USA
| | - Gregory W Schwartz
- Department of Ophthalmology, Northwestern University, Chicago, IL, USA; Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
29
|
Guo L, Normando EM, Shah PA, De Groef L, Cordeiro MF. Oculo-visual abnormalities in Parkinson's disease: Possible value as biomarkers. Mov Disord 2018; 33:1390-1406. [DOI: 10.1002/mds.27454] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 05/16/2018] [Accepted: 05/18/2018] [Indexed: 12/16/2022] Open
Affiliation(s)
- Li Guo
- Glaucoma and Retinal Degenerative Disease Research Group, Institute of Ophthalmology; University College London; London UK
| | - Eduardo M. Normando
- Glaucoma and Retinal Degenerative Disease Research Group, Institute of Ophthalmology; University College London; London UK
- Western Eye Hospital, Imperial College Healthcare National Health Service Trust; London UK
- Imperial College Ophthalmology Research Group, Department of Surgery and Cancer, Imperial College London; London UK
| | - Parth Arvind Shah
- Glaucoma and Retinal Degenerative Disease Research Group, Institute of Ophthalmology; University College London; London UK
| | - Lies De Groef
- Glaucoma and Retinal Degenerative Disease Research Group, Institute of Ophthalmology; University College London; London UK
- Neural Circuit Development and Regeneration Research Group, Department of Biology; University of Leuven; Leuven Belgium
| | - M. Francesca Cordeiro
- Glaucoma and Retinal Degenerative Disease Research Group, Institute of Ophthalmology; University College London; London UK
- Western Eye Hospital, Imperial College Healthcare National Health Service Trust; London UK
- Imperial College Ophthalmology Research Group, Department of Surgery and Cancer, Imperial College London; London UK
| |
Collapse
|
30
|
Caravaggio F, Scifo E, Sibille EL, Hernandez-Da Mota SE, Gerretsen P, Remington G, Graff-Guerrero A. Expression of dopamine D2 and D3 receptors in the human retina revealed by positron emission tomography and targeted mass spectrometry. Exp Eye Res 2018; 175:32-41. [PMID: 29883636 DOI: 10.1016/j.exer.2018.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/24/2018] [Accepted: 06/04/2018] [Indexed: 11/26/2022]
Abstract
Dopamine D2 receptors (D2R) are expressed in the human retina and play an important role in the modulation of neural responses to light-adaptation. However, it is unknown whether dopamine D3 receptors (D3R) are expressed in the human retina. Using positron emission tomography (PET), we have observed significant uptake of the D3R-preferring agonist radiotracer [11C]-(+)-PHNO into the retina of humans in vivo. This led us to examine whether [11C]-(+)-PHNO binding in the retina was quantifiable using reference tissue methods and if D3R are expressed in human post-mortem retinal tissue. [11C]-(+)-PHNO data from 49 healthy controls (mean age: 39.96 ± 14.36; 16 female) and 12 antipsychotic-naïve patients with schizophrenia (mean age: 25.75 ± 6.25; 4 female) were analyzed. We observed no differences in [11C]-(+)-PHNO binding in the retina between first-episode, drug-naïve patients with schizophrenia and healthy controls. Post-mortem retinal tissues from four healthy persons (mean age: 59.75 ± 9.11; 2 female) and four patients with schizophrenia (mean age: 54 ± 17.11; 2 female) were analyzed using a targeted mass spectrometry technique: parallel reaction monitoring (PRM) analysis. Using targeted mass spectrometry, we confirmed that D3R are expressed in human retinal tissue ex vivo. Notably, there was far greater expression of D2R relative to D3R in the healthy human retina (∼12:1). Moreover, PRM analysis revealed reduced D2R, but not D3R, expression in the retinas of non-first episode patients with schizophrenia compared to healthy controls. We confirm that D3R are expressed in the human retina. Future studies are needed to determine what proportion of the [11C]-(+)-PHNO signal in the human retina in vivo is due to binding to D3R versus D2R. Knowledge that both D2R and D3R are expressed in the human retina, and potentially quantifiable in vivo using [11C]-(+)-PHNO, poses new research avenues for better understanding the role of retinal dopamine in human vision. This work may have important implications for elucidating pathophysiological and antipsychotic induced visual deficits in schizophrenia.
Collapse
Affiliation(s)
- Fernando Caravaggio
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada.
| | - Enzo Scifo
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases(DZNE), Bonn, Germany
| | - Etienne L Sibille
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Pharmacology and Toxicology, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | | | - Philip Gerretsen
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Gary Remington
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Ariel Graff-Guerrero
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| |
Collapse
|
31
|
Cheng Q, Wang H, Wu Y, Zhao S, Kong X, Chen Y, Jiang J. Highly selective enzymatic-free electrochemical sensor for dopamine detection based on the self-assemblied film of a sandwich mixed (phthalocyaninato) (porphyrinato) europium derivative. J PORPHYR PHTHALOCYA 2018. [DOI: 10.1142/s108842461750081x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
An efficient enzymatic-free electrochemical sensor is firstly developed based on the self-assemblied film of a sandwich mixed (phthalocyaninato) (porphyrinato) europium(III) double-decker complex, Eu(Pc)[T(OH)PP], [Pc = phthalocyaninate, T(OH)PP = 5,10,15,tris (4-tert-butylphenyl)-20-(4-hydroxyphenyl)porphyrinate] prepared by using a solution-processing QLS method. The Eu(Pc)[T(OH)PP]semiconducting active layer on an ITO working electrode leads to a good sensing property for the detection of dopamine with an excellent selectivity, due to the high Eu(Pc)[T(OH)PP] molecular ordering/packing in the QLS film and more favorable interaction between the Eu(Pc)[T(OH)PP] and DA molecules. The amperometric responses are linearly proportional to the concentration of dopamine in the range of 8–100 [Formula: see text]M, with a low detection limit of 4.8 [Formula: see text]M and good sensitivity, indicating the great potential of electroactive tetrapyrrole rare earth sandwich type complexes in the field of nonenzymatic electrochemical sensors.
Collapse
Affiliation(s)
- Qianqian Cheng
- School of Science, China University of Petroleum (East China), Qingdao 266580, China
| | - Haoyuan Wang
- School of Science, China University of Petroleum (East China), Qingdao 266580, China
| | - Yanling Wu
- School of Science, China University of Petroleum (East China), Qingdao 266580, China
| | - Shuai Zhao
- School of Science, China University of Petroleum (East China), Qingdao 266580, China
| | - Xia Kong
- School of Science, China University of Petroleum (East China), Qingdao 266580, China
| | - Yanli Chen
- School of Science, China University of Petroleum (East China), Qingdao 266580, China
| | - Jianzhuang Jiang
- School of Science, China University of Petroleum (East China), Qingdao 266580, China
- Beijing Key Laboratory for Science and Application of Functional Molecular and Crystalline Materials, Department of Chemistry, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
32
|
Dai H, Jackson CR, Davis GL, Blakely RD, McMahon DG. Is dopamine transporter-mediated dopaminergic signaling in the retina a noninvasive biomarker for attention-deficit/ hyperactivity disorder? A study in a novel dopamine transporter variant Val559 transgenic mouse model. J Neurodev Disord 2017; 9:38. [PMID: 29281965 PMCID: PMC5745861 DOI: 10.1186/s11689-017-9215-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 09/14/2017] [Indexed: 12/14/2022] Open
Abstract
Background Dopamine (DA) is a critical neuromodulator in the retina. Disruption of retinal DA synthesis and signaling significantly attenuates light-adapted, electroretinogram (ERG) responses, as well as contrast sensitivity and acuity. As these measures can be detected noninvasively, they may provide opportunities to detect disease processes linked to perturbed DA signaling. Recently, we identified a rare, functional DA transporter (DAT, SLC6A3) coding substitution, Ala559Val, in subjects with attention-deficit/hyperactivity disorder (ADHD), demonstrating that DAT Val559 imparts anomalous DA efflux (ADE) with attendant physiological, pharmacological, and behavioral phenotypes. To understand the broader impact of ADE on ADHD, noninvasive measures sensitive to DAT reversal are needed. Methods Here, we explored this question through ERG-based analysis of retinal light responses, as well as HPLC measurements of retinal DA in DAT Val559 mice. Results Male mice homozygous (HOM) for the DAT Val559 variant demonstrated increased, light-adapted ERG b-wave amplitudes compared to wild type (WT) and heterozygous (HET) mice, whereas dark-adapted responses were indistinguishable across genotypes. The elevated amplitude of the photopic light responses in HOM mice could be mimicked in WT mice by applying D1 and D4 DA receptor agonists and suppressed in HOM mice by introducing D4 antagonist, supporting elevated retinal DA signaling arising from ADE. Following the challenge with amphetamine, WT exhibited an increase in light-adapted response amplitudes, while HOM did not. Total retinal DA content was similar across genotypes. Interestingly, female DAT Val559 HOM animals revealed no significant difference in photopic ERG responses when compared with WT and HET littermates. Conclusions These data reveal that noninvasive, in vivo evaluation of retinal responses to light can reveal physiological signatures of ADE, suggesting a possible approach to the segregation of neurobehavioral disorders based on the DAT-dependent control of DA signaling.
Collapse
Affiliation(s)
- Heng Dai
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA
| | - Chad R Jackson
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA.,Present address: Defense Threat Reduction Agency, 8211 Terminal Road, Lorton, VA, 22079, USA
| | - Gwynne L Davis
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Randy D Blakely
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Douglas G McMahon
- Department of Biological Sciences, Vanderbilt University, Box 35-1634 Station B, Nashville, TN, 37235-1634, USA.
| |
Collapse
|
33
|
Oliveira FG, Nascimento-Júnior ESD, Cavalcante JC, Guzen FP, Cavalcante JDS, Soares JG, Cavalcanti JRLDP, Freitas LMD, Costa MSMDO, Andrade-da-Costa BLDS. Topographic specializations of catecholaminergic cells and ganglion cells and distribution of calcium binding proteins in the crepuscular rock cavy (Kerodon rupestris) retina. J Chem Neuroanat 2017; 90:57-69. [PMID: 29277705 DOI: 10.1016/j.jchemneu.2017.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 01/16/2023]
Abstract
The rock cavy (Kerodon rupestris) is a crepuscular Hystricomorpha rodent that has been used in comparative analysis of retinal targets, but its retinal organization remains to be investigated. In order to better characterize its visual system, the present study analyzed neurochemical features related to the topographic organization of catecholaminergic cells and ganglion cells, as well the distribution of calcium-binding proteins in the outer and inner retina. Retinal sections and/or wholemounts were processed using tyrosine hydroxylase (TH), GABA, calbindin, parvalbumin and calretinin immunohistochemistry or Nissl staining. Two types of TH-immunoreactive (TH-IR) cells were found which differ in soma size, dendritic arborization, intensity of TH immunoreactivity and stratification pattern in the inner plexiform layer. The topographic distribution of all TH-IR cells defines a visual streak along the horizontal meridian in the superior retina. The ganglion cells are also distributed in a visual streak and the visual acuity estimated considering their peak density is 4.13 cycles/degree. A subset of TH-IR cells express GABA or calbindin. Calretinin is abundant in most of retinal layers and coexists with calbindin in horizontal cells. Parvalbumin is less abundant and expressed by presumed amacrine cells in the INL and some ganglion cells in the GCL. The topographic distribution of TH-IR cells and ganglion cells in the rock cavy retina indicate a suitable adaptation for using a broad extension of its inferior visual field in aspects that involve resolution, adjustment to ambient light intensity and movement detection without specialized eye movements.
Collapse
Affiliation(s)
- Francisco Gilberto Oliveira
- Departamento de Ciências Biológicas, Centro de Ciências Biológicas e da Saúde, Universidade Regional do Cariri - URCA, Crato, CE, Brazil; Departamento de Morfologia, Laboratório de Neuroanatomia, Centro de Biociências, Universidade Federal do Rio Grande do Norte - UFRN, Natal, RN, Brazil
| | - Expedito Silva do Nascimento-Júnior
- Departamento de Morfologia, Laboratório de Neuroanatomia, Centro de Biociências, Universidade Federal do Rio Grande do Norte - UFRN, Natal, RN, Brazil
| | - Judney Cley Cavalcante
- Departamento de Morfologia, Laboratório de Neuroanatomia, Centro de Biociências, Universidade Federal do Rio Grande do Norte - UFRN, Natal, RN, Brazil
| | - Fausto Pierdoná Guzen
- Faculdade de Ciências da Saúde, Departamento de Ciências Biomédicas, Universidade do Estado do Rio Grande do Norte - UERN, Mossoró, RN, Brazil
| | - Jeferson de Souza Cavalcante
- Departamento de Morfologia, Laboratório de Neuroanatomia, Centro de Biociências, Universidade Federal do Rio Grande do Norte - UFRN, Natal, RN, Brazil; Departamento de Fisiologia, Laboratório de Neuroanatomia, Centro de Biociências, Universidade Federal do Rio Grande do Norte - UFRN, Natal, RN, Brazil
| | - Joacil Germano Soares
- Departamento de Morfologia, Laboratório de Neuroanatomia, Centro de Biociências, Universidade Federal do Rio Grande do Norte - UFRN, Natal, RN, Brazil
| | | | - Leandro Moura de Freitas
- Departamento de Morfologia, Laboratório de Neuroanatomia, Centro de Biociências, Universidade Federal do Rio Grande do Norte - UFRN, Natal, RN, Brazil
| | - Miriam Stela Maris de Oliveira Costa
- Departamento de Morfologia, Laboratório de Neuroanatomia, Centro de Biociências, Universidade Federal do Rio Grande do Norte - UFRN, Natal, RN, Brazil
| | | |
Collapse
|
34
|
Pons C, Mazade R, Jin J, Dul MW, Zaidi Q, Alonso JM. Neuronal mechanisms underlying differences in spatial resolution between darks and lights in human vision. J Vis 2017; 17:5. [PMID: 29196762 PMCID: PMC5713488 DOI: 10.1167/17.14.5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Artists and astronomers noticed centuries ago that humans perceive dark features in an image differently from light ones; however, the neuronal mechanisms underlying these dark/light asymmetries remained unknown. Based on computational modeling of neuronal responses, we have previously proposed that such perceptual dark/light asymmetries originate from a luminance/response saturation within the ON retinal pathway. Consistent with this prediction, here we show that stimulus conditions that increase ON luminance/response saturation (e.g., dark backgrounds) or its effect on light stimuli (e.g., optical blur) impair the perceptual discrimination and salience of light targets more than dark targets in human vision. We also show that, in cat visual cortex, the magnitude of the ON luminance/response saturation remains relatively constant under a wide range of luminance conditions that are common indoors, and only shifts away from the lowest luminance contrasts under low mesopic light. Finally, we show that the ON luminance/response saturation affects visual salience mostly when the high spatial frequencies of the image are reduced by poor illumination or optical blur. Because both low luminance and optical blur are risk factors in myopia, our results suggest a possible neuronal mechanism linking myopia progression with the function of the ON visual pathway.
Collapse
Affiliation(s)
- Carmen Pons
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, NY, USA
| | - Reece Mazade
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, NY, USA
| | - Jianzhong Jin
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, NY, USA
| | - Mitchell W Dul
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, NY, USA
| | - Qasim Zaidi
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, NY, USA
| | - Jose-Manuel Alonso
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, NY, USA
| |
Collapse
|
35
|
Liu LL, Spix NJ, Zhang DQ. NMDA Receptors Contribute to Retrograde Synaptic Transmission from Ganglion Cell Photoreceptors to Dopaminergic Amacrine Cells. Front Cell Neurosci 2017; 11:279. [PMID: 28959188 PMCID: PMC5603656 DOI: 10.3389/fncel.2017.00279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/29/2017] [Indexed: 11/13/2022] Open
Abstract
Recently, a line of evidence has demonstrated that the vertebrate retina possesses a novel retrograde signaling pathway. In this pathway, phototransduction is initiated by the photopigment melanopsin, which is expressed in a small population of retinal ganglion cells. These ganglion cell photoreceptors then signal to dopaminergic amacrine cells (DACs) through glutamatergic synapses, influencing visual light adaptation. We have previously demonstrated that in Mg2+-containing solution, α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptors mediate this glutamatergic transmission. Here, we demonstrate that removing extracellular Mg2+ enhances melanopsin-based DAC light responses at membrane potentials more negative than −40 mV. Melanopsin-based responses in Mg2+-free solution were profoundly suppressed by the selective N-methyl-D-aspartate (NMDA) receptor antagonist D-AP5. In addition, application of NMDA to the retina produced excitatory inward currents in DACs. These data strongly suggest that DACs express functional NMDA receptors. We further found that in the presence of Mg2+, D-AP5 reduced the peak amplitude of melanopsin-based DAC responses by ~70% when the cells were held at their resting membrane potential (−50 mV), indicating that NMDA receptors are likely to contribute to retrograde signal transmission to DACs under physiological conditions. Moreover, our data show that melanopsin-based NMDA-receptor-mediated responses in DACs are suppressed by antagonists specific to either the NR2A or NR2B receptor subtype. Immunohistochemical results show that NR2A and NR2B subunits are expressed on DAC somata and processes. These results suggest that DACs express functional NMDA receptors containing both NR2A and NR2B subunits. Collectively, our data reveal that, along with AMPA receptors, NR2A- and NR2B-containing NMDA receptors mediate retrograde signal transmission from ganglion cell photoreceptors to DACs.
Collapse
Affiliation(s)
- Lei-Lei Liu
- Eye Research Institute, Oakland UniversityRochester, MI, United States
| | - Nathan J Spix
- Eye Research Institute, Oakland UniversityRochester, MI, United States
| | - Dao-Qi Zhang
- Eye Research Institute, Oakland UniversityRochester, MI, United States
| |
Collapse
|
36
|
Qiao SN, Zhou W, Liu LL, Zhang DQ, Zhong YM. Orexin-A Suppresses Signal Transmission to Dopaminergic Amacrine Cells From Outer and Inner Retinal Photoreceptors. Invest Ophthalmol Vis Sci 2017; 58:4712-4721. [PMID: 28910447 PMCID: PMC5598320 DOI: 10.1167/iovs.17-21835] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Purpose The neuropeptides orexin-A and orexin-B are widely expressed in the vertebrate retina; however, their role in visual function is unclear. This study investigates whether and how orexins modulate signal transmission to dopaminergic amacrine cells (DACs) from both outer retinal photoreceptors (rods and cones) and inner retinal photoreceptors (melanopsin-expressing intrinsically photosensitive retinal ganglion cells [ipRGCs]). Methods A whole-cell voltage-clamp technique was used to record light-induced responses from genetically labeled DACs in flat-mount mouse retinas. Rod and cone signaling to DACs was confirmed pharmacologically (in wild-type retinas), whereas retrograde melanopsin signaling to DACs was isolated either pharmacologically (in wild-type retinas) or by genetic deletion of rod and cone function (in transgenic mice). Results Orexin-A attenuated rod/cone-mediated light responses in the majority of DACs and inhibited all DACs that exhibited melanopsin-based light responses, suggesting that exogenous orexin suppresses signal transmission from rods, cones, and ipRGCs to DACs. In addition, orexin receptor 1 antagonist SB334867 and orexin receptor 2 antagonist TCS OX229 enhanced melanopsin-based DAC responses, indicating that endogenous orexins inhibit signal transmission from ipRGCs to DACs. We further found that orexin-A inhibits melanopsin-based DAC responses via orexin receptors on DACs, whereas orexin-A may modulate signal transmission from rods and cones to DACs through activation of orexin receptors on DACs and their upstream neurons. Conclusions Our results suggest that orexins could influence visual function via the dopaminergic system in the mammalian retina.
Collapse
Affiliation(s)
- Sheng-Nan Qiao
- Institutes of Brain Science, Fudan University, Shanghai, China.,Eye Research Institute, Oakland University, Rochester, Michigan, United States
| | - Wei Zhou
- Institutes of Brain Science, Fudan University, Shanghai, China
| | - Lei-Lei Liu
- Eye Research Institute, Oakland University, Rochester, Michigan, United States
| | - Dao-Qi Zhang
- Eye Research Institute, Oakland University, Rochester, Michigan, United States
| | - Yong-Mei Zhong
- Institutes of Brain Science, Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Chaffiol A, Ishii M, Cao Y, Mangel SC. Dopamine Regulation of GABA A Receptors Contributes to Light/Dark Modulation of the ON-Cone Bipolar Cell Receptive Field Surround in the Retina. Curr Biol 2017; 27:2600-2609.e4. [PMID: 28844643 DOI: 10.1016/j.cub.2017.07.063] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 05/22/2017] [Accepted: 07/27/2017] [Indexed: 10/19/2022]
Abstract
Cone bipolar cells are interneurons that receive synaptic input from cone photoreceptor cells and provide the output of the first synaptic layer of the retina. These cells exhibit center-surround receptive fields, a prototype of lateral inhibition between neighboring sensory cells in which stimulation of the receptive field center excites the cell whereas stimulation of the surrounding region laterally inhibits the cell. This fundamental sensory coding mechanism facilitates spatial discrimination and detection of stimulus edges. However, although it is well established that the receptive field surround is strongest when ambient or background illumination is most intense, e.g., at midday, and that the surround is minimal following maintained darkness, the synaptic mechanisms that produce and modulate the surround have not been resolved. Using electrical recording of bipolar cells under experimental conditions in which the cells exhibited surround light responses, and light and electron microscopic immunocytochemistry, we show in the rabbit retina that bright-light-induced activation of dopamine D1 receptors located on ON-center cone bipolar cell dendrites increases the expression and activity of GABAA receptors on the dendrites of the cells and that surround light responses depend on endogenous GABAA receptor activation. We also show that maintained darkness and D1 receptor blockade following maintained illumination and D1 receptor activation result in minimal GABAA receptor expression and activity and greatly diminished surrounds. Modulation of the D1/GABAA receptor signaling pathway of ON-cBC dendrites by the ambient light level facilitates detection of spatial details on bright days and large dim objects on moonless nights.
Collapse
Affiliation(s)
- Antoine Chaffiol
- Department of Neuroscience, The Ohio State University College Of Medicine, Columbus, OH 43210, USA
| | - Masaaki Ishii
- Department of Neuroscience, The Ohio State University College Of Medicine, Columbus, OH 43210, USA
| | - Yu Cao
- Department of Neuroscience, The Ohio State University College Of Medicine, Columbus, OH 43210, USA
| | - Stuart C Mangel
- Department of Neuroscience, The Ohio State University College Of Medicine, Columbus, OH 43210, USA.
| |
Collapse
|
38
|
Dopamine Activation Preserves Visual Motion Perception Despite Noise Interference of Human V5/MT. J Neurosci 2017; 36:9303-12. [PMID: 27605607 DOI: 10.1523/jneurosci.4452-15.2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 06/27/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED When processing sensory signals, the brain must account for noise, both noise in the stimulus and that arising from within its own neuronal circuitry. Dopamine receptor activation is known to enhance both visual cortical signal-to-noise-ratio (SNR) and visual perceptual performance; however, it is unknown whether these two dopamine-mediated phenomena are linked. To assess this, we used single-pulse transcranial magnetic stimulation (TMS) applied to visual cortical area V5/MT to reduce the SNR focally and thus disrupt visual motion discrimination performance to visual targets located in the same retinotopic space. The hypothesis that dopamine receptor activation enhances perceptual performance by improving cortical SNR predicts that dopamine activation should antagonize TMS disruption of visual perception. We assessed this hypothesis via a double-blinded, placebo-controlled study with the dopamine receptor agonists cabergoline (a D2 agonist) and pergolide (a D1/D2 agonist) administered in separate sessions (separated by 2 weeks) in 12 healthy volunteers in a William's balance-order design. TMS degraded visual motion perception when the evoked phosphene and the visual stimulus overlapped in time and space in the placebo and cabergoline conditions, but not in the pergolide condition. This suggests that dopamine D1 or combined D1 and D2 receptor activation enhances cortical SNR to boost perceptual performance. That local visual cortical excitability was unchanged across drug conditions suggests the involvement of long-range intracortical interactions in this D1 effect. Because increased internal noise (and thus lower SNR) can impair visual perceptual learning, improving visual cortical SNR via D1/D2 agonist therapy may be useful in boosting rehabilitation programs involving visual perceptual training. SIGNIFICANCE STATEMENT In this study, we address the issue of whether dopamine activation improves visual perception despite increasing sensory noise in the visual cortex. We show specifically that dopamine D1 (or combined D1/D2) receptor activation enhances the cortical signal-to-noise-ratio to boost perceptual performance. Together with the previously reported effects of dopamine upon brain plasticity and learning (Wolf et al., 2003; Hansen and Manahan-Vaughan, 2014), our results suggest that combining rehabilitation with dopamine agonists could enhance both the saliency of the training signal and the long-term effects on brain plasticity to boost rehabilitation regimens for brain injury.
Collapse
|
39
|
Zhao X, Wong KY, Zhang DQ. Mapping physiological inputs from multiple photoreceptor systems to dopaminergic amacrine cells in the mouse retina. Sci Rep 2017; 7:7920. [PMID: 28801634 PMCID: PMC5554153 DOI: 10.1038/s41598-017-08172-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/07/2017] [Indexed: 01/28/2023] Open
Abstract
In the vertebrate retina, dopamine is synthesized and released by a specialized type of amacrine cell, the dopaminergic amacrine cell (DAC). DAC activity is stimulated by rods, cones, and melanopsin-expressing intrinsically photosensitive retinal ganglion cells upon illumination. However, the relative contributions of these three photoreceptor systems to the DAC light-induced response are unknown. Here we found that rods excite dark-adapted DACs across a wide range of stimulation intensities, primarily through connexin-36-dependent rod pathways. Similar rod-driven responses were observed in both ventral and dorsal DACs. We further found that in the dorsal retina, M-cones and melanopsin contribute to dark-adapted DAC responses with a similar threshold intensity. In the ventral retina, however, the threshold intensity for M-cone-driven responses was two log units greater than that observed in dorsal DACs, and melanopsin-driven responses were almost undetectable. We also examined the DAC response to prolonged adapting light and found such responses to be mediated by rods under dim lighting conditions, rods/M-cones/melanopsin under intermediate lighting conditions, and cones and melanopsin under bright lighting conditions. Our results elucidate the relative contributions of the three photoreceptor systems to DACs under different lighting conditions, furthering our understanding of the role these cells play in the visual system.
Collapse
Affiliation(s)
- Xiwu Zhao
- Eye Research Institute, Oakland University, Rochester, MI, United States.,Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Kwoon Y Wong
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States.,Department of Molecular, Cellular & Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Dao-Qi Zhang
- Eye Research Institute, Oakland University, Rochester, MI, United States.
| |
Collapse
|
40
|
M1 ipRGCs Influence Visual Function through Retrograde Signaling in the Retina. J Neurosci 2017; 36:7184-97. [PMID: 27383593 DOI: 10.1523/jneurosci.3500-15.2016] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 05/26/2016] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED Melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs, with five subtypes named M1-M5) are a unique subclass of RGCs with axons that project directly to many brain nuclei involved in non-image-forming functions such as circadian photoentrainment and the pupillary light reflex. Recent evidence suggests that melanopsin-based signals also influence image-forming visual function, including light adaptation, but the mechanisms involved are unclear. Intriguingly, a small population of M1 ipRGCs have intraretinal axon collaterals that project toward the outer retina. Using genetic mouse models, we provide three lines of evidence showing that these axon collaterals make connections with upstream dopaminergic amacrine cells (DACs): (1) ipRGC signaling to DACs is blocked by tetrodotoxin both in vitro and in vivo, indicating that ipRGC-to-DAC transmission requires voltage-gated Na(+) channels; (2) this transmission is partly dependent on N-type Ca(2+) channels, which are possibly expressed in the axon collateral terminals of ipRGCs; and (3) fluorescence microscopy reveals that ipRGC axon collaterals make putative presynaptic contact with DACs. We further demonstrate that elimination of M1 ipRGCs attenuates light adaptation, as evidenced by an impaired electroretinogram b-wave from cones, whereas a dopamine receptor agonist can potentiate the cone-driven b-wave of retinas lacking M1 ipRGCs. Together, the results strongly suggest that ipRGCs transmit luminance signals retrogradely to the outer retina through the dopaminergic system and in turn influence retinal light adaptation. SIGNIFICANCE STATEMENT Melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) comprise a third class of retinal photoreceptors that are known to mediate physiological responses such as circadian photoentrainment. However, investigation into whether and how ipRGCs contribute to vision has just begun. Here, we provide convergent anatomical and physiological evidence that axon collaterals of ipRGCs constitute a centrifugal pathway to DACs, conveying melanopsin-based signals from the innermost retina to the outer retina. We further demonstrate that retrograde signals likely influence visual processing because elimination of axon collateral-bearing ipRGCs impairs light adaptation by limiting dopamine-dependent facilitation of the cone pathway. Our findings strongly support the hypothesis that retrograde melanopsin-based signaling influences visual function locally within the retina, a notion that refutes the dogma that RGCs only provide physiological signals to the brain.
Collapse
|
41
|
Spix NJ, Liu LL, Zhang Z, Hohlbein JP, Prigge CL, Chintala S, Ribelayga CP, Zhang DQ. Vulnerability of Dopaminergic Amacrine Cells to Chronic Ischemia in a Mouse Model of Oxygen-Induced Retinopathy. Invest Ophthalmol Vis Sci 2017; 57:3047-57. [PMID: 27281270 PMCID: PMC4913805 DOI: 10.1167/iovs.16-19346] [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] [Indexed: 11/24/2022] Open
Abstract
Purpose Retinal dopamine deficiency is a potential cause of myopia and visual deficits in retinopathy of prematurity (ROP). We investigated the cellular mechanisms responsible for lowered levels of retinal dopamine in an oxygen-induced retinopathy (OIR) mouse model of ROP. Methods Retinopathy was induced by exposing mice to 75% oxygen from postnatal day 7 (P7) to P12. Oxygen-induced retinopathy and age-matched control mice were euthanized at P12, P17, P25, or P42 to P50. Immunohistochemistry, electrophysiology, and biochemical approaches were used to determine the effect of OIR on the structure and function of dopaminergic amacrine cells (DACs). Results The total number of DACs was unchanged in OIR retinas at P12 despite significant capillary dropout in the central retina. However, a significant loss of DACs was observed in P17 OIR retinas (in which neovascularization was maximal), with the cell loss being more profound in the central (avascular) than in the peripheral (neovascular) regions. Cell loss was persistent in both regions at P25, at which time retinal neovascularization had regressed. At P42, the percentage of DACs lost (54%) was comparable to the percent decrease in total dopamine content (53%). Additionally, it was found that DACs recorded in OIR retinas at P42 to P50 had a complete dendritic field and exhibited relatively normal spontaneous and light-induced electrical activity. Conclusions The results suggest that remaining DACs are structurally and functionally intact and that loss of DACs is primarily responsible for the decreased levels of retinal dopamine observed after OIR.
Collapse
Affiliation(s)
- Nathan J Spix
- Eye Research Institute Oakland University, Rochester, Michigan, United States
| | - Lei-Lei Liu
- Eye Research Institute Oakland University, Rochester, Michigan, United States
| | - Zhijing Zhang
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States
| | - Joshua P Hohlbein
- Eye Research Institute Oakland University, Rochester, Michigan, United States
| | - Cameron L Prigge
- Eye Research Institute Oakland University, Rochester, Michigan, United States
| | - Shravan Chintala
- Eye Research Institute Oakland University, Rochester, Michigan, United States
| | - Christophe P Ribelayga
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States 3Graduate School of Biomedical Sciences, The University of Texas Health Science Center at
| | - Dao-Qi Zhang
- Eye Research Institute Oakland University, Rochester, Michigan, United States
| |
Collapse
|
42
|
Sabbah S, Berg D, Papendorp C, Briggman KL, Berson DM. A Cre Mouse Line for Probing Irradiance- and Direction-Encoding Retinal Networks. eNeuro 2017; 4:ENEURO.0065-17.2017. [PMID: 28466070 PMCID: PMC5411164 DOI: 10.1523/eneuro.0065-17.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/31/2017] [Accepted: 04/10/2017] [Indexed: 01/18/2023] Open
Abstract
Cell type-specific Cre driver lines have revolutionized the analysis of retinal cell types and circuits. We show that the transgenic mouse Rbp4-Cre selectively labels several retinal neuronal types relevant to the encoding of absolute light intensity (irradiance) and visual motion. In the ganglion cell layer (GCL), most marked cells are wide-field spiking polyaxonal amacrine cells (ACs) with sustained irradiance-encoding ON responses that persist during chemical synaptic blockade. Their arbors spread about 1 mm across the retina and are restricted to the inner half of the ON sublamina of the inner plexiform layer (IPL). There, they costratify with dendrites of M2 intrinsically photosensitive retinal ganglion cells (ipRGCs), to which they are tracer coupled. We propose that synaptically driven and intrinsic photocurrents of M2 cells pass through gap junctions to drive AC light responses. Also marked in this mouse are two types of RGCs. R-cells have a bistratified dendritic arbor, weak directional tuning, and irradiance-encoding ON responses. However, they also receive excitatory OFF input, revealed during ON-channel blockade. Serial blockface electron microscopic (SBEM) reconstruction confirms OFF bipolar input, and reveals that some OFF input derives from a novel type of OFF bipolar cell (BC). R-cells innervate specific layers of the dorsal lateral geniculate nucleus (dLGN) and superior colliculus (SC). The other marked RGC type (RDS) is bistratified, transient, and ON-OFF direction selective (DS). It apparently innervates the nucleus of the optic tract (NOT). The Rbp4-Cre mouse will be valuable for targeting these cell types for further study and for selectively manipulating them for circuit analysis.
Collapse
Affiliation(s)
- Shai Sabbah
- Department of Neuroscience, Brown University, Providence, RI 02912
| | - Daniel Berg
- Department of Neuroscience, Brown University, Providence, RI 02912
| | - Carin Papendorp
- Department of Neuroscience, Brown University, Providence, RI 02912
| | - Kevin L. Briggman
- National Institute for Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - David M. Berson
- Department of Neuroscience, Brown University, Providence, RI 02912
| |
Collapse
|
43
|
Fasoli A, Dang J, Johnson JS, Gouw AH, Fogli Iseppe A, Ishida AT. Somatic and neuritic spines on tyrosine hydroxylase-immunopositive cells of rat retina. J Comp Neurol 2017; 525:1707-1730. [PMID: 28035673 DOI: 10.1002/cne.24166] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/13/2016] [Accepted: 12/27/2016] [Indexed: 12/27/2022]
Abstract
Dopamine- and tyrosine hydroxylase-immunopositive cells (TH cells) modulate visually driven signals as they flow through retinal photoreceptor, bipolar, and ganglion cells. Previous studies suggested that TH cells release dopamine from varicose axons arborizing in the inner and outer plexiform layers after glutamatergic synapses depolarize TH cell dendrites in the inner plexiform layer and these depolarizations propagate to the varicosities. Although it has been proposed that these excitatory synapses are formed onto appendages resembling dendritic spines, spines have not been found on TH cells of most species examined to date or on TH cell somata that release dopamine when exposed to glutamate receptor agonists. By use of protocols that preserve proximal retinal neuron morphology, we have examined the shape, distribution, and synapse-related immunoreactivity of adult rat TH cells. We report here that TH cell somata, tapering and varicose inner plexiform layer neurites, and varicose outer plexiform layer neurites all bear spines, that some of these spines are immunopositive for glutamate receptor and postsynaptic density proteins (viz., GluR1, GluR4, NR1, PSD-95, and PSD-93), that TH cell somata and tapering neurites are also immunopositive for a γ-aminobutyric acid (GABA) receptor subunit (GABAA Rα1 ), and that a synaptic ribbon-specific protein (RIBEYE) is found adjacent to some colocalizations of GluR1 and TH in the inner plexiform layer. These results identify previously undescribed sites at which glutamatergic and GABAergic inputs may stimulate and inhibit dopamine release, especially at somata and along varicose neurites that emerge from these somata and arborize in various levels of the retina. J. Comp. Neurol. 525:1707-1730, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Anna Fasoli
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California
| | - James Dang
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California
| | - Jeffrey S Johnson
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California
| | - Aaron H Gouw
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California
| | - Alex Fogli Iseppe
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California
| | - Andrew T Ishida
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California.,Department of Ophthalmology and Vision Science, University of California, Sacramento, California
| |
Collapse
|
44
|
Qiao SN, Zhang Z, Ribelayga CP, Zhong YM, Zhang DQ. Multiple cone pathways are involved in photic regulation of retinal dopamine. Sci Rep 2016; 6:28916. [PMID: 27356880 PMCID: PMC4928117 DOI: 10.1038/srep28916] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/10/2016] [Indexed: 12/24/2022] Open
Abstract
Dopamine is a key neurotransmitter in the retina and plays a central role in the light adaptive processes of the visual system. The sole source of retinal dopamine is dopaminergic amacrine cells (DACs). We and others have previously demonstrated that DACs are activated by rods, cones, and intrinsically photosensitive retinal ganglion cells (ipRGCs) upon illumination. However, it is still not clear how each class of photosensitive cells generates light responses in DACs. We genetically isolated cone function in mice to specifically examine the cone-mediated responses of DACs and their neural pathways. In addition to the reported excitatory input to DACs from light-increment (ON) bipolar cells, we found that cones alternatively signal to DACs via a retrograde signalling pathway from ipRGCs. Cones also produce ON and light-decrement (OFF) inhibitory responses in DACs, which are mediated by other amacrine cells, likely driven by type 1 and type 2/3a OFF bipolar cells, respectively. Dye injections indicated that DACs had similar morphological profiles with or without ON/OFF inhibition. Our data demonstrate that cones utilize specific parallel excitatory and inhibitory circuits to modulate DAC activity and efficiently regulate dopamine release and the light-adaptive state of the retina.
Collapse
Affiliation(s)
- Sheng-Nan Qiao
- Institutes of Brain Science, Fudan University, Shanghai 200032, China
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA
| | - Zhijing Zhang
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Christophe P. Ribelayga
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yong-Mei Zhong
- Institutes of Brain Science, Fudan University, Shanghai 200032, China
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
- Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Dao-Qi Zhang
- Eye Research Institute, Oakland University, Rochester, MI 48309, USA
| |
Collapse
|
45
|
Arroyo DA, Kirkby LA, Feller MB. Retinal Waves Modulate an Intraretinal Circuit of Intrinsically Photosensitive Retinal Ganglion Cells. J Neurosci 2016; 36:6892-905. [PMID: 27358448 PMCID: PMC4926237 DOI: 10.1523/jneurosci.0572-16.2016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/11/2016] [Accepted: 05/16/2016] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Before the maturation of rod and cone photoreceptors, the developing retina relies on light detection by intrinsically photosensitive retinal ganglion cells (ipRGCs) to drive early light-dependent behaviors. ipRGCs are output neurons of the retina; however, they also form functional microcircuits within the retina itself. Whether ipRGC microcircuits exist during development and whether they influence early light detection remain unknown. Here, we investigate the neural circuit that underlies the ipRGC-driven light response in developing mice. We use a combination of calcium imaging, tracer coupling, and electrophysiology experiments to show that ipRGCs form extensive gap junction networks that strongly contribute to the overall light response of the developing retina. Interestingly, we found that gap junction coupling was modulated by spontaneous retinal waves, such that acute blockade of waves dramatically increased the extent of coupling and hence increased the number of light-responsive neurons. Moreover, using an optical sensor, we found that this wave-dependent modulation of coupling is driven by dopamine that is phasically released by retinal waves. Our results demonstrate that ipRGCs form gap junction microcircuits during development that are modulated by retinal waves; these circuits determine the extent of the light response and thus potentially impact the processing of early visual information and light-dependent developmental functions. SIGNIFICANCE STATEMENT Light-dependent functions in early development are mediated by intrinsically photosensitive retinal ganglion cells (ipRGCs). Here we show that ipRGCs form an extensive gap junction network with other retinal neurons, including other ipRGCs, which shapes the retina's overall light response. Blocking cholinergic retinal waves, which are the primary source of neural activity before maturation of photoreceptors, increased the extent of ipRGC gap junction networks, thus increasing the number of light-responsive cells. We determined that this modulation of ipRGC gap junction networks occurs via dopamine released by waves. These results demonstrate that retinal waves mediate dopaminergic modulation of gap junction networks to regulate pre-vision light responses.
Collapse
Affiliation(s)
| | | | - Marla B Feller
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California-Berkeley, Berkeley, California 94720-3200
| |
Collapse
|
46
|
Trakhtenberg EF, Pita-Thomas W, Fernandez SG, Patel KH, Venugopalan P, Shechter JM, Morkin MI, Galvao J, Liu X, Dombrowski SM, Goldberg JL. Serotonin receptor 2C regulates neurite growth and is necessary for normal retinal processing of visual information. Dev Neurobiol 2016; 77:419-437. [PMID: 26999672 DOI: 10.1002/dneu.22391] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 01/25/2016] [Accepted: 03/12/2016] [Indexed: 12/21/2022]
Abstract
Serotonin (5HT) is present in a subpopulation of amacrine cells, which form synapses with retinal ganglion cells (RGCs), but little is known about the physiological role of retinal serotonergic circuitry. We found that the 5HT receptor 2C (5HTR2C) is upregulated in RGCs after birth. Amacrine cells generate 5HT and about half of RGCs respond to 5HTR2C agonism with calcium elevation. We found that there are on average 83 5HT+ amacrine cells randomly distributed across the adult mouse retina, all negative for choline acetyltransferase and 90% positive for tyrosine hydroxylase. We also investigated whether 5HTR2C and 5HTR5A affect RGC neurite growth. We found that both suppress neurite growth, and that RGCs from the 5HTR2C knockout (KO) mice grow longer neurites. Furthermore, 5HTR2C is subject to post-transcriptional editing, and we found that only the edited isoform's suppressive effect on neurite growth could be reversed by a 5HTR2C inverse agonist. Next, we investigated the physiological role of 5HTR2C in the retina, and found that 5HTR2C KO mice showed increased amplitude on pattern electroretinogram. Finally, RGC transcriptional profiling and pathways analysis suggested partial developmental compensation for 5HTR2C absence. Taken together, our findings demonstrate that 5HTR2C regulates neurite growth and RGC activity and is necessary for normal amplitude of RGC response to physiologic stimuli, and raise the hypothesis that these functions are modulated by a subset of 5HT+/ChAT-/TH+ amacrine cells as part of retinal serotonergic circuitry. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 419-437, 2017.
Collapse
Affiliation(s)
- Ephraim F Trakhtenberg
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurosurgery, Harvard Medical School, Boston, Massachusetts.,Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Wolfgang Pita-Thomas
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida.,Department of Anatomy and Neurobiology, Washington University, St. Louis, Missouri
| | - Stephanie G Fernandez
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Karan H Patel
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Praseeda Venugopalan
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurosurgery, Harvard Medical School, Boston, Massachusetts.,Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Jesse M Shechter
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Melina I Morkin
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Joana Galvao
- Shiley Eye Center, University of California, San Diego, California
| | - Xiongfei Liu
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Susan M Dombrowski
- Genomatix Software, Ann Arbor, Michigan.,Department of Obstetrics & Gynecology, Wayne State University School of Medicine, Detroit, Michigan
| | - Jeffrey L Goldberg
- Bascom Palmer Eye Institute and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, Florida.,Shiley Eye Center, University of California, San Diego, California.,Byers Eye Institute, Stanford University, Palo Alto, California
| |
Collapse
|
47
|
Parallel Inhibition of Dopamine Amacrine Cells and Intrinsically Photosensitive Retinal Ganglion Cells in a Non-Image-Forming Visual Circuit of the Mouse Retina. J Neurosci 2016; 35:15955-70. [PMID: 26631476 DOI: 10.1523/jneurosci.3382-15.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
An inner retinal microcircuit composed of dopamine (DA)-containing amacrine cells and melanopsin-containing, intrinsically photosensitive retinal ganglion cells (M1 ipRGCs) process information about the duration and intensity of light exposures, mediating light adaptation, circadian entrainment, pupillary reflexes, and other aspects of non-image-forming vision. The neural interaction is reciprocal: M1 ipRGCs excite DA amacrine cells, and these, in turn, feed inhibition back onto M1 ipRGCs. We found that the neuropeptide somatostatin [somatotropin release inhibiting factor (SRIF)] also inhibits the intrinsic light response of M1 ipRGCs and postulated that, to tune the bidirectional interaction of M1 ipRGCs and DA amacrine cells, SRIF amacrine cells would provide inhibitory modulation to both cell types. SRIF amacrine cells, DA amacrine cells, and M1 ipRGCs form numerous contacts. DA amacrine cells and M1 ipRGCs express the SRIF receptor subtypes sst(2A) and sst4 respectively. SRIF modulation of the microcircuit was investigated with targeted patch-clamp recordings of DA amacrine cells in TH-RFP mice and M1 ipRGCs in OPN4-EGFP mice. SRIF increases K(+) currents, decreases Ca(2+) currents, and inhibits spike activity in both cell types, actions reproduced by the selective sst(2A) agonist L-054,264 (N-[(1R)-2-[[[(1S*,3R*)-3-(aminomethyl)cyclohexyl]methyl]amino]-1-(1H-indol-3-ylmethyl)-2-oxoethyl]spiro[1H-indene-1,4'-piperidine]-1'-carboxamide) in DA amacrine cells and the selective sst4 agonist L-803,087 (N(2)-[4-(5,7-difluoro-2-phenyl-1H-indol-3-yl)-1-oxobutyl]-L-arginine methyl ester trifluoroacetate) in M1 ipRGCs. These parallel actions of SRIF may serve to counteract the disinhibition of M1 ipRGCs caused by SRIF inhibition of DA amacrine cells. This allows the actions of SRIF on DA amacrine cells to proceed with adjusting retinal DA levels without destabilizing light responses by M1 ipRGCs, which project to non-image-forming targets in the brain.
Collapse
|
48
|
Hirasawa H, Contini M, Raviola E. Extrasynaptic release of GABA and dopamine by retinal dopaminergic neurons. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0186. [PMID: 26009765 DOI: 10.1098/rstb.2014.0186] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In the mouse retina, dopaminergic amacrine (DA) cells synthesize both dopamine and GABA. Both transmitters are released extrasynaptically and act on neighbouring and distant retinal neurons by volume transmission. In simultaneous recordings of dopamine and GABA release from isolated perikarya of DA cells, a proportion of the events of dopamine and GABA exocytosis were simultaneous, suggesting co-release. In addition, DA cells establish GABAergic synapses onto AII amacrine cells, the neurons that transfer rod bipolar signals to cone bipolars. GABAA but not dopamine receptors are clustered in the postsynaptic membrane. Therefore, dopamine, irrespective of its site of release-synaptic or extrasynaptic-exclusively acts by volume transmission. Dopamine is released upon illumination and sets the gain of retinal neurons for vision in bright light. The GABA released at DA cells' synapses probably prevents signals from the saturated rods from entering the cone pathway when the dark-adapted retina is exposed to bright illumination. The GABA released extrasynaptically by DA and other amacrine cells may set a 'GABAergic tone' in the inner plexiform layer and thus counteract the effects of a spillover of glutamate released at the bipolar cell synapses of adjacent OFF and ON strata, thus preserving segregation of signals between ON and OFF pathways.
Collapse
Affiliation(s)
- Hajime Hirasawa
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA Department of Physiology, Faculty of Medicine, Saitama Medical University, 38 Morohongo, Moroyama, Saitama 350-0495, Japan
| | - Massimo Contini
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA Dipartimento di Medicina Sperimentale e Clinica, Viale Morgagni, 63, Firenze 50134, Italy
| | - Elio Raviola
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
49
|
Lin HJ, Hong ZY, Li YK, Liau I. Fluorescent tracer of dopamine enables selective labelling and interrogation of dopaminergic amacrine cells in the retina of living zebrafish. RSC Adv 2016. [DOI: 10.1039/c6ra13073a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
A ‘fluorescent dopamine’ that enables selective labeling and interrogation of retinal dopaminergic amacrine cells in living zebrafish was demonstrated.
Collapse
Affiliation(s)
- Hui-Jen Lin
- Department of Applied Chemistry
- Institute of Molecular Science
- National Chiao Tung University
- Hsinchu 300
- Taiwan
| | - Zhen-Yi Hong
- Department of Applied Chemistry
- Institute of Molecular Science
- National Chiao Tung University
- Hsinchu 300
- Taiwan
| | - Yaw-Kuen Li
- Department of Applied Chemistry
- Institute of Molecular Science
- National Chiao Tung University
- Hsinchu 300
- Taiwan
| | - Ian Liau
- Department of Applied Chemistry
- Institute of Molecular Science
- National Chiao Tung University
- Hsinchu 300
- Taiwan
| |
Collapse
|
50
|
Farshi P, Fyk-Kolodziej B, Krolewski DM, Walker PD, Ichinose T. Dopamine D1 receptor expression is bipolar cell type-specific in the mouse retina. J Comp Neurol 2015; 524:2059-79. [PMID: 26587737 DOI: 10.1002/cne.23932] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 11/16/2015] [Accepted: 11/17/2015] [Indexed: 01/25/2023]
Abstract
In the retina, dopamine is a key molecule for daytime vision. Dopamine is released by retinal dopaminergic amacrine cells and transmits signaling either by conventional synaptic or by volume transmission. By means of volume transmission, dopamine modulates all layers of retinal neurons; however, it is not well understood how dopamine modulates visual signaling pathways in bipolar cells. Here we analyzed Drd1a-tdTomato BAC transgenic mice and found that the dopamine D1 receptor (D1R) is expressed in retinal bipolar cells in a type-dependent manner. Strong tdTomato fluorescence was detected in the inner nuclear layer and localized to type 1, 3b, and 4 OFF bipolar cells and type 5-2, XBC, 6, and 7 ON bipolar cells. In contrast, type 2, 3a, 5-1, 9, and rod bipolar cells did not express Drd1a-tdTomato. Other interneurons were also found to express tdTomato including horizontal cells and a subset (25%) of AII amacrine cells. Diverse visual processing pathways, such as color or motion-coded pathways, are thought to be initiated in retinal bipolar cells. Our results indicate that dopamine sculpts bipolar cell performance in a type-dependent manner to facilitate daytime vision. J. Comp. Neurol. 524:2059-2079, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Pershang Farshi
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Bozena Fyk-Kolodziej
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - David M Krolewski
- Molecular and Behavioral Neuroscience Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Paul D Walker
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Tomomi Ichinose
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Ophthalmology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|