1
|
MacIsaac AR, Wellington AJ, Filicetti K, Eggers ED. Impaired dopamine signaling in early diabetic retina: Insights from D1R and D4R agonist effects on whole retina responses. Exp Eye Res 2024; 247:110049. [PMID: 39151774 PMCID: PMC11392630 DOI: 10.1016/j.exer.2024.110049] [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: 06/07/2024] [Revised: 07/23/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
The retina has low dopamine levels early in diabetes. To determine how low dopamine levels affected dopamine signaling, the effects of dopamine receptor agonists and mRNA localization were measured after 6 weeks of diabetes. Whole retina ex vivo electroretinogram (ERG) recordings were used to analyze how dopamine type 1 receptor (D1R) and type 4 (D4R) agonists change the light-evoked retinal responses of non-diabetic and 6-week diabetic (STZ injected) mouse retinas. Fluorescence in situ hybridization was utilized to analyze D4R and D1R mRNA locations and expression levels. D4R activation reduced A- and B-wave ERG amplitudes and increased B-wave implicit time and rise-time in the non-diabetic group without a corresponding change in the diabetic group. D1R activation increased B-wave rise-time and oscillatory potential peak time in the non-diabetic group also with no change in the diabetic group. The lack of responsivity to D1R or D4R agonists shows an impairment of dopamine signaling in the diabetic retina. D4R mRNA was found primarily in the outer nuclear layer where photoreceptor cell bodies reside. D1R mRNA was found in the inner nuclear layer and ganglion cell layer that contain bipolar, amacrine, horizontal and ganglion cells. There was no change in D4R or D1R mRNA expression between the non-diabetic and diabetic retinas. This suggests that the significant dopamine signaling changes observed were not from lower receptor expression levels but could be due to changes in dopamine receptor activity or protein levels. These studies show that changes in retinal dopamine signaling could be an important mechanism of diabetic retinal dysfunction.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Diabetes Mellitus, Experimental/metabolism
- Diabetic Retinopathy/metabolism
- Dopamine/metabolism
- Dopamine Agonists/pharmacology
- Electroretinography
- In Situ Hybridization, Fluorescence
- Mice, Inbred C57BL
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D1/agonists
- Receptors, Dopamine D4/metabolism
- Receptors, Dopamine D4/genetics
- Receptors, Dopamine D4/agonists
- Retina/metabolism
- Retina/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Angela R MacIsaac
- Department of Physiology, University of Arizona, Tucson, AZ, USA; Department of Biomedical Engineering, University of Arizona, Tucson, AZ, USA.
| | - Andrea J Wellington
- Department of Physiology, University of Arizona, Tucson, AZ, USA; Department of Biomedical Engineering, University of Arizona, Tucson, AZ, USA.
| | - Kyle Filicetti
- Department of Physiology, University of Arizona, Tucson, AZ, USA; Department of Biomedical Engineering, University of Arizona, Tucson, AZ, USA.
| | - Erika D Eggers
- Department of Physiology, University of Arizona, Tucson, AZ, USA; Department of Biomedical Engineering, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
2
|
Swygart D, Yu WQ, Takeuchi S, Wong ROL, Schwartz GW. A presynaptic source drives differing levels of surround suppression in two mouse retinal ganglion cell types. Nat Commun 2024; 15:599. [PMID: 38238324 PMCID: PMC10796971 DOI: 10.1038/s41467-024-44851-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 01/05/2024] [Indexed: 01/22/2024] Open
Abstract
In early sensory systems, cell-type diversity generally increases from the periphery into the brain, resulting in a greater heterogeneity of responses to the same stimuli. Surround suppression is a canonical visual computation that begins within the retina and is found at varying levels across retinal ganglion cell types. Our results show that heterogeneity in the level of surround suppression occurs subcellularly at bipolar cell synapses. Using single-cell electrophysiology and serial block-face scanning electron microscopy, we show that two retinal ganglion cell types exhibit very different levels of surround suppression even though they receive input from the same bipolar cell types. This divergence of the bipolar cell signal occurs through synapse-specific regulation by amacrine cells at the scale of tens of microns. These findings indicate that each synapse of a single bipolar cell can carry a unique visual signal, expanding the number of possible functional channels at the earliest stages of visual processing.
Collapse
Affiliation(s)
- David Swygart
- Northwestern University Interdepartmental Neuroscience Program, Chicago, IL, USA
| | - Wan-Qing Yu
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Shunsuke Takeuchi
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Rachel O L Wong
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Gregory W Schwartz
- Northwestern University Interdepartmental Neuroscience Program, Chicago, IL, USA.
- Departments of Ophthalmology and Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
3
|
Abstract
Although diabetic retinopathy (DR) is clinically diagnosed as a vascular disease, many studies find retinal neuronal and visual dysfunction before the onset of vascular DR. This suggests that DR should be viewed as a neurovascular disease. Prior to the onset of DR, human patients have compromised electroretinograms that indicate a disruption of normal function, particularly in the inner retina. They also exhibit reduced contrast sensitivity. These early changes, especially those due to dysfunction in the inner retina, are also seen in rodent models of diabetes in the early stages of the disease. Rodent models of diabetes exhibit several neuronal mechanisms, such as reduced evoked GABA release, increased excitatory glutamate signaling, and reduced dopamine signaling, that suggest specific neuronal deficits. This suggests that understanding neuronal deficits may lead to early diabetes treatments to ameliorate neuronal dysfunction.
Collapse
Affiliation(s)
- Erika D Eggers
- Departments of Physiology and Biomedical Engineering, University of Arizona, Tucson, Arizona, USA;
| |
Collapse
|
4
|
Yin N, Wang HN, Ding WW, Zhou H, Li SY, Miao Y, Li F, Lei B, Wang Z. Dopamine receptor-mediated roles on retinal ganglion cell hyperexcitability and injury in experimental glaucoma. Cell Signal 2023:110781. [PMID: 37354963 DOI: 10.1016/j.cellsig.2023.110781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023]
Abstract
Extraordinary excitability (hyperexcitability) is closely related to retinal ganglion cell (RGC) injury in glaucoma. Dopamine (DA) and its receptors are involved in modulating RGC excitability. We investigated how DA system affects RGC injury in chronic ocular hypertension (COH) experimental glaucoma model. Western blotting and immunohistochemistry results revealed that expression of DA D2-like receptor (D2R) in RGCs was increased in COH retinas. Patch-clamp recordings showed that outward K+ currents were downregulated, while Na+ currents and NaV1.6 expression were upregulated in RGCs of COH retinas, which could be reversed by intravitreal pre-injection of the D2R antagonist sulpiride, but not by the D1-like receptor (D1R) antagonist SCH23390. However, pre-injection of the D1R agonist SKF81297 could partially reverse the increased expression of NaV1.6 proteins. Consistently, the numbers of evoked action potentials induced by current injections were increased in RGCs of COH retinas, indicating that RGCs may be in a condition of hyperexcitability. The increased frequency of evoked action potentials could be partially block by pre-injection of sulpiride, SKF81297 or DA, respectively. Furthermore, the increased number of TUNEL-positive RGCs in COH retinas could be partially reduced by intravitreal pre-injection of sulpiride, but not by pre-injection of SCH23390. Moreover, pre-injection of SKF81297 or DA could reduce the number of TUNEL-positive RGCs in COH retinas. All these results indicate that in COH retina, activation of D2R enhances RGC hyperexcitability and injury, while activation of D1R results in the opposite effects. Selective inhibition of D2R or activation of D1R may be an effective strategy for treatment of glaucoma.
Collapse
Affiliation(s)
- Ning Yin
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Hong-Ning Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Wen-Wen Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Han Zhou
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Shu-Ying Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yanying Miao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Fang Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Bo Lei
- Institutes of Neuroscience and Third Affiliated Hospital, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450003, China
| | - Zhongfeng Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
5
|
Zhang C, Zhu Z, Zhao J, Li Y, Zhang Z, Zheng Y. Ubiquitous light-emitting diodes: Potential threats to retinal circadian rhythms and refractive development. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 862:160809. [PMID: 36502986 DOI: 10.1016/j.scitotenv.2022.160809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
The use of light-emitting diodes (LEDs) has increased considerably in the 21st century with humans living in a modern photoperiod with brighter nights and dimmer days. Prolonged exposure to LEDs, especially at night, is considered a new source of pollution because it may affect the synthesis and secretion of retinal melatonin and dopamine, resulting in negative impacts on retinal circadian clocks and potentially disrupting retinal circadian rhythms. The control of ocular refraction is believed to be related to retinal circadian rhythms. Moreover, the global prevalence of myopia has increased at an alarming rate in recent decades. The widespread use of LEDs and the rapid increase in the prevalence of myopia overlap, which is unlikely to be a coincidence. The connection among LEDs, retinal circadian rhythms, and refractive development is both fascinating and confusing. In this review, we aim to develop a systematic framework that includes LEDs, retinal circadian rhythms and refractive development. This paper summarizes the possible mechanisms by which LEDs may disrupt retinal circadian rhythms. We propose that prolonged exposure to LEDs may induce myopia by disrupting retinal circadian rhythms. Finally, we suggest several possible countermeasures to prevent LED interference on retinal circadian rhythms, with the hope of reducing the onset and progression of myopia.
Collapse
Affiliation(s)
- Chenchen Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Zhe Zhu
- Department of Ophthalmology, Eye Hospital of Shandong First Medical University, Shandong Eye Institute, Jinan 250000, China
| | - Jing Zhao
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Yanxia Li
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Zhaoying Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Yajuan Zheng
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
6
|
The Role of Atropine in Preventing Myopia Progression: An Update. Pharmaceutics 2022; 14:pharmaceutics14050900. [PMID: 35631486 PMCID: PMC9147984 DOI: 10.3390/pharmaceutics14050900] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 12/24/2022] Open
Abstract
Several approaches have been investigated for preventing myopia progression in children and teenagers. Among them, topical atropine has shown promising results and it is being adopted in clinical practice more and more frequently. However, the optimal formulation and treatment algorithm are still to be determined. We discuss the pharmacokinetic, pharmacodynamic, clinical, and tolerability profile revealed first by the multicenter, randomized ATOM 1 and 2 trials and, more recently, by the LAMP Study. Results from these trials confirmed the efficacy of low-concentration atropine with a concentration-dependent response. Although atropine at 0.025% and 0.05% concentrations has shown the most encouraging results in large-scale studies, these formulations are not yet commonplace in worldwide clinical practice. Moreover, their rebound effect and the possibility of reaching a stabilization effect have not been fully investigated with real-life studies. Thus, further larger-scale studies should better characterize the clinical efficacy of atropine over longer follow-up periods, in order to define the optimal dosage and treatment regimen.
Collapse
|
7
|
Adámek P, Langová V, Horáček J. Early-stage visual perception impairment in schizophrenia, bottom-up and back again. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2022; 8:27. [PMID: 35314712 PMCID: PMC8938488 DOI: 10.1038/s41537-022-00237-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 02/17/2022] [Indexed: 01/01/2023]
Abstract
Visual perception is one of the basic tools for exploring the world. However, in schizophrenia, this modality is disrupted. So far, there has been no clear answer as to whether the disruption occurs primarily within the brain or in the precortical areas of visual perception (the retina, visual pathways, and lateral geniculate nucleus [LGN]). A web-based comprehensive search of peer-reviewed journals was conducted based on various keyword combinations including schizophrenia, saliency, visual cognition, visual pathways, retina, and LGN. Articles were chosen with respect to topic relevance. Searched databases included Google Scholar, PubMed, and Web of Science. This review describes the precortical circuit and the key changes in biochemistry and pathophysiology that affect the creation and characteristics of the retinal signal as well as its subsequent modulation and processing in other parts of this circuit. Changes in the characteristics of the signal and the misinterpretation of visual stimuli associated with them may, as a result, contribute to the development of schizophrenic disease.
Collapse
Affiliation(s)
- Petr Adámek
- Third Faculty of Medicine, Charles University, Prague, Czech Republic. .,Center for Advanced Studies of Brain and Consciousness, National Institute of Mental Health, Klecany, Czech Republic.
| | - Veronika Langová
- Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Center for Advanced Studies of Brain and Consciousness, National Institute of Mental Health, Klecany, Czech Republic
| | - Jiří Horáček
- Third Faculty of Medicine, Charles University, Prague, Czech Republic.,Center for Advanced Studies of Brain and Consciousness, National Institute of Mental Health, Klecany, Czech Republic
| |
Collapse
|
8
|
Flood MD, Wellington AJ, Eggers ED. Impaired Light Adaptation of ON-Sustained Ganglion Cells in Early Diabetes Is Attributable to Diminished Response to Dopamine D4 Receptor Activation. Invest Ophthalmol Vis Sci 2022; 63:33. [PMID: 35077550 PMCID: PMC8802033 DOI: 10.1167/iovs.63.1.33] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Retinal neuronal signaling is disrupted early in diabetes, before the onset of the vascular pathologies associated with diabetic retinopathy. There is also growing evidence that retinal dopamine, a neuromodulator that mediates light adaptation, is reduced in early diabetes. Previously, we have shown that after 6 weeks of diabetes, light adaptation is impaired in ON-sustained (ON-s) ganglion cells in the mouse retina. The purpose of this study was to determine whether changes in the response to dopamine receptor activation contribute to this dysfunction. Methods Single-cell retinal patch-clamp recordings from the mouse retina were used to determine how activating dopamine type D4 receptors (D4Rs) changes the light-evoked and spontaneous excitatory inputs to ON-s ganglion cells, in both control and 6-week diabetic (STZ-injected) animals. Fluorescence in situ hybridization was also used to assess whether D4R expression was affected by diabetes. Results D4R activation decreased light-evoked and spontaneous inputs to ON-s ganglion cells in control and diabetic retinas. However, D4R activation caused a smaller reduction in light-evoked excitatory inputs to ON-s ganglion cells in diabetic retinas compared to controls. This impaired D4R signaling is not attributable to a decline in D4R expression, as there was no change in D4R mRNA density in the diabetic retinas. Conclusions These results suggest that the cellular response to dopamine signaling is disrupted in early diabetes and may be amenable to chronic dopamine supplementation therapy.
Collapse
Affiliation(s)
- Michael D Flood
- Departments of Physiology and Biomedical Engineering, University of Arizona, Tucson, Arizona, United States
| | - Andrea J Wellington
- Departments of Physiology and Biomedical Engineering, University of Arizona, Tucson, Arizona, United States
| | - Erika D Eggers
- Departments of Physiology and Biomedical Engineering, University of Arizona, Tucson, Arizona, United States
| |
Collapse
|
9
|
Flood MD, Eggers ED. Dopamine D1 and D4 receptors contribute to light adaptation in ON-sustained retinal ganglion cells. J Neurophysiol 2021; 126:2039-2052. [PMID: 34817291 PMCID: PMC8715048 DOI: 10.1152/jn.00218.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/08/2021] [Accepted: 11/19/2021] [Indexed: 01/21/2023] Open
Abstract
The adaptation of ganglion cells to increasing light levels is a crucial property of the retina. The retina must respond to light intensities that vary by 10-12 orders of magnitude, but the dynamic range of ganglion cell responses covers only ∼3 orders of magnitude. Dopamine is a crucial neuromodulator for light adaptation and activates receptors in the D1 and D2 families. Dopamine type D1 receptors (D1Rs) are expressed on horizontal cells and some bipolar, amacrine, and ganglion cells. In the D2 family, D2Rs are expressed on dopaminergic amacrine cells and D4Rs are primarily expressed on photoreceptors. However, the roles of activating these receptors to modulate the synaptic properties of the inputs to ganglion cells are not yet clear. Here, we used single-cell retinal patch-clamp recordings from the mouse retina to determine how activating D1Rs and D4Rs changed the light-evoked and spontaneous excitatory inputs to ON-sustained (ON-s) ganglion cells. We found that both D1R and D4R activation decrease the light-evoked excitatory inputs to ON-s ganglion cells, but that only the sum of the peak response decrease due to activating the two receptors was similar to the effect of light adaptation to a rod-saturating background. The largest effects on spontaneous excitatory activity of both D1R and D4R agonists was on the frequency of events, suggesting that both D1Rs and D4Rs are acting upstream of the ganglion cells.NEW & NOTEWORTHY Dopamine by bright light conditions allows retinal neurons to reduce sensitivity to adapt to bright light conditions. It is not clear how and why dopamine receptors modulate retinal ganglion cell signaling. We found that both D1 and D4 dopamine receptors in photoreceptors and inner retinal neurons contribute significantly to the reduction in sensitivity of ganglion cells with light adaptation. However, light adaptation also requires dopamine-independent mechanisms that could reflect inherent sensitivity changes in photoreceptors.
Collapse
Affiliation(s)
- Michael D Flood
- Department of Physiology, University of Arizona, Tucson, Arizona
- Department Biomedical Engineering, University of Arizona, Tucson, Arizona
| | - Erika D Eggers
- Department of Physiology, University of Arizona, Tucson, Arizona
- Department Biomedical Engineering, University of Arizona, Tucson, Arizona
| |
Collapse
|
10
|
Glycine Release Is Potentiated by cAMP via EPAC2 and Ca 2+ Stores in a Retinal Interneuron. J Neurosci 2021; 41:9503-9520. [PMID: 34620721 PMCID: PMC8612479 DOI: 10.1523/jneurosci.0670-21.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 11/21/2022] Open
Abstract
Neuromodulation via the intracellular second messenger cAMP is ubiquitous at presynaptic nerve terminals. This modulation of synaptic transmission allows exocytosis to adapt to stimulus levels and reliably encode information. The AII amacrine cell (AII-AC) is a central hub for signal processing in the mammalian retina. The main apical dendrite of the AII-AC is connected to several lobular appendages that release glycine onto OFF cone bipolar cells and ganglion cells. However, the influence of cAMP on glycine release is not well understood. Using membrane capacitance measurements from mouse AII-ACs to directly measure exocytosis, we observe that intracellular dialysis of 1 mm cAMP enhances exocytosis without affecting the L-type Ca2+ current. Responses to depolarizing pulses of various durations show that the size of the readily releasable pool of vesicles nearly doubles with cAMP, while paired-pulse depression experiments suggest that release probability does not change. Specific agonists and antagonists for exchange protein activated by cAMP 2 (EPAC2) revealed that the cAMP-induced enhancement of exocytosis requires EPAC2 activation. Furthermore, intact Ca2+ stores were also necessary for the cAMP potentiation of exocytosis. Postsynaptic recordings from OFF cone bipolar cells showed that increasing cAMP with forskolin potentiated the frequency of glycinergic spontaneous IPSCs. We propose that cAMP elevations in the AII-AC lead to a robust enhancement of glycine release through an EPAC2 and Ca2+ store signaling pathway. Our results thus contribute to a better understanding of how AII-AC crossover inhibitory circuits adapt to changes in ambient luminance.SIGNIFICANCE STATEMENT The mammalian retina operates over a wide dynamic range of light intensities and contrast levels. To optimize the signal-to-noise ratio of processed visual information, both excitatory and inhibitory synapses within the retina must modulate their gain in synaptic transmission to adapt to different levels of ambient light. Here we show that increases of cAMP concentration within AII amacrine cells produce enhanced exocytosis from these glycinergic interneurons. Therefore, we propose that light-sensitive neuromodulators may change the output of glycine release from AII amacrine cells. This novel mechanism may fine-tune the amount of tonic and phasic synaptic inhibition received by bipolar cell terminals and, consequently, the spiking patterns that ganglion cells send to the upstream visual areas of the brain.
Collapse
|
11
|
Zhong L, Gleason EL. Adenylate Cyclase 1 Links Calcium Signaling to CFTR-Dependent Cytosolic Chloride Elevations in Chick Amacrine Cells. Front Cell Neurosci 2021; 15:726605. [PMID: 34456687 PMCID: PMC8385318 DOI: 10.3389/fncel.2021.726605] [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: 06/17/2021] [Accepted: 07/19/2021] [Indexed: 01/03/2023] Open
Abstract
The strength and sign of synapses involving ionotropic GABA and glycine receptors are dependent upon the Cl- gradient. We have shown that nitric oxide (NO) elicits the release of Cl- from internal acidic stores in retinal amacrine cells (ACs); temporarily altering the Cl- gradient and the strength or even sign of incoming GABAergic or glycinergic synapses. The underlying mechanism for this effect of NO requires the cystic fibrosis transmembrane regulator (CFTR) but the link between NO and CFTR activation has not been determined. Here, we test the hypothesis that NO-dependent Ca2+ elevations activate the Ca2+-dependent adenylate cyclase 1 (AdC1) leading to activation of protein kinase A (PKA) whose activity is known to open the CFTR channel. Using the reversal potential of GABA-gated currents to monitor cytosolic Cl-, we established the requirement for Ca2+ elevations. Inhibitors of AdC1 suppressed the NO-dependent increases in cytosolic Cl- whereas inhibitors of other AdC subtypes were ineffective suggesting that AdC1 is involved. Inhibition of PKA also suppressed the action of NO. To address the sufficiency of this pathway in linking NO to elevations in cytosolic Cl-, GABA-gated currents were measured under internal and external zero Cl- conditions to isolate the internal Cl- store. Activators of the cAMP pathway were less effective than NO in producing GABA-gated currents. However, coupling the cAMP pathway activators with the release of Ca2+ from stores produced GABA-gated currents indistinguishable from those stimulated with NO. Together, these results demonstrate that cytosolic Ca2+ links NO to the activation of CFTR and the elevation of cytosolic Cl-.
Collapse
Affiliation(s)
- Li Zhong
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
| | - Evanna L Gleason
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
12
|
Wang WY, Chen C, Chang J, Chien L, Shih YF, Lin LLK, Pang CP, Wang IJ. Pharmacotherapeutic candidates for myopia: A review. Biomed Pharmacother 2021; 133:111092. [PMID: 33378986 DOI: 10.1016/j.biopha.2020.111092] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/24/2020] [Accepted: 11/28/2020] [Indexed: 01/11/2023] Open
Abstract
This review provides insights into the mechanism underlying the pathogenesis of myopia and potential targets for clinical intervention. Although the etiology of myopia involves both environmental and genetic factors, recent evidence has suggested that the prevalence and severity of myopia appears to be affected more by environmental factors. Current pharmacotherapeutics are aimed at inhibiting environmentally induced changes in visual input and subsequent changes in signaling pathways during myopia pathogenesis and progression. Recent studies on animal models of myopia have revealed specific molecules potentially involved in the regulation of eye development. Among them, the dopamine receptor plays a critical role in controlling myopia. Subsequent studies have reported pharmacotherapeutic treatments to control myopia progression. In particular, atropine treatment yielded favorable outcomes and has been extensively used; however, current studies are aimed at optimizing its efficacy and confirming its safety. Furthermore, future studies are required to assess the efficacy of combinatorial use of low-dose atropine and contact lenses or orthokeratology.
Collapse
Affiliation(s)
- Wen-Yi Wang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Camille Chen
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Justine Chang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Lillian Chien
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yung-Feng Shih
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Luke L K Lin
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, Hong Kong Eye Hospital, 147K Argyle Street, KLN, Hong Kong, China.
| | - I-Jong Wang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
13
|
Indrieri A, Pizzarelli R, Franco B, De Leonibus E. Dopamine, Alpha-Synuclein, and Mitochondrial Dysfunctions in Parkinsonian Eyes. Front Neurosci 2020; 14:567129. [PMID: 33192254 PMCID: PMC7604532 DOI: 10.3389/fnins.2020.567129] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/22/2020] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by motor dysfunctions including bradykinesia, tremor at rest and motor instability. These symptoms are associated with the progressive degeneration of dopaminergic neurons originating in the substantia nigra pars compacta and projecting to the corpus striatum, and by accumulation of cytoplasmic inclusions mainly consisting of aggregated alpha-synuclein, called Lewy bodies. PD is a complex, multifactorial disorder and its pathogenesis involves multiple pathways and mechanisms such as α-synuclein proteostasis, mitochondrial function, oxidative stress, calcium homeostasis, axonal transport, and neuroinflammation. Motor symptoms manifest when there is already an extensive dopamine denervation. There is therefore an urgent need for early biomarkers to apply disease-modifying therapeutic strategies. Visual defects and retinal abnormalities, including decreased visual acuity, abnormal spatial contrast sensitivity, color vision defects, or deficits in more complex visual tasks are present in the majority of PD patients. They are being considered for early diagnosis together with retinal imaging techniques are being considered as non-invasive biomarkers for PD. Dopaminergic cells can be found in the retina in a subpopulation of amacrine cells; however, the molecular mechanisms leading to visual deficits observed in PD patients are still largely unknown. This review provides a comprehensive analysis of the retinal abnormalities observed in PD patients and animal models and of the molecular mechanisms underlying neurodegeneration in parkinsonian eyes. We will review the role of α-synuclein aggregates in the retina pathology and/or in the onset of visual symptoms in PD suggesting that α-synuclein aggregates are harmful for the retina as well as for the brain. Moreover, we will summarize experimental evidence suggesting that the optic nerve pathology observed in PD resembles that seen in mitochondrial optic neuropathies highlighting the possible involvement of mitochondrial abnormalities in the development of PD visual defects. We finally propose that the eye may be considered as a complementary experimental model to identify possible novel disease’ pathways or to test novel therapeutic approaches for PD.
Collapse
Affiliation(s)
- Alessia Indrieri
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Institute for Genetic and Biomedical Research, National Research Council, Milan, Italy
| | - Rocco Pizzarelli
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy
| | - Brunella Franco
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Medical Genetics, Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.,Institute of Biochemistry and Cellular Biology, National Research Council, Rome, Italy
| |
Collapse
|
14
|
Flood MD, Wellington AJ, Cruz LA, Eggers ED. Early diabetes impairs ON sustained ganglion cell light responses and adaptation without cell death or dopamine insensitivity. Exp Eye Res 2020; 200:108223. [PMID: 32910942 DOI: 10.1016/j.exer.2020.108223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/17/2020] [Accepted: 09/03/2020] [Indexed: 10/23/2022]
Abstract
Retinal signaling under dark-adapted conditions is perturbed during early diabetes. Additionally, dopamine, the main neuromodulator of retinal light adaptation, is diminished in diabetic retinas. However, it is not known if this dopamine deficiency changes how the retina responds to increased light or dopamine. Here we determine whether light adaptation is impaired in the diabetic retina, and investigate potential mechanism(s) of impairment. Diabetes was induced in C57BL/6J male mice via 3 intraperitoneal injections of streptozotocin (75 mg/kg) and confirmed by blood glucose levels more than 200 mg/dL. After 6 weeks, whole-cell recordings of light-evoked and spontaneous inhibitory postsynaptic currents (IPSCs) or excitatory postsynaptic currents (EPSCs) were made from rod bipolar cells and ON sustained ganglion cells, respectively. Light responses were recorded before and after D1 receptor (D1R) activation (SKF-38393, 20 μM) or light adaptation (background of 950 photons·μm-2 ·s-1). Retinal whole mounts were stained for either tyrosine hydroxylase and activated caspase-3 or GAD65/67, GlyT1 and RBPMS and imaged. D1R activation and light adaptation both decreased inhibition, but the disinhibition was not different between control and diabetic rod bipolar cells. However, diabetic ganglion cell light-evoked EPSCs were increased in the dark and showed reduced light adaptation. No differences were found in light adaptation of spontaneous EPSC parameters, suggesting upstream changes. No changes in cell density were found for dopaminergic, glycinergic or GABAergic amacrine cells, or ganglion cells. Thus, in early diabetes, ON sustained ganglion cells receive excessive excitation under dark- and light-adapted conditions. Our results show that this is not attributable to loss in number or dopamine sensitivity of inhibitory amacrine cells or loss of dopaminergic amacrine cells.
Collapse
Affiliation(s)
- Michael D Flood
- Departments of Physiology and Biomedical Engineering, P.O. Box 245051, University of Arizona, Tucson, AZ, 85724, USA.
| | - Andrea J Wellington
- Departments of Physiology and Biomedical Engineering, P.O. Box 245051, University of Arizona, Tucson, AZ, 85724, USA.
| | - Luis A Cruz
- Departments of Physiology and Biomedical Engineering, P.O. Box 245051, University of Arizona, Tucson, AZ, 85724, USA.
| | - Erika D Eggers
- Departments of Physiology and Biomedical Engineering, P.O. Box 245051, University of Arizona, Tucson, AZ, 85724, USA.
| |
Collapse
|
15
|
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: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
16
|
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: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
17
|
Inhibitory components of retinal bipolar cell receptive fields are differentially modulated by dopamine D1 receptors. Vis Neurosci 2020; 37:E01. [PMID: 32046810 DOI: 10.1017/s0952523819000129] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
During adaptation to an increase in environmental luminance, retinal signaling adjustments are mediated by the neuromodulator dopamine. Retinal dopamine is released with light and can affect center-surround receptive fields, the coupling state between neurons, and inhibitory pathways through inhibitory receptors and neurotransmitter release. While the inhibitory receptive field surround of bipolar cells becomes narrower and weaker during light adaptation, it is unknown how dopamine affects bipolar cell surrounds. If dopamine and light have similar effects, it would suggest that dopamine could be a mechanism for light-adapted changes. We tested the hypothesis that dopamine D1 receptor activation is sufficient to elicit the magnitude of light-adapted reductions in inhibitory bipolar cell surrounds. Surrounds were measured from OFF bipolar cells in dark-adapted mouse retinas while stimulating D1 receptors, which are located on bipolar, horizontal, and inhibitory amacrine cells. The D1 agonist SKF-38393 narrowed and weakened OFF bipolar cell inhibitory receptive fields but not to the same extent as with light adaptation. However, the receptive field surround reductions differed between the glycinergic and GABAergic components of the receptive field. GABAergic inhibitory strength was reduced only at the edges of the surround, while glycinergic inhibitory strength was reduced across the whole receptive field. These results expand the role of retinal dopamine to include modulation of bipolar cell receptive field surrounds. Additionally, our results suggest that D1 receptor pathways may be a mechanism for the light-adapted weakening of glycinergic surround inputs and the furthest wide-field GABAergic inputs to bipolar cells. However, remaining differences between light-adapted and D1 receptor-activated inhibition demonstrate that non-D1 receptor mechanisms are necessary to elicit the full effect of light adaptation on inhibitory surrounds.
Collapse
|
18
|
Kovács-Öller T, Szarka G, Ganczer A, Tengölics Á, Balogh B, Völgyi B. Expression of Ca 2+-Binding Buffer Proteins in the Human and Mouse Retinal Neurons. Int J Mol Sci 2019; 20:E2229. [PMID: 31067641 PMCID: PMC6539911 DOI: 10.3390/ijms20092229] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/31/2022] Open
Abstract
Ca2+-binding buffer proteins (CaBPs) are widely expressed by various neurons throughout the central nervous system (CNS), including the retina. While the expression of CaBPs by photoreceptors, retinal interneurons and the output ganglion cells in the mammalian retina has been extensively studied, a general description is still missing due to the differences between species, developmental expression patterns and study-to-study discrepancies. Furthermore, CaBPs are occasionally located in a compartment-specific manner and two or more CaBPs can be expressed by the same neuron, thereby sharing the labor of Ca2+ buffering in the intracellular milieu. This article reviews this topic by providing a framework on CaBP functional expression by neurons of the mammalian retina with an emphasis on human and mouse retinas and the three most abundant and extensively studied buffer proteins: parvalbumin, calretinin and calbindin.
Collapse
Affiliation(s)
- Tamás Kovács-Öller
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
- Medical School, University of Pécs, 7624 Pécs, Hungary.
| | - Gergely Szarka
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Alma Ganczer
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Ádám Tengölics
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Boglárka Balogh
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
| | - Béla Völgyi
- János Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary.
- Retinal Electrical Synapses Research Group, National Brain Research Program (NAP 2.0), Hungarian Academy of Sciences, 1051 Budapest, Hungary.
- Department of Experimental Zoology and Neurobiology, University of Pécs, 7624 Pécs, Hungary.
- Medical School, University of Pécs, 7624 Pécs, Hungary.
| |
Collapse
|
19
|
Yadav SC, Tetenborg S, Dedek K. Gap Junctions in A8 Amacrine Cells Are Made of Connexin36 but Are Differently Regulated Than Gap Junctions in AII Amacrine Cells. Front Mol Neurosci 2019; 12:99. [PMID: 31065239 PMCID: PMC6489437 DOI: 10.3389/fnmol.2019.00099] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/03/2019] [Indexed: 01/01/2023] Open
Abstract
In the mammalian retina, amacrine cells represent the most diverse cell class and are involved in the spatio-temporal processing of visual signals in the inner plexiform layer. They are connected to bipolar, other amacrine and ganglion cells, forming complex networks via electrical and chemical synapses. The small-field A8 amacrine cell was shown to receive non-selective glutamatergic input from OFF and ON cone bipolar cells at its bistratified dendrites in sublamina 1 and 4 of the inner plexiform layer. Interestingly, it was also shown to form electrical synapses with ON cone bipolar cells, thus resembling the rod pathway-specific AII amacrine cell. In contrast to the AII cell, however, the electrical synapses of A8 cells are poorly understood. Therefore, we made use of the Ier5-GFP mouse line, in which A8 cells are labeled by GFP, to study the gap junction composition and frequency in A8 cells. We found that A8 cells form <20 gap junctions per cell and these gap junctions consist of connexin36. Connexin36 is present at both OFF and ON dendrites of A8 cells, preferentially connecting A8 cells to type 1 OFF and type 6 and 7 ON bipolar cells and presumably other amacrine cells. Additionally, we show that the OFF dendrites of A8 cells co-stratify with the processes of dopaminergic amacrine cells from which they may receive GABAergic input via GABAA receptor subunit α3. As we found A8 cells to express dopamine receptor D1 (but not D2), we also tested whether A8 cell coupling is modulated by D1 receptor agonists and antagonists as was shown for the coupling of AII cells. However, this was not the case. In summary, our data suggests that A8 coupling is differently regulated than AII cells and may even be independent of ambient light levels and serve signal facilitation rather than providing a separate neuronal pathway.
Collapse
Affiliation(s)
- Shubhash C Yadav
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Stephan Tetenborg
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany
| | - Karin Dedek
- Animal Navigation/Neurosensorics, Institute for Biology and Environmental Sciences, University of Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, University of Oldenburg, Oldenburg, Germany
| |
Collapse
|
20
|
Mazade RE, Flood MD, Eggers ED. Dopamine D1 receptor activation reduces local inner retinal inhibition to light-adapted levels. J Neurophysiol 2019; 121:1232-1243. [PMID: 30726156 PMCID: PMC6485729 DOI: 10.1152/jn.00448.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 01/26/2023] Open
Abstract
During adaptation from dim to bright environments, changes in retinal signaling are mediated, in part, by dopamine. Dopamine is released with light and can modulate retinal receptive fields, neuronal coupling, inhibitory receptors, and rod pathway inhibition. However, it is unclear how dopamine affects inner retinal inhibition to cone bipolar cells, which relay visual information from photoreceptors to ganglion cells and are important signal processing sites. We tested the hypothesis that dopamine (D)1 receptor activation is sufficient to elicit light-adapted inhibitory changes. Local light-evoked inhibition and spontaneous activity were measured from OFF cone bipolar cells in dark-adapted mouse retinas while stimulating D1 receptors, which are located on bipolar, horizontal, and inhibitory amacrine cells. The D1 agonist SKF38393 reduced local inhibitory light-evoked response magnitude and increased response transience, which mimicked changes measured with light adaptation. D1-mediated reductions in local inhibition were more pronounced for glycinergic than GABAergic inputs, comparable with light adaptation. The effects of D1 receptors on light-evoked input were similar to the effects on spontaneous input. D1 receptor activation primarily decreased glycinergic spontaneous current frequency, similar to light adaptation, suggesting mainly a presynaptic amacrine cell site of action. These results expand the role of dopamine to include signal modulation of cone bipolar cell local inhibition. In this role, D1 receptor activation, acting primarily through glycinergic amacrine cells, may be an important mechanism for the light-adapted reduction in OFF bipolar cell inhibition since the actions are similar and dopamine is released during light adaptation. NEW & NOTEWORTHY Retinal adaptation to different luminance conditions requires the adjustment of local circuits for accurate signaling of visual scenes. Understanding mechanisms behind luminance adaptation at different retinal levels is important for understanding how the retina functions in a dynamic environment. In the mouse, we show that dopamine pathways reduce inner retinal inhibition similar to increased background luminance, suggesting the two are linked and highlighting a possible mechanism for light adaptation at an early retinal processing center.
Collapse
Affiliation(s)
- Reece E Mazade
- Departments of Physiology and Biomedical Engineering, University of Arizona , Tucson, Arizona
| | - Michael D Flood
- Departments of Physiology and Biomedical Engineering, University of Arizona , Tucson, Arizona
| | - Erika D Eggers
- Departments of Physiology and Biomedical Engineering, University of Arizona , Tucson, Arizona
| |
Collapse
|
21
|
Sankaran M, Keeley PW, He L, Iuvone PM, Reese BE. Dopaminergic amacrine cell number, plexus density, and dopamine content in the mouse retina: Strain differences and effects of Bax gene disruption. Exp Eye Res 2018; 177:208-212. [PMID: 30240584 DOI: 10.1016/j.exer.2018.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/07/2018] [Accepted: 09/17/2018] [Indexed: 01/01/2023]
Abstract
Many types of retinal neuron modulate the distribution of their processes to ensure a uniform coverage of the retinal surface. Dendritic field area, for instance, is inversely related to the variation in cellular density for many cell types, observed either across retinal eccentricity or between different strains of mice that differ in cell number. Dopaminergic amacrine (DA) cells, by contrast, have dendritic arbors that bear no spatial relationship to the presence of their immediate homotypic neighbors, yet it remains to be determined whether their coverage upon the retina, as a population, is conserved across variation in their total number. The present study assessed the overall density of the dopaminergic plexus in the inner plexiform layer in the presence of large variation in the total number of DA cells, as well as their retinal dopamine content, to determine whether either of these features is conserved. We first compared these traits between two strains of mice (C57BL/6J and A/J) that exhibit a two-fold difference in DA cell number. We subsequently examined these same traits in littermate mice for which the pro-apoptotic Bax gene was either intact or knocked out, yielding a five-fold difference in DA cell number. In both comparisons, we found greater plexus density and DA content in the strain or condition with the greater number of DA cells. The population of DA cells, therefore, does not appear to self-regulate its process coverage to achieve a constant density as the DA mosaic is established during development, nor its functional dopamine content in maturity.
Collapse
Affiliation(s)
- Mathangi Sankaran
- Neuroscience Research Institute, University of California, Santa Barbara, CA, 93106, USA; Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, 93106, USA
| | - Patrick W Keeley
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, 93106, USA
| | - Li He
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, 30322, USA; Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Benjamin E Reese
- Neuroscience Research Institute, University of California, Santa Barbara, CA, 93106, USA; Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
22
|
Flood MD, Moore-Dotson JM, Eggers ED. Dopamine D1 receptor activation contributes to light-adapted changes in retinal inhibition to rod bipolar cells. J Neurophysiol 2018; 120:867-879. [PMID: 29847232 PMCID: PMC6139461 DOI: 10.1152/jn.00855.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/22/2018] [Accepted: 05/25/2018] [Indexed: 12/27/2022] Open
Abstract
Dopamine modulation of retinal signaling has been shown to be an important part of retinal adaptation to increased background light levels, but the role of dopamine modulation of retinal inhibition is not clear. We previously showed that light adaptation causes a large reduction in inhibition to rod bipolar cells, potentially to match the decrease in excitation after rod saturation. In this study, we determined how dopamine D1 receptors in the inner retina contribute to this modulation. We found that D1 receptor activation significantly decreased the magnitude of inhibitory light responses from rod bipolar cells, whereas D1 receptor blockade during light adaptation partially prevented this decline. To determine what mechanisms were involved in the modulation of inhibitory light responses, we measured the effect of D1 receptor activation on spontaneous currents and currents evoked from electrically stimulating amacrine cell inputs to rod bipolar cells. D1 receptor activation decreased the frequency of spontaneous inhibition with no change in event amplitudes, suggesting a presynaptic change in amacrine cell activity in agreement with previous reports that rod bipolar cells lack D1 receptors. Additionally, we found that D1 receptor activation reduced the amplitude of electrically evoked responses, showing that D1 receptors can modulate amacrine cells directly. Our results suggest that D1 receptor activation can replicate a large portion but not all of the effects of light adaptation, likely by modulating release from amacrine cells onto rod bipolar cells. NEW & NOTEWORTHY We demonstrated a new aspect of dopaminergic signaling that is involved in mediating light adaptation of retinal inhibition. This D1 receptor-dependent mechanism likely acts through receptors located directly on amacrine cells, in addition to its potential role in modulating the strength of serial inhibition between amacrine cells. Our results also suggest that another D2/D4 receptor-dependent or dopamine-independent mechanism must also be involved in light adaptation of inhibition to rod bipolar cells.
Collapse
Affiliation(s)
- Michael D Flood
- Departments of Physiology and Biomedical Engineering, University of Arizona , Tucson, Arizona
| | - Johnnie M Moore-Dotson
- Departments of Physiology and Biomedical Engineering, University of Arizona , Tucson, Arizona
| | - Erika D Eggers
- Departments of Physiology and Biomedical Engineering, University of Arizona , Tucson, Arizona
| |
Collapse
|
23
|
Pardue MT, Allen RS. Neuroprotective strategies for retinal disease. Prog Retin Eye Res 2018; 65:50-76. [PMID: 29481975 PMCID: PMC6081194 DOI: 10.1016/j.preteyeres.2018.02.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/14/2018] [Accepted: 02/20/2018] [Indexed: 12/20/2022]
Abstract
Diseases that affect the eye, including photoreceptor degeneration, diabetic retinopathy, and glaucoma, affect 11.8 million people in the US, resulting in vision loss and blindness. Loss of sight affects patient quality of life and puts an economic burden both on individuals and the greater healthcare system. Despite the urgent need for treatments, few effective options currently exist in the clinic. Here, we review research on promising neuroprotective strategies that promote neuronal survival with the potential to protect against vision loss and retinal cell death. Due to the large number of neuroprotective strategies, we restricted our review to approaches that we had direct experience with in the laboratory. We focus on drugs that target survival pathways, including bile acids like UDCA and TUDCA, steroid hormones like progesterone, therapies that target retinal dopamine, and neurotrophic factors. In addition, we review rehabilitative methods that increase endogenous repair mechanisms, including exercise and electrical stimulation therapies. For each approach, we provide background on the neuroprotective strategy, including history of use in other diseases; describe potential mechanisms of action; review the body of research performed in the retina thus far, both in animals and in humans; and discuss considerations when translating each treatment to the clinic and to the retina, including which therapies show the most promise for each retinal disease. Despite the high incidence of retinal diseases and the complexity of mechanisms involved, several promising neuroprotective treatments provide hope to prevent blindness. We discuss attractive candidates here with the goal of furthering retinal research in critical areas to rapidly translate neuroprotective strategies into the clinic.
Collapse
Affiliation(s)
- Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332, USA.
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA
| |
Collapse
|
24
|
Hoshi H, Sato F. The morphological characterization of orientation-biased displaced large-field ganglion cells in the central part of goldfish retina. J Comp Neurol 2018; 526:243-261. [PMID: 28921532 DOI: 10.1002/cne.24331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 11/10/2022]
Abstract
The vertebrate retina has about 30 subtypes of ganglion cells. Each ganglion cell receives synaptic inputs from specific types of bipolar and amacrine cells ramifying at the same depth of the inner plexiform layer (IPL), each of which is thought to process a specific aspect of visual information. Here, we identified one type of displaced ganglion cell in the goldfish retina which had a large and elongated dendritic field. As a population, all of these ganglion cells were oriented in the horizontal axis and perpendicular to the dorsal-ventral axis of the goldfish eye in the central part of retina. This ganglion cell has previously been classified as Type 1.2. However, the circuit elements which synapse with this ganglion cell are not yet characterized. We found that this displaced ganglion cell was directly tracer-coupled only with homologous ganglion cells at sites containing Cx35/36 puncta. We further illustrated that the processes of dopaminergic neurons often terminated next to intersections between processes of ganglion cells, close to where dopamine D1 receptors were localized. Finally, we showed that Mb1 ON bipolar cells had ribbon synapses in the axonal processes passing through the IPL and made ectopic synapses with this displaced ganglion cell that stratified into stratum 1 of the IPL. These results suggest that the displaced ganglion cell may synapse with both Mb1 cells using ectopic ribbon synapses and OFF cone bipolar cells with regular ribbon synapses in the IPL to function in both scotopic and photopic light conditions.
Collapse
Affiliation(s)
- Hideo Hoshi
- Department of Anatomy, School of Medicine, Toho University, Tokyo, Japan
| | - Fumi Sato
- Department of Anatomy, School of Medicine, Toho University, Tokyo, Japan
| |
Collapse
|
25
|
Chesnokova NB, Pavlenko TA, Ugrumov MV. [Ophthalmic disorders as a manifestation of Parkinson's disease]. Zh Nevrol Psikhiatr Im S S Korsakova 2017; 117:124-131. [PMID: 29053133 DOI: 10.17116/jnevro201711791124-131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Parkinson's disease is a severe neurodegenerative disease accompanied with the degeneration of dopaminergic neurons in the central and peripheral nervous system. The diagnosis of Parkinson's disease can still be made only on the stage of irreversible and nearly total degeneration of the nigrostriatum dopaminergic system and exhaustion of brain compensatory mechanisms that explains the low efficacy of therapy. Ophthalmic pathology is one of the nonmotor symptoms of Parkinson's disease. This can be explained firstly by the fact that eye is a 'peripheral part of brain' and secondly by the involvement of dopaminergic neurons (dopamine-producing cells) that are subject to the selective degeneration during Parkinson's disease in the regulation of visual function in the eye and brain. Dopaminergic neurons and dopamine receptors are present in all structures of the eye. Parkinson's disease cause abnormalities not only in the retina but in the whole optic tract and can be considered as peripheral manifestations of the disease that precede the well-known motor dysfunctions. This review describes ophthalmological symptoms of Parkinson's disease, possible pathophysiological mechanisms of their development, optical disorders in experimental models of Parkinson's disease and also the perspectives of experimental and clinical studies of visual disorders for the development of preclinical diagnosis of Parkinson's disease.
Collapse
Affiliation(s)
- N B Chesnokova
- Helmholtz Moscow Research Institute of Eye Diseases, Moscow, Russia
| | - T A Pavlenko
- Helmholtz Moscow Research Institute of Eye Diseases, Moscow, Russia
| | - M V Ugrumov
- Koltzov Institute of Developmental Biology, Russian of Sciences, Moscow, Russia
| |
Collapse
|
26
|
Cui P, Li XY, Zhao Y, Li Q, Gao F, Li LZ, Yin N, Sun XH, Wang Z. Activation of dopamine D1 receptors enhances the temporal summation and excitability of rat retinal ganglion cells. Neuroscience 2017; 355:71-83. [DOI: 10.1016/j.neuroscience.2017.04.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/24/2017] [Accepted: 04/30/2017] [Indexed: 01/11/2023]
|
27
|
Ward AH, Siegwart JT, Frost MR, Norton TT. Intravitreally-administered dopamine D2-like (and D4), but not D1-like, receptor agonists reduce form-deprivation myopia in tree shrews. Vis Neurosci 2017; 34:E003. [PMID: 28304244 PMCID: PMC5567805 DOI: 10.1017/s0952523816000195] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We examined the effect of intravitreal injections of D1-like and D2-like dopamine receptor agonists and antagonists and D4 receptor drugs on form-deprivation myopia (FDM) in tree shrews, mammals closely related to primates. In eleven groups (n = 7 per group), we measured the amount of FDM produced by monocular form deprivation (FD) over an 11-day treatment period. The untreated fellow eye served as a control. Animals also received daily 5 µL intravitreal injections in the FD eye. The reference group received 0.85% NaCl vehicle. Four groups received a higher, or lower, dose of a D1-like receptor agonist (SKF38393) or antagonist (SCH23390). Four groups received a higher, or lower, dose of a D2-like receptor agonist (quinpirole) or antagonist (spiperone). Two groups received the D4 receptor agonist (PD168077) or antagonist (PD168568). Refractions were measured daily; axial component dimensions were measured on day 1 (before treatment) and day 12. We found that in groups receiving the D1-like receptor agonist or antagonist, the development of FDM and altered ocular component dimensions did not differ from the NaCl group. Groups receiving the D2-like receptor agonist or antagonist at the higher dose developed significantly less FDM and had shorter vitreous chambers than the NaCl group. The D4 receptor agonist, but not the antagonist, was nearly as effective as the D2-like agonist in reducing FDM. Thus, using intravitreally-administered agents, we did not find evidence supporting a role for the D1-like receptor pathway in reducing FDM in tree shrews. The reduction of FDM by the dopamine D2-like agonist supported a role for the D2-like receptor pathway in the control of FDM. The reduction of FDM by the D4 receptor agonist, but not the D4 antagonist, suggests an important role for activation of the dopamine D4 receptor in the control of axial elongation and refractive development.
Collapse
Affiliation(s)
- Alexander H. Ward
- Genetics, Genomics and Bioinformatics Theme, University of Alabama at Birmingham, Birmingham, AL 35294
| | - John T. Siegwart
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Michael R. Frost
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Thomas T. Norton
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
28
|
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: 41] [Impact Index Per Article: 4.6] [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
|
29
|
Li Q, Wu N, Cui P, Gao F, Qian WJ, Miao Y, Sun XH, Wang Z. Suppression of outward K(+) currents by activating dopamine D1 receptors in rat retinal ganglion cells through PKA and CaMKII signaling pathways. Brain Res 2016; 1635:95-104. [PMID: 26826585 DOI: 10.1016/j.brainres.2016.01.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/17/2016] [Accepted: 01/21/2016] [Indexed: 01/11/2023]
Abstract
Dopamine plays an important role in regulating neuronal functions in the central nervous system by activating the specific G-protein coupled receptors. Both D1 and D2 dopamine receptors are extensively distributed in the retinal neurons. In the present study, we investigated the effects of D1 receptor signaling on outward K(+) currents in acutely isolated rat retinal ganglion cells (RGCs) by patch-clamp techniques. Extracellular application of SKF81297 (10 μM), a specific D1 receptor agonist, significantly and reversibly suppressed outward K(+) currents of the cells, which was reversed by SCH23390 (10 μM), a selective D1 receptor antagonist. We further showed that SKF81297 mainly suppressed the glybenclamide (Gb)- and 4-aminopyridine (4-AP)-sensitive K(+) current components, but did not show effect on the tetraethylammonium (TEA)-sensitive one. Both protein kinase A (PKA) and calcium/calmodulin-dependent protein kinase II (CaMKII) signaling pathways were likely involved in the SKF81297-induced suppression of the K(+) currents since either Rp-cAMP (10 μM), a cAMP/PKA signaling inhibitor, or KN-93 (10 μM), a specific CaMKII inhibitor, eliminated the SKF81297 effect. In contrast, neither protein kinase C (PKC) nor mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathway seemed likely to be involved because both the PKC inhibitor bisindolylmaleimide IV (Bis IV) (10 μM) and the MAPK/ERK1/2 inhibitor U0126 (10 μM) did not block the SKF81297-induced suppression of the K(+) currents. These results suggest that activation of D1 receptors suppresses the Gb- and 4-AP-sensitive K(+) current components in rat RGCs through the intracellular PKA and CaMKII signaling pathways, thus modulating the RGC excitability.
Collapse
Affiliation(s)
- Qian Li
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, 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.
| | - Na Wu
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Peng Cui
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, 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.
| | - Feng Gao
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Wen-Jing Qian
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, 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.
| | - Yanying Miao
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Institute of Neurobiology, 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.
| | - Xing-Huai Sun
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Zhongfeng Wang
- Institutes of Brain Science, Fudan University, Shanghai 200032, China; Department of Ophthalmology at Eye & ENT Hospital, Fudan University, Shanghai 200031, China; Institute of Neurobiology, Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
30
|
Jensen R. Effects of Dopamine D2-Like Receptor Antagonists on Light Responses of Ganglion Cells in Wild-Type and P23H Rat Retinas. PLoS One 2015; 10:e0146154. [PMID: 26717015 PMCID: PMC4696741 DOI: 10.1371/journal.pone.0146154] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 12/14/2015] [Indexed: 12/04/2022] Open
Abstract
In animal models of retinitis pigmentosa the dopaminergic system in the retina appears to be dysfunctional, which may contribute to the debilitated sight experienced by retinitis pigmentosa patients. Since dopamine D2-like receptors are known to modulate the activity of dopaminergic neurons, I examined the effects of dopamine D2-like receptor antagonists on the light responses of retinal ganglion cells (RGCs) in the P23H rat model of retinitis pigmentosa. Extracellular electrical recordings were made from RGCs in isolated transgenic P23H rat retinas and wild-type Sprague-Dawley rat retinas. Intensity-response curves to flashes of light were evaluated prior to and during bath application of a dopamine D2-like receptor antagonist. The dopamine D2/D3 receptor antagonists sulpiride and eticlopride and the D4 receptor antagonist L-745,870 increased light sensitivity of P23H rat RGCs but decreased light sensitivity in Sprague-Dawley rat RGCs. In addition, L-745,870, but not sulpiride or eticlopride, reduced the maximum peak responses of Sprague-Dawley rat RGCs. I describe for the first time ON-center RGCs in P23H rats that exhibit an abnormally long-latency (>200 ms) response to the onset of a small spot of light. Both sulpiride and eticlopride, but not L-745,870, reduced this ON response and brought out a short-latency OFF response, suggesting that these cells are in actuality OFF-center cells. Overall, the results show that the altered dopaminergic system in degenerate retinas contributes to the deteriorated light responses of RGCs.
Collapse
Affiliation(s)
- Ralph Jensen
- VA Boston Healthcare System, Mail Stop 151E, 150 South Huntington Avenue, Boston, Massachusetts 02130, United States of America
- * E-mail:
| |
Collapse
|
31
|
Firsov ML, Astakhova LA. The Role of Dopamine in Controlling Retinal Photoreceptor Function in Vertebrates. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s11055-015-0210-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
32
|
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: 3.0] [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
|
33
|
Kakiuchi D, Uehara T, Shiotani M, Nakano-Ito K, Suganuma A, Aoki T, Tsukidate K, Sawada K. Oscillatory potentials in electroretinogram as an early marker of visual abnormalities in vitamin A deficiency. Mol Med Rep 2014; 11:995-1003. [PMID: 25369780 DOI: 10.3892/mmr.2014.2852] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 09/12/2014] [Indexed: 11/05/2022] Open
Abstract
Vitamin A deficiency (VAD) caused by malnutrition and certain intestinal diseases induces visual impairments, including night blindness and photoreceptor cell dysfunction as indicated by reduced a‑ and b‑waves in an electroretinogram (ERG). The effects of VAD on the inner retinal layer cells, including amacrine and ganglion cells, remain to be elucidated. The functions of these cells are reflected in oscillatory potentials (OPs), another component of the ERG. The present study investigated inner retinal layer cell function in VAD rats by analyzing OPs. In the present study, VAD was induced by feeding Brown Norway rats a vitamin A deficient diet for 10 weeks. A reduced body weight and peri‑papillary opacification indicative of papilledema without histopathological alterations were observed, which are considered early symptoms of VAD. At this stage, the ERG revealed reduced OPs as well as a‑ and b‑waves at various intensities of light stimulation. Further analysis indicated that the ratio of the alterations in OPs was more significant than those of a‑ and b‑waves. After 5 weeks of recovery, these changes returned to control levels. These results suggest that OPs are the most sensitive and early marker of VAD‑associated visual impairment in the ERG.
Collapse
Affiliation(s)
- Dai Kakiuchi
- Life Science Center of Tsukuba Advanced Research Alliance, Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki 305‑8577, Japan
| | - Taisuke Uehara
- Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki 300‑2635, Japan
| | - Motohiro Shiotani
- Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki 300‑2635, Japan
| | - Kyoko Nakano-Ito
- Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki 300‑2635, Japan
| | - Akiyoshi Suganuma
- Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki 300‑2635, Japan
| | - Toyohiko Aoki
- Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki 300‑2635, Japan
| | - Kazuo Tsukidate
- Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki 300‑2635, Japan
| | - Kohei Sawada
- Eisai Product Creation Systems, Eisai Co., Ltd., Tokodai, Tsukuba, Ibaraki 300‑2635, Japan
| |
Collapse
|
34
|
Popova E. Role of dopamine in distal retina. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2014; 200:333-58. [PMID: 24728309 DOI: 10.1007/s00359-014-0906-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/24/2014] [Accepted: 03/25/2014] [Indexed: 01/11/2023]
Abstract
Dopamine is the most abundant catecholamine in the vertebrate retina. Despite the description of retinal dopaminergic cells three decades ago, many aspects of their function in the retina remain unclear. There is no consensus among the authors about the stimulus conditions for dopamine release (darkness, steady or flickering light) as well as about its action upon the various types of retinal cells. Many contradictory results exist concerning the dopamine effect on the gross electrical activity of the retina [reflected in electroretinogram (ERG)] and the receptors involved in its action. This review summarized current knowledge about the types of the dopaminergic neurons and receptors in the retina as well as the effects of dopamine receptor agonists and antagonists on the light responses of photoreceptors, horizontal and bipolar cells in both nonmammalian and mammalian retina. Special focus of interest concerns their effects upon the diffuse ERG as a useful tool for assessment of the overall function of the distal retina. An attempt is made to reveal some differences between the dopamine actions upon the activity of the ON versus OFF channel in the distal retina. The author has included her own results demonstrating such differences.
Collapse
Affiliation(s)
- E Popova
- Department of Physiology, Medical Faculty, Medical University, 1431, Sofia, Bulgaria,
| |
Collapse
|
35
|
Ogata G, Stradleigh TW, Partida GJ, Ishida AT. Dopamine and full-field illumination activate D1 and D2-D5-type receptors in adult rat retinal ganglion cells. J Comp Neurol 2013; 520:4032-49. [PMID: 22678972 DOI: 10.1002/cne.23159] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Dopamine can regulate signal generation and transmission by activating multiple receptors and signaling cascades, especially in striatum, hippocampus, and cerebral cortex. Dopamine modulates an even larger variety of cellular properties in retina, yet has been reported to do so by only D1 receptor-driven cyclic adenosine monophosphate (cAMP) increases or D2 receptor-driven cAMP decreases. Here, we test the possibility that dopamine operates differently on retinal ganglion cells, because the ganglion cell layer binds D1 and D2 receptor ligands, and displays changes in signaling components other than cAMP under illumination that should release dopamine. In adult rat retinal ganglion cells, based on patch-clamp recordings, Ca(2+) imaging, and immunohistochemistry, we find that 1) spike firing is inhibited by dopamine and SKF 83959 (an agonist that does not activate homomeric D1 receptors or alter cAMP levels in other systems); 2) D1 and D2 receptor antagonists (SCH 23390, eticlopride, raclopride) counteract these effects; 3) these antagonists also block light-induced rises in cAMP, light-induced activation of Ca(2+) /calmodulin-dependent protein kinase II, and dopamine-induced Ca(2+) influx; and 4) the Ca(2+) rise is markedly reduced by removing extracellular Ca(2+) and by an IP3 receptor antagonist (2-APB). These results provide the first evidence that dopamine activates a receptor in adult mammalian retinal neurons that is distinct from classical D1 and D2 receptors, and that dopamine can activate mechanisms in addition to cAMP and cAMP-dependent protein kinase to modulate retinal ganglion cell excitability.
Collapse
Affiliation(s)
- Genki Ogata
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California 95616, USA
| | | | | | | |
Collapse
|
36
|
Feldkaemper M, Schaeffel F. An updated view on the role of dopamine in myopia. Exp Eye Res 2013; 114:106-19. [PMID: 23434455 DOI: 10.1016/j.exer.2013.02.007] [Citation(s) in RCA: 258] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/23/2013] [Accepted: 02/05/2013] [Indexed: 10/27/2022]
Abstract
A large body of data is available to support the hypothesis that dopamine (DA) is one of the retinal neurotransmitters involved in the signaling cascade that controls eye growth by vision. Initially, reduced retinal DA levels were observed in eyes deprived of sharp vision by either diffusers ("deprivation myopia", DM) or negative lenses ("lens induced myopia", LIM). Simulating high retinal DA levels by intravitreal application of a DA agonist can suppress the development of both DM and LIM. Also more recent studies using knock-out mouse models of DA receptors support the idea of an association between decreased DA levels and DM. There seem to be differences in the magnitude of the effects of DA on DM and LIM, with larger changes in DM but the degrees of image degradation by both treatments need to be matched to support this conclusion. Although a number of studies have shown that the inhibitory effects of dopamine agonists on DM and LIM are mediated through stimulation of the D2-receptor, there is also recent evidence that the balance of D2- and D1-receptor activation is important. Inhibition of D2-receptors can also slow the development of spontaneous myopia in albino guinea pigs. Retinal DA content displays a distinct endogenous diurnal, and partially circadian rhythm. In addition, retinal DA is regulated by a number of visual stimuli like retinal illuminance, spatial frequency content of the image, temporal contrast and, in chicks, by the light input from the pineal organ. A close interaction was found between muscarinergic and dopaminergic systems, and between nitric oxide and dopaminergic pathways, and there is evidence for crosstalk between the different pathways, perhaps multiple binding of the ligands to different receptors. It was shown that DA agonists interact with the immediate early signaling molecule ZENK which triggers the first steps in eye growth regulation. However, since long treatment periods were often needed to induce significant changes in retinal dopamine synthesis and release, the role of dopamine in the early steps is unclear. The wide spatial distribution of dopaminergic amacrine cells in the retina and the observation that changes in dopamine levels can be locally induced by local retinal deprivation is in line with the assumption that dopaminergic mechanisms control both central and peripheral eye growth. The protective effect of outdoor activity on myopia development in children seems to be partly mediated by the stimulatory effect of light on retinal dopamine production and release. However, the dose-response function linking light exposure to dopamine and to the suppression of myopia is not known and requires further studies.
Collapse
Affiliation(s)
- Marita Feldkaemper
- Centre for Ophthalmology, Institute for Ophthalmic Research, Section of Neurobiology of the Eye, Calwerstraße 7/1, 72076 Tuebingen, Germany.
| | | |
Collapse
|
37
|
Irons TD, Kelly PE, Hunter DL, Macphail RC, Padilla S. Acute administration of dopaminergic drugs has differential effects on locomotion in larval zebrafish. Pharmacol Biochem Behav 2012; 103:792-813. [PMID: 23274813 DOI: 10.1016/j.pbb.2012.12.010] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 12/10/2012] [Accepted: 12/13/2012] [Indexed: 12/26/2022]
Abstract
Altered dopaminergic signaling causes behavioral changes in mammals. In general, dopaminergic receptor agonists increase locomotor activity, while antagonists decrease locomotor activity. In order to determine if zebrafish (a model organism becoming popular in pharmacology and toxicology) respond similarly, the acute effects of drugs known to target dopaminergic receptors in mammals were assessed in zebrafish larvae. Larvae were maintained in 96-well microtiter plates (1 larva/well). Non-lethal concentrations (0.2-50 μM) of dopaminergic agonists (apomorphine, SKF-38393, and quinpirole) and antagonists (butaclamol, SCH-23390, and haloperidol) were administered at 6 days post-fertilization (dpf). An initial experiment identified the time of peak effect of each drug (20-260 min post-dosing, depending on the drug). Locomotor activity was then assessed for 70 min in alternating light and dark at the time of peak effect for each drug to delineate dose-dependent effects. All drugs altered larval locomotion in a dose-dependent manner. Both the D1- and D2-like selective agonists (SKF-38393 and quinpirole, respectively) increased activity, while the selective antagonists (SCH-23390 and haloperidol, respectively) decreased activity. Both selective antagonists also blunted the response of the larvae to changes in lighting conditions at higher doses. The nonselective drugs had biphasic effects on locomotor activity: apomorphine increased activity at the low dose and at high doses, while butaclamol increased activity at low to intermediate doses, and decreased activity at high doses. This study demonstrates that (1) larval zebrafish locomotion can be altered by dopamine receptor agonists and antagonists, (2) receptor agonists and antagonists generally have opposite effects, and (3) drugs that target dopaminergic receptors in mammals appear, in general, to elicit similar locomotor responses in zebrafish larvae.
Collapse
Affiliation(s)
- T D Irons
- Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | | | | | | | | |
Collapse
|
38
|
Shen W, Purpura LA, Li B, Nan C, Chang IJ, Ripps H. Regulation of synaptic transmission at the photoreceptor terminal: a novel role for the cation-chloride co-transporter NKCC1. J Physiol 2012; 591:133-47. [PMID: 23090945 DOI: 10.1113/jphysiol.2012.241042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The Na(+)-K(+)-2Cl(-) co-transporter type 1 (NKCC1) is localized primarily throughout the outer plexiform layer (OPL) of the distal retina, a synaptic lamina that is comprised of the axon terminals of photoreceptors and the dendrites of horizontal and bipolar cells. Although known to play a key role in development, signal transmission and the gating of sensory signals in other regions of the retina and in the CNS, the contribution of NKCC1 to synaptic transmission within the OPL is largely unknown. In the present study, we investigated the function of NKCC1 at the photoreceptor-horizontal cell synapse by recording the electrical responses of photoreceptors and horizontal cells before and after blocking the activity of the transporter with bumetanide (BMN). Because NKCC1 co-transports 1 Na(+), 1 K(+) and 2 Cl(-), it is electroneutral and its activation had little effect on membrane conductance. However, recordings from postsynaptic horizontal cells revealed that inhibiting NKCC1 with BMN greatly increased glutamate release from both rod and cone terminals. In addition, we found that NKCC1 directly regulates Ca(2+)-dependent exocytosis at the photoreceptor synapse, raising the possibility that NKCC1 serves to suppress bulk release of glutamate vesicles from photoreceptor terminals in the dark and at light offset. Interestingly, NKCC1 gene and protein expressions were upregulated by light, which we attribute to the light-induced release of dopamine acting on D1-like receptors. In sum, our study reveals a new role for NKCC1 in the regulation of synaptic transmission in photoreceptors.
Collapse
Affiliation(s)
- Wen Shen
- Department of Biomedical Science, Charles E Schmidt College of Medicine, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Regnell CE, Hildrestrand GA, Sejersted Y, Medin T, Moldestad O, Rolseth V, Krokeide SZ, Suganthan R, Luna L, Bjørås M, Bergersen LH. Hippocampal adult neurogenesis is maintained by Neil3-dependent repair of oxidative DNA lesions in neural progenitor cells. Cell Rep 2012; 2:503-10. [PMID: 22959434 DOI: 10.1016/j.celrep.2012.08.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 06/14/2012] [Accepted: 08/10/2012] [Indexed: 12/31/2022] Open
Abstract
Accumulation of oxidative DNA damage has been proposed as a potential cause of age-related cognitive decline. The major pathway for removal of oxidative DNA base lesions is base excision repair, which is initiated by DNA glycosylases. In mice, Neil3 is the main DNA glycosylase for repair of hydantoin lesions in single-stranded DNA of neural stem/progenitor cells, promoting neurogenesis. Adult neurogenesis is crucial for maintenance of hippocampus-dependent functions involved in behavior. Herein, behavioral studies reveal learning and memory deficits and reduced anxiety-like behavior in Neil3(-/-) mice. Neural stem/progenitor cells from aged Neil3(-/-) mice show impaired proliferative capacity and reduced DNA repair activity. Furthermore, hippocampal neurons in Neil3(-/-) mice display synaptic irregularities. It appears that Neil3-dependent repair of oxidative DNA damage in neural stem/progenitor cells is required for maintenance of adult neurogenesis to counteract the age-associated deterioration of cognitive performance.
Collapse
Affiliation(s)
- Christine Elisabeth Regnell
- Department of Anatomy, Centre for Molecular Biology and Neuroscience, University of Oslo, N-0317 Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hartveit E, Veruki ML. Electrical synapses between AII amacrine cells in the retina: Function and modulation. Brain Res 2012; 1487:160-72. [PMID: 22776293 DOI: 10.1016/j.brainres.2012.05.060] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 05/09/2012] [Indexed: 12/24/2022]
Abstract
Adaptation enables the visual system to operate across a large range of background light intensities. There is evidence that one component of this adaptation is mediated by modulation of gap junctions functioning as electrical synapses, thereby tuning and functionally optimizing specific retinal microcircuits and pathways. The AII amacrine cell is an interneuron found in most mammalian retinas and plays a crucial role for processing visual signals in starlight, twilight and daylight. AII amacrine cells are connected to each other by gap junctions, potentially serving as a substrate for signal averaging and noise reduction, and there is evidence that the strength of electrical coupling is modulated by the level of background light. Whereas there is extensive knowledge concerning the retinal microcircuits that involve the AII amacrine cell, it is less clear which signaling pathways and intracellular transduction mechanisms are involved in modulating the junctional conductance between electrically coupled AII amacrine cells. Here we review the current state of knowledge, with a focus on the recent evidence that suggests that the modulatory control involves activity-dependent changes in the phosphorylation of the gap junction channels between AII amacrine cells, potentially linked to their intracellular Ca(2+) dynamics. This article is part of a Special Issue entitled Electrical Synapses.
Collapse
Affiliation(s)
- Espen Hartveit
- University of Bergen, Department of Biomedicine, Bergen, Norway.
| | | |
Collapse
|
41
|
Jackson CR, Ruan GX, Aseem F, Abey J, Gamble K, Stanwood G, Palmiter RD, Iuvone PM, McMahon DG. Retinal dopamine mediates multiple dimensions of light-adapted vision. J Neurosci 2012; 32:9359-68. [PMID: 22764243 PMCID: PMC3400466 DOI: 10.1523/jneurosci.0711-12.2012] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 05/14/2012] [Accepted: 05/19/2012] [Indexed: 11/21/2022] Open
Abstract
Dopamine is a key neuromodulator in the retina and brain that supports motor, cognitive, and visual function. Here, we developed a mouse model on a C57 background in which expression of the rate-limiting enzyme for dopamine synthesis, tyrosine hydroxylase, is specifically disrupted in the retina. This model enabled assessment of the overall role of retinal dopamine in vision using electrophysiological (electroretinogram), psychophysical (optokinetic tracking), and pharmacological techniques. Significant disruptions were observed in high-resolution, light-adapted vision caused by specific deficits in light responses, contrast sensitivity, acuity, and circadian rhythms in this retinal dopamine-depleted mouse model. These global effects of retinal dopamine on vision are driven by the differential actions of dopamine D1 and D4 receptors on specific retinal functions and appear to be due to the ongoing bioavailability of dopamine rather than developmental effects. Together, our data indicate that dopamine is necessary for the circadian nature of light-adapted vision as well as optimal contrast detection and acuity.
Collapse
Affiliation(s)
- Chad R. Jackson
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37235
| | - Guo-Xiang Ruan
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37235
| | - Fazila Aseem
- Departments of Ophthalmology and Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Jane Abey
- Departments of Ophthalmology and Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Karen Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama-Birmingham, Birmingham, Alabama 35294-0017
| | - Greg Stanwood
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, and
| | - Richard D. Palmiter
- Howard Hughes Medical Institute and Department of Biochemistry, University of Washington, Seattle, Washington 98195
| | - P. Michael Iuvone
- Departments of Ophthalmology and Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Douglas G. McMahon
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37235
| |
Collapse
|
42
|
Abstract
Amacrine cells represent the most diverse class of retinal neuron, comprising dozens of distinct cell types. Each type exhibits a unique morphology and generates specific visual computations through its synapses with a subset of excitatory interneurons (bipolar cells), other amacrine cells, and output neurons (ganglion cells). Here, we review the intrinsic and network properties that underlie the function of the most common amacrine cell in the mammalian retina, the AII amacrine cell. The AII connects rod and cone photoreceptor pathways, forming an essential link in the circuit for rod-mediated (scotopic) vision. As such, the AII has become known as the rod-amacrine cell. We, however, now understand that AII function extends to cone-mediated (photopic) vision, and AII function in scotopic and photopic conditions utilizes the same underlying circuit: AIIs are electrically coupled to each other and to the terminals of some types of ON cone bipolar cells. The direction of signal flow, however, varies with illumination. Under photopic conditions, the AII network constitutes a crossover inhibition pathway that allows ON signals to inhibit OFF ganglion cells and contributes to motion sensitivity in certain ganglion cell types. We discuss how the AII's combination of intrinsic and network properties accounts for its unique role in visual processing.
Collapse
|
43
|
Haj-Yasein NN, Jensen V, Østby I, Omholt SW, Voipio J, Kaila K, Ottersen OP, Hvalby Ø, Nagelhus EA. Aquaporin-4 regulates extracellular space volume dynamics during high-frequency synaptic stimulation: a gene deletion study in mouse hippocampus. Glia 2012; 60:867-74. [PMID: 22419561 DOI: 10.1002/glia.22319] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 02/14/2012] [Indexed: 11/10/2022]
Abstract
Little is known about the physiological roles of aquaporin-4 (AQP4) in the central nervous system. AQP4 water channels are concentrated in endfeet membranes of astrocytes but also localize to the fine astrocytic processes that abut central synapses. Based on its pattern of expression, we predicted that AQP4 could be involved in controlling water fluxes and changes in extracellular space (ECS) volume that are associated with activation of excitatory pathways. Here, we show that deletion of Aqp4 accentuated the shrinkage of the ECS that occurred in the mouse hippocampal CA1 region during activation of Schaffer collateral/commissural fibers. This effect was found in the stratum radiatum (where perisynaptic astrocytic processes abound) but not in the pyramidal cell layer (where astrocytic processes constitute but a minor volume fraction). For both genotypes the ECS shrinkage was most pronounced in the pyramidal cell layer. Our data attribute a physiological role to AQP4 and indicate that this water channel regulates extracellular volume dynamics in the mammalian brain.
Collapse
Affiliation(s)
- Nadia Nabil Haj-Yasein
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Van Hook MJ, Wong KY, Berson DM. Dopaminergic modulation of ganglion-cell photoreceptors in rat. Eur J Neurosci 2012; 35:507-18. [PMID: 22304466 DOI: 10.1111/j.1460-9568.2011.07975.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A novel class of photoreceptors, the intrinsically photosensitive retinal ganglion cells (ipRGCs), express the photopigment melanopsin and drive non-image-forming responses to light such as circadian photoentrainment, the pupillary light reflex and suppression of nocturnal melatonin production in the pineal. Because dendrites from one subclass of these cells - the M1-type ipRGCs - make presumptive synaptic contacts at sites of dopamine release from dopaminergic amacrine cells, they are prime targets for modulation by dopamine, a neuromodulator implicated in retinal circadian rhythms and light adaptation. In patch-clamp recordings from ipRGCs in intact rat retinas, dopamine attenuated the melanopsin-based photocurrent. We confirmed that this was the result of direct action on ipRGCs by replicating the effect in dissociated ipRGCs that were isolated from influences of other retinal neurons. In these recordings, the D1-family dopamine receptor agonist SKF38393 attenuated the photocurrent, caused a modest depolarization, and reduced the input resistance of ipRGCs. The D2-family agonist quinpirole had no effect on the photocurrent. Single-cell reverse-transcriptase polymerase chain reaction revealed that the majority of ipRGCs tested expressed drd1a, the gene coding for the D1a dopamine receptor. This finding was supported by immunohistochemical localization of D1a receptor protein in melanopsin-expressing ganglion cells. Finally, the adenylate cyclase activator forskolin, applied in combination with the phosphodiesterase inhibitor IBMX (isobutylmethylxanthine), mimicked the effects of SKF38393 on the ipRGC photocurrent, membrane potential and input resistance, consistent with a D1-receptor signaling pathway. These data suggest that dopamine, acting via D1-family receptors, alters the responses of ipRGCs and thus of non-image-forming vision.
Collapse
Affiliation(s)
- Matthew J Van Hook
- Department of Neuroscience, Brown University, Box G-LN, Providence, RI, USA
| | | | | |
Collapse
|
45
|
Herrmann R, Heflin SJ, Hammond T, Lee B, Wang J, Gainetdinov RR, Caron MG, Eggers ED, Frishman LJ, McCall MA, Arshavsky VY. Rod vision is controlled by dopamine-dependent sensitization of rod bipolar cells by GABA. Neuron 2011; 72:101-10. [PMID: 21982372 DOI: 10.1016/j.neuron.2011.07.030] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2011] [Indexed: 11/19/2022]
Abstract
Dark and light adaptation of retinal neurons allow our vision to operate over an enormous light intensity range. Here we report a mechanism that controls the light sensitivity and operational range of rod-driven bipolar cells that mediate dim-light vision. Our data indicate that the light responses of these cells are enhanced by sustained chloride currents via GABA(C) receptor channels. This sensitizing GABAergic input is controlled by dopamine D1 receptors, with horizontal cells serving as a plausible source of GABA release. Our findings expand the role of dopamine in vision from its well-established function of suppressing rod-driven signals in bright light to enhancing the same signals under dim illumination. They further reveal a role for GABA in sensitizing the circuitry for dim-light vision, thereby complementing GABA's traditional role in providing dynamic feedforward and feedback inhibition in the retina.
Collapse
Affiliation(s)
- Rolf Herrmann
- Albert Eye Research Institute, Duke University, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Nickla DL, Totonelly K, Dhillon B. Dopaminergic agonists that result in ocular growth inhibition also elicit transient increases in choroidal thickness in chicks. Exp Eye Res 2010; 91:715-20. [PMID: 20801115 PMCID: PMC2962673 DOI: 10.1016/j.exer.2010.08.021] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 08/12/2010] [Accepted: 08/19/2010] [Indexed: 10/19/2022]
Abstract
The dopaminergic system has been implicated in ocular growth regulation in chicks and monkeys. In both, dopamine D2 agonists inhibit the development of myopia in response to form deprivation, and in chicks, to negative lenses as well. Because there is mounting evidence that the choroidal response to defocus plays a role in ocular growth regulation, we asked whether the effective agonists also elicit transient thickening of the choroid concomitant with the growth inhibition. Negative lenses mounted on velcro rings were worn on one eye starting at age 8-12 days. Intravitreal injections (20 μl; dose = 10 nmole) of the agonist (dissolved in saline) or saline, were given through the superior temporal sclera using a 30G needle. Eyes were injected daily at noon, for 4 days, and the lenses immediately replaced. Agonists used were apomorphine (non-specific; n = 17), quinpirole (D2; n = 10), SKF-38393 (D1; n = 9), and saline controls (n = 22). For the antagonists, the same protocol was used, but on each day, the lenses were removed for 2 h. Immediately prior to lens-removal, the antagonist was injected (20 μl; dose = 5 nmole). Antagonists used were methylergonovine (non-specific; n = 12), spiperone (D2; n = 20), SCH-23390 (D1; n = 6) and saline controls (n = 27). Comparisons to saline (continuous lens wear) controls were from the agonist experiment. Axial dimensions were measured using high frequency A-scan ultrasonography at the start of lens wear, and on day 4 prior to the injections, and then again 3 h later. Refractive errors were measured using a Hartinger's refractometer at the end of the experiment. Apomorphine and quinpirole inhibited the refractive response to the hyperopic defocus induced by the negative lenses (drug vs saline controls: -1.3 and 1.2 D vs -5.6 D; p < 0.005 for both). This effect was axial: both drugs prevented the excessive ocular elongation (change in axial length: 233 and 205 μm vs 417 μm; p < 0.01 for both). Both drugs were also associated with a transient thickening of the choroid over 3 h (41 and 32 μm vs -1 μm; p < 0.01; p = 0.059 respectively) that did not summate: choroids thinned significantly over the 4 day period in all lens-wearing eyes. Two daily hours of unrestricted vision during negative lens wear normally prevents the development of myopia. Spiperone and SCH-23390 inhibited the ameliorating effects of periods of vision on lens-induced refractive error (-2.9 and -2.8 D vs 0.6 D; p < 0.0001), however, the effects on neither axial length nor choroidal thickness were significant. These data support a role for both D1 and D2 receptors in the ocular growth responses.
Collapse
Affiliation(s)
- Debora L Nickla
- The New England College of Optometry, Dept. of Biosciences, 424 Beacon St., Boston, MA 02115, USA.
| | | | | |
Collapse
|
47
|
Leonelli M, Martins DO, Britto LRG. TRPV1 receptors are involved in protein nitration and Müller cell reaction in the acutely axotomized rat retina. Exp Eye Res 2010; 91:755-68. [PMID: 20826152 DOI: 10.1016/j.exer.2010.08.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 06/24/2010] [Accepted: 08/30/2010] [Indexed: 12/20/2022]
Abstract
We report here the protein expression of TRPV1 receptor in axotomized rat retinas and its possible participation in mechanisms involved in retinal ganglion cell (RGC) death. Adult rats were subjected to unilateral, intraorbital axotomy of the optic nerve, and the retinal tissue was removed for further processing. TRPV1 total protein expression decreased progressively after optic nerve transection, reaching 66.2% of control values 21 days after axotomy. The number of cells labeled for TRPV1 in the remnant GCL decreased after 21 days post-lesion (to 63%). Fluoro-Jade B staining demonstrated that the activation of TRPV1 in acutely-lesioned eyes elicited more intense neuronal degeneration in the GCL and in the inner nuclear layer than in sham-operated retinas. A single intraocular injection of capsazepine (100 μM), a TRPV1 antagonist, 5 days after optic nerve lesion, decreased the number of GFAP-expressing Müller cells (72.5% of control values) and also decreased protein nitration in the retinal vitreal margin (75.7% of control values), but did not affect lipid peroxidation. Furthermore, retinal explants were treated with capsaicin (100 μM), and remarkable protein nitration was then present, which was reduced by blockers of the constitutive and inducible nitric oxide synthases (7-NI and aminoguanidine, respectively). TRPV1 activation also increased GFAP expression, which was reverted by both TRPV1 antagonism with capsazepine and by 7-NI and aminoguanidine. Given that Müller cells do not express TRPV1, we suppose that the increased GFAP expression in these cells might be elicited by TRPV1 activation and by its indirect effect upon nitric oxide overproduction and peroxynitrite formation. We incubated Fluorogold pre-labeled retinal explants in the presence of capsazepine (1 μM) during 48 h. The numbers of surviving RGCs stained with fluorogold and the numbers of apoptotic cells in the GCL detected with TUNEL were similar in lesioned and control retinas. We conclude that TRPV1 receptor expression decreased after optic nerve injury due to death of TRPV1-containing cells. Furthermore, these data indicate that TRPV1 might be involved in intrinsic protein nitration and Müller cell reaction observed after optic nerve injury.
Collapse
Affiliation(s)
- Mauro Leonelli
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-900, Brazil.
| | | | | |
Collapse
|
48
|
Keeley PW, Reese BE. Morphology of dopaminergic amacrine cells in the mouse retina: independence from homotypic interactions. J Comp Neurol 2010; 518:1220-31. [PMID: 20148440 PMCID: PMC2865197 DOI: 10.1002/cne.22270] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To determine the role of homotypic interactions between neighboring dopaminergic amacrine (DA) cells upon dendritic morphogenesis, the morphology of single cells was examined relative to the positioning of all neighboring homotypic cells. For each labeled cell, the dendritic field was reconstructed, its Voronoi domain was calculated, and the two were related. The dendritic fields of DA cells were observed to be large, sparse, and highly irregular. Dendrites readily overlapped those of neighboring cells, showing no evidence for dendritic tiling or inter-digitation consistent with homotypic repulsion or avoidance. Furthermore, a direct comparison of dendritic field area with the Voronoi domain area of the same cell showed no evidence for dendritic growth being constrained or biased by the local distribution of homotypic neighbors in wild-type retinas. A comparison of the processes of adjacent filled cells confirmed their immediate proximity to one another within the inner plexiform layer, indicating that they do not engage in mutual avoidance by coursing at different depths. Together, these results suggest that the morphogenesis of DA cells is independent of homotypic interactions. However, in the absence of the pro-apoptotic Bax gene, which yields a fourfold increase in DA cell number, a small but significant reduction in dendritic field size was obtained, although not so great as would be predicted by the increase in density. The present results are considered in light of recent studies on the role of cell adhesion molecules expressed by developing DA cells.
Collapse
Affiliation(s)
- Patrick W Keeley
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California 93106-9625, USA
| | | |
Collapse
|
49
|
Hayashida Y, Rodríguez CV, Ogata G, Partida GJ, Oi H, Stradleigh TW, Lee SC, Colado AF, Ishida AT. Inhibition of adult rat retinal ganglion cells by D1-type dopamine receptor activation. J Neurosci 2009; 29:15001-16. [PMID: 19940196 PMCID: PMC3236800 DOI: 10.1523/jneurosci.3827-09.2009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 10/01/2009] [Accepted: 10/22/2009] [Indexed: 11/21/2022] Open
Abstract
The spike output of neural pathways can be regulated by modulating output neuron excitability and/or their synaptic inputs. Dopaminergic interneurons synapse onto cells that route signals to mammalian retinal ganglion cells, but it is unknown whether dopamine can activate receptors in these ganglion cells and, if it does, how this affects their excitability. Here, we show D(1a) receptor-like immunoreactivity in ganglion cells identified in adult rats by retrogradely transported dextran, and that dopamine, D(1)-type receptor agonists, and cAMP analogs inhibit spiking in ganglion cells dissociated from adult rats. These ligands curtailed repetitive spiking during constant current injections and reduced the number and rate of rise of spikes elicited by fluctuating current injections without significantly altering the timing of the remaining spikes. Consistent with mediation by D(1)-type receptors, SCH-23390 [R-(+)-7-chloro-8-hydroxy-3-methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine] reversed the effects of dopamine on spikes. Contrary to a recent report, spike inhibition by dopamine was not precluded by blocking I(h). Consistent with the reduced rate of spike rise, dopamine reduced voltage-gated Na(+) current (I(Na)) amplitude, and tetrodotoxin, at doses that reduced I(Na) as moderately as dopamine, also inhibited spiking. These results provide the first direct evidence that D(1)-type dopamine receptor activation can alter mammalian retinal ganglion cell excitability and demonstrate that dopamine can modulate spikes in these cells by a mechanism different from the presynaptic and postsynaptic means proposed by previous studies. To our knowledge, our results also provide the first evidence that dopamine receptor activation can reduce excitability without altering the temporal precision of spike firing.
Collapse
Affiliation(s)
- Yuki Hayashida
- Departments of Neurobiology, Physiology, and Behavior, and
| | | | - Genki Ogata
- Departments of Neurobiology, Physiology, and Behavior, and
| | | | - Hanako Oi
- Departments of Neurobiology, Physiology, and Behavior, and
| | | | - Sherwin C. Lee
- Departments of Neurobiology, Physiology, and Behavior, and
| | | | - Andrew T. Ishida
- Departments of Neurobiology, Physiology, and Behavior, and
- Ophthalmology and Vision Science, University of California, Davis, Davis, California 95616
| |
Collapse
|
50
|
Hirasawa H, Puopolo M, Raviola E. Extrasynaptic release of GABA by retinal dopaminergic neurons. J Neurophysiol 2009; 102:146-58. [PMID: 19403749 DOI: 10.1152/jn.00130.2009] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
GABA release by dopaminergic amacrine (DA) cells of the mouse retina was detected by measuring Cl- currents generated by isolated perikarya in response to their own neurotransmitter. The possibility that the Cl- currents were caused by GABA release from synaptic endings that had survived the dissociation of the retina was ruled out by examining confocal Z series of the surface of dissociated tyrosine hydroxylase-positive perikarya after staining with antibodies to pre and postsynaptic markers. GABA release was caused by exocytosis because 1) the current events were transient on the millisecond time scale and thus resembled miniature synaptic currents; 2) they were abolished by treatment with a blocker of the vesicular proton pump, bafilomycin A1; and 3) their frequency was controlled by the intracellular Ca2+ concentration. Because DA cell perikarya do not contain presynaptic active zones, release was by necessity extrasynaptic. A range of depolarizing stimuli caused GABA exocytosis, showing that extrasynaptic release of GABA is controlled by DA cell electrical activity. With all modalities of stimulation, including long-lasting square pulses, segments of pacemaker activity delivered by the action-potential-clamp method and high-frequency trains of ramps, discharge of GABAergic currents exhibited considerable variability in latency and duration, suggesting that coupling between Ca2+ influx and transmitter exocytosis is extremely loose in comparison with the synapse. Paracrine signaling based on extrasynaptic release of GABA by DA cells and other GABAergic amacrines may participate in controlling the excitability of the neuronal processes that interact synaptically in the inner plexiform layer.
Collapse
Affiliation(s)
- Hajime Hirasawa
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA
| | | | | |
Collapse
|