1
|
Gonschorek D, Goldin MA, Oesterle J, Schwerd-Kleine T, Arlinghaus R, Zhao Z, Schubert T, Marre O, Euler T. Nitric oxide modulates contrast suppression in a subset of mouse retinal ganglion cells. eLife 2025; 13:RP98742. [PMID: 39783858 PMCID: PMC11717361 DOI: 10.7554/elife.98742] [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] [Indexed: 01/12/2025] Open
Abstract
Neuromodulators have major influences on the regulation of neural circuit activity across the nervous system. Nitric oxide (NO) has been shown to be a prominent neuromodulator in many circuits and has been extensively studied in the retina. Here, it has been associated with the regulation of light adaptation, gain control, and gap junctional coupling, but its effect on the retinal output, specifically on the different types of retinal ganglion cells (RGCs), is still poorly understood. In this study, we used two-photon Ca2+ imaging and multi-electrode array (MEA) recordings to measure light-evoked activity of RGCs in the ganglion cell layer in the ex vivo mouse retina. This approach allowed us to investigate the neuromodulatory effects of NO on a cell type-level. Our findings reveal that NO selectively modulates the suppression of temporal responses in a distinct subset of contrast-suppressed RGC types, increasing their activity without altering the spatial properties of their receptive fields. Given that under photopic conditions, NO release is triggered by quick changes in light levels, we propose that these RGC types signal fast contrast changes to higher visual regions. Remarkably, we found that about one-third of the RGC types, recorded using two-photon Ca2+ imaging, exhibited consistent, cell type-specific adaptational response changes throughout an experiment, independent of NO. By employing a sequential-recording paradigm, we could disentangle those additional adaptational response changes from drug-induced modulations. Taken together, our research highlights the selective neuromodulatory effects of NO on RGCs and emphasizes the need of considering non-pharmacological activity changes, like adaptation, in such study designs.
Collapse
Affiliation(s)
- Dominic Gonschorek
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
| | - Matías A Goldin
- Institut de la Vision, Sorbonne Université, INSERM, CNRSParisFrance
| | - Jonathan Oesterle
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- Hertie Institute for AI in Brain Health, Tübingen AI Center, University of TübingenTübingenGermany
| | - Tom Schwerd-Kleine
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
| | - Ryan Arlinghaus
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Zhijian Zhao
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Timm Schubert
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
| | - Olivier Marre
- Institut de la Vision, Sorbonne Université, INSERM, CNRSParisFrance
| | - Thomas Euler
- Werner Reichardt Centre for Integrative Neuroscience, University of TübingenTübingenGermany
- Institute for Ophthalmic Research, University of TübingenTübingenGermany
- GRK 2381 ’cGMP: From Bedside to Bench’, University of TübingenTübingenGermany
- Bernstein Center for Computational Neuroscience, University of TübingenTübingenGermany
| |
Collapse
|
2
|
Merkley MB, Soriano D, Jones KL, Summers JA. The Effects of Nitric Oxide on Choroidal Gene Expression. JOURNAL OF BIOINFORMATICS AND SYSTEMS BIOLOGY : OPEN ACCESS 2024; 7:39-51. [PMID: 39867920 PMCID: PMC11759056 DOI: 10.26502/jbsb.5107077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Purpose Nitric oxide (NO) is recognized as an important biological mediator that controls several physiological functions, and evidence is now emerging that this molecule may play a significant role in the postnatal control of ocular growth and myopia development. We therefore sought to understand the role that nitric oxide plays in visually-guided ocular growth in order to gain insight into the underlying mechanisms of this process. Methods Choroids were incubated in organ culture in the presence of the NO donor, PAPA- NONOate (1.5 mM). Following RNA extraction, bulk RNA-seq was used to quantify and compare choroidal gene expression in the presence and absence of PAPA-NONOate. We used bioinformatics to identify enriched canonical pathways, predicted diseases and functions, and regulatory effects of NO in the choroid. Results Upon treatment of normal chick choroids with the NO donor, PAPA-NONOate, we identified a total of 837 differentially expressed genes (259 upregulated genes, 578 down-regulated genes) compared with untreated controls. Among these, the top five upregulated genes were LSMEM1, STEAP4, HSPB9, and CCL19, and the top five down-regulated genes were CDCA3, SMC2, a novel gene (ENSALGALG00000050836), an uncharacterized gene (LOC107054158), and SPAG5. Bioinformatics predicted that NO treatment will activate pathways involved in cell and organismal death, necrosis, and cardiovascular system development, and inhibit pathways involved in cell proliferation, cell movement, and gene expression. Conclusions The findings reported herein may provide insight into possible effects of NO in the choroid during visually regulated eye growth, and help to identify targeted therapies for the treatment of myopia and other ocular diseases.
Collapse
Affiliation(s)
- Makenzie B Merkley
- Department of Biology, University of Oklahoma, Norman, Oklahoma, 73019, United States
| | - Diana Soriano
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, United States
| | | | - Jody A Summers
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, United States
| |
Collapse
|
3
|
Merkley MB, Soriano D, Jones KL, Summers JA. The Effects of Nitric Oxide on Choroidal Gene Expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545343. [PMID: 37398322 PMCID: PMC10312785 DOI: 10.1101/2023.06.16.545343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Purpose Nitric oxide (NO) is recognized as an important biological mediator that controls several physiological functions, and evidence is now emerging that this molecule may play a significant role in the postnatal control of ocular growth and myopia development. We therefore sought to understand the role that nitric oxide plays in visually-guided ocular growth in order to gain insight into the underlying mechanisms of this process. Methods Choroids were incubated in organ culture in the presence of the NO donor, PAPA-NONOate (1.5 mM). Following RNA extraction, bulk RNA-seq was used to quantify and compare choroidal gene expression in the presence and absence of PAPA-NONOate. We used bioinformatics to identify enriched canonical pathways, predicted diseases and functions, and regulatory effects of NO in the choroid. Results Upon treatment of normal chick choroids with the NO donor, PAPA-NONOate, we identified a total of 837 differentially expressed genes (259 upregulated genes, 578 down-regulated genes) compared with untreated controls. Among these, the top five upregulated genes were LSMEM1, STEAP4, HSPB9, and CCL19, and the top five down-regulated genes were CDCA3, SMC2, a novel gene (ENSALGALG00000050836), an uncharacterized gene (LOC107054158), and SPAG5. Bioinformatics predicted that NO treatment will activate pathways involved in cell and organismal death, necrosis, and cardiovascular system development, and inhibit pathways involved in cell proliferation, cell movement, and gene expression. Conclusions The findings reported herein may provide insight into possible effects of NO in the choroid during visually regulated eye growth, and help to identify targeted therapies for the treatment of myopia and other ocular diseases.
Collapse
Affiliation(s)
- Makenzie B Merkley
- Department of Biology, University of Oklahoma, Norman, Oklahoma, 73019, United States
| | - Diana Soriano
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, United States
| | | | - Jody A Summers
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, United States
| |
Collapse
|
4
|
Duarte-Silva AT, Ximenes LGR, Guimarães-Souza M, Domith I, Paes-de-Carvalho R. Chemical signaling in the developing avian retina: Focus on cyclic AMP and AKT-dependent pathways. Front Cell Dev Biol 2022; 10:1058925. [PMID: 36568967 PMCID: PMC9780464 DOI: 10.3389/fcell.2022.1058925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Communication between developing progenitor cells as well as differentiated neurons and glial cells in the nervous system is made through direct cell contacts and chemical signaling mediated by different molecules. Several of these substances are synthesized and released by developing cells and play roles since early stages of Central Nervous System development. The chicken retina is a very suitable model for neurochemical studies, including the study of regulation of signaling pathways during development. Among advantages of the model are its very well-known histogenesis, the presence of most neurotransmitter systems found in the brain and the possibility to make cultures of neurons and/or glial cells where many neurochemical functions develop in a similar way than in the intact embryonic tissue. In the chicken retina, some neurotransmitters or neuromodulators as dopamine, adenosine, and others are coupled to cyclic AMP production or adenylyl cyclase inhibition since early stages of development. Other substances as vitamin C and nitric oxide are linked to the major neurotransmitter glutamate and AKT metabolism. All these different systems regulate signaling pathways, including PKA, PKG, SRC, AKT and ERK, and the activation of the transcription factor CREB. Dopamine and adenosine stimulate cAMP accumulation in the chick embryo retina through activation of D1 and A2a receptors, respectively, but the onset of dopamine stimulation is much earlier than that of adenosine. However, adenosine can inhibit adenylyl cyclase and modulate dopamine-dependent cAMP increase since early developmental stages through A1 receptors. Dopamine stimulates different PKA as well as EPAC downstream pathways both in intact tissue and in culture as the CSK-SRC pathway modulating glutamate NMDA receptors as well as vitamin C release and CREB phosphorylation. By the other hand, glutamate modulates nitric oxide production and AKT activation in cultured retinal cells and this pathway controls neuronal survival in retina. Glutamate and adenosine stimulate the release of vitamin C and this vitamin regulates the transport of glutamate, activation of NMDA receptors and AKT phosphorylation in cultured retinal cells. In the present review we will focus on these reciprocal interactions between neurotransmitters or neuromodulators and different signaling pathways during retinal development.
Collapse
Affiliation(s)
- A. T. Duarte-Silva
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - L. G. R. Ximenes
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - M. Guimarães-Souza
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - I. Domith
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - R. Paes-de-Carvalho
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil,Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil,*Correspondence: R. Paes-de-Carvalho,
| |
Collapse
|
5
|
Rodriguez TC, Zhong L, Simpson H, Gleason E. Reduced Expression of TMEM16A Impairs Nitric Oxide-Dependent Cl− Transport in Retinal Amacrine Cells. Front Cell Neurosci 2022; 16:937060. [PMID: 35966201 PMCID: PMC9363626 DOI: 10.3389/fncel.2022.937060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Postsynaptic cytosolic Cl− concentration determines whether GABAergic and glycinergic synapses are inhibitory or excitatory. We have shown that nitric oxide (NO) initiates the release of Cl− from acidic internal stores into the cytosol of retinal amacrine cells (ACs) thereby elevating cytosolic Cl−. In addition, we found that cystic fibrosis transmembrane conductance regulator (CFTR) expression and Ca2+ elevations are necessary for the transient effects of NO on cytosolic Cl− levels, but the mechanism remains to be elucidated. Here, we investigated the involvement of TMEM16A as a possible link between Ca2+ elevations and cytosolic Cl− release. TMEM16A is a Ca2+-activated Cl− channel that is functionally coupled with CFTR in epithelia. Both proteins are also expressed in neurons. Based on this and its Ca2+ dependence, we test the hypothesis that TMEM16A participates in the NO-dependent elevation in cytosolic Cl− in ACs. Chick retina ACs express TMEM16A as shown by Western blot analysis, single-cell PCR, and immunocytochemistry. Electrophysiology experiments demonstrate that TMEM16A functions in amacrine cells. Pharmacological inhibition of TMEM16A with T16inh-AO1 reduces the NO-dependent Cl− release as indicated by the diminished shift in the reversal potential of GABAA receptor-mediated currents. We confirmed the involvement of TMEM16A in the NO-dependent Cl− release using CRISPR/Cas9 knockdown of TMEM16A. Two different modalities targeting the gene for TMEM16A (ANO1) were tested in retinal amacrine cells: an all-in-one plasmid vector and crRNA/tracrRNA/Cas9 ribonucleoprotein. The all-in-one CRISPR/Cas9 modality did not change the expression of TMEM16A protein and produced no change in the response to NO. However, TMEM16A-specific crRNA/tracrRNA/Cas9 ribonucleoprotein effectively reduces both TMEM16A protein levels and the NO-dependent shift in the reversal potential of GABA-gated currents. These results show that TMEM16A plays a role in the NO-dependent Cl− release from retinal ACs.
Collapse
|
6
|
Samaha D, Diaconu V, Bouchard JF, Desalliers C, Dupont A. Effect of Latanoprostene Bunod on Optic Nerve Head Blood Flow. Optom Vis Sci 2022; 99:172-176. [PMID: 34889858 DOI: 10.1097/opx.0000000000001842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
SIGNIFICANCE Topical latanoprostene bunod increases capillary oxygen saturation and blood volume at the optic nerve head in healthy individuals. PURPOSE This study aimed to evaluate the effect of topical latanoprostene bunod on optic nerve blood volume and oxygen saturation in a population of healthy participants. METHODS In this prospective double-blind crossover study, 23 healthy participants aged from 21 to 62 years were recruited. Optic nerve head capillary blood volume (ONHvol) and oxygen saturation (ONHSaO2) baselines were measured over a period of 2 hours using multichannel spectroscopic reflectometry and were remeasured after a 7-day once-daily instillation regimen of either latanoprost 0.005% or latanoprostene bunod 0.024%. After a 30-day washout period, participants were crossed over to the alternate product, and measurements were repeated. Participants were used as their own baselines to calculate variation in ONHvol and ONHSaO2 across time and pharmacological agents. The Friedman test was used to establish significant differences in optic nerve head parameters from baseline values, and Conover post hoc analysis was carried for multiple between-group comparisons. RESULTS Latanoprostene bunod 0.024% induced a significant increase of 4% in ONHSaO2 compared with latanoprost 0.005% (P < .001). Furthermore, latanoprostene bunod increased ONHvol levels by more than twofold at all time points (P < .001 at T60, T90, and T120). The increase in ONHvol was 66.2% higher than levels achieved with latanoprost at T60 (P = .001), 47% higher at T90 (P < .001), and 45% higher at T120 (P < .01). CONCLUSIONS Latanoprostene bunod 0.024% induces a significant increase in optic nerve head blood volume and oxygen saturation in healthy subjects, when compared with latanoprost 0.005%. Future studies are needed to evaluate whether similar responses are elicited in patients suffering from glaucomatous optic neuropathy.
Collapse
Affiliation(s)
| | - Vasile Diaconu
- Montreal University School of Optometry, Montreal, Quebec, Canada
| | | | | | - Ariane Dupont
- Montreal University School of Optometry, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Thébault S. Minireview: Insights into the role of TRP channels in the retinal circulation and function. Neurosci Lett 2021; 765:136285. [PMID: 34634394 DOI: 10.1016/j.neulet.2021.136285] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/25/2021] [Accepted: 08/28/2021] [Indexed: 12/17/2022]
Abstract
Consistent with their wide distribution throughout the CNS, transcripts of all transient receptor potential (TRP) cation channel superfamily members have been detected in both neuronal and non-neuronal cells of the mammalian retina. Evidence shows that members of the TRPC (canonical, TRPC1/4/5/6), TRPV (vanilloid, TRPV1/2/4), TRPM (melastatin, TRPM1/2/3/5), TRPA (ankyrin, TRPA1), and TRPP (polycystin, TRPP2) subfamilies contribute to retinal function and circulation in health and disease, but the relevance of most TRPs has yet to be determined. Their principal role in light detection is far better understood than their participation in the control of intraocular pressure, retinal blood flow, oxidative stress, ion homeostasis, and transmitter signaling for retinal information processing. Moreover, if the therapeutic potential of targeting some TRPs to treat various retinal diseases remains speculative, recent studies highlight that vision restoration strategies are very likely to benefit from the thermo- and mechanosensitive properties of TRPs. This minireview focuses on the evidence of the past 5 years about the role of TRPs in the retina and retinal circulation, raises some possibilities about the function of TRPs in the retina, and discusses the potential sources of endogenous stimuli for TRPs in this tissue, as a reflection for future studies.
Collapse
Affiliation(s)
- Stéphanie Thébault
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, 76230 Querétaro, Mexico.
| |
Collapse
|
8
|
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
|
9
|
Vargas A, Yamamoto KL, Craft CM, Lee EJ. Clusterin enhances cell survival by suppressing neuronal nitric-oxide synthase expression in the rhodopsin S334ter-line3 retinitis pigmentosa model. Brain Res 2021; 1768:147575. [PMID: 34242654 DOI: 10.1016/j.brainres.2021.147575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/20/2021] [Accepted: 06/26/2021] [Indexed: 11/18/2022]
Abstract
Environmental changes in the retina, including oxidative stress-induced cell death, influence photoreceptor degeneration in Retinitis Pigmentosa (RP). Previously, we tested and discovered that a cytoprotective chaperone protein, clusterin, produced robust preservation of rod photoreceptors of a rat autosomal dominant rhodopsin transgenic model of RP, S334ter-line3. To investigate the biochemical and molecular cytoprotective pathways of clusterin, we examined and compared a known source of cone cell death, nitric oxide (NO), observing nNOS expression using antibody against nNOS in RP retinas with intravitreal injections of saline, clusterin (10 μg/ml), or a non-isoform-selective NOS inhibitor (25 mM), L-NAME, or with an intraperitoneal injection (IP) of L-NAME (100 mg/kg). Rhodopsin-immunoreactive rod photoreceptor cells and nNOS-immunoreactive cells were quantified with immunohistochemistry in the presence or absence of L-NAME or clusterin, and the total nNOS retinal expression was determined by immunoblot analysis. In this study, the level of nNOS expression was significantly up-regulated postnatally (P) at P15 (P < 0.05), P30 (P < 0.001) and P60 (P < 0.0001) in RP retinas compared to normal controls. Clusterin treatment suppressed the up-regulated nNOS expression in RP retinas (P < 0.0001) and was enhanced in Type II amacrine cells. Additionally, IP injection of L-NAME at P15 prolonged rod survival in the later stage of RP retinas (P < 0.001). Conversely, rod survival in L-NAME-treated RP retinas was not equivalent to the rod survival number seen in clusterin-treated retinas, which suggests induction of nNOS expression in RP retinas and its reduction by clusterin is only partly responsible for the rescue observed through the reduction of nNOS expression in S334ter-line3 rat retinas.
Collapse
Affiliation(s)
- Andrew Vargas
- Mary D. Allen Vision Research Laboratory, USC Roski Eye Institute, Department of Ophthalmology, Los Angeles, CA, United States
| | - Kyra L Yamamoto
- Mary D. Allen Vision Research Laboratory, USC Roski Eye Institute, Department of Ophthalmology, Los Angeles, CA, United States
| | - Cheryl Mae Craft
- Mary D. Allen Vision Research Laboratory, USC Roski Eye Institute, Department of Ophthalmology, Los Angeles, CA, United States; Department of Integrative Anatomical Sciences, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Eun-Jin Lee
- Mary D. Allen Vision Research Laboratory, USC Roski Eye Institute, Department of Ophthalmology, Los Angeles, CA, United States; Department of Ophthalmology, Stanford University, Palo Alto, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Palo Alto, CA, United States.
| |
Collapse
|
10
|
Xu K, Qian H, Zou M. Triamcinolone acetonide combined with aminoguanidine inhibits inflammation and oxidative stress, improves vascular endothelial and retinal function and reduces VEGF expression in diabetic retinopathy patients. Exp Ther Med 2020; 19:2519-2526. [PMID: 32256730 PMCID: PMC7086164 DOI: 10.3892/etm.2020.8478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/09/2020] [Indexed: 11/21/2022] Open
Abstract
Effects of triamcinolone acetonide (TA) combined with aminoguanidine (AG) on vascular endothelial function, vascular endothelial growth factor (VEGF) expression and retinal function in diabetic retinopathy (DR) patients were studied. A total of 100 patients with DR admitted to the Taizhou Second People's Hospital Affiliated to Yangzhou University from January 2016 to December 2018 were collected and randomly divided into observation group (n=50) and control group (n=50). The control group was only treated with TA (4 mg), while the observation group was additionally treated with AG (50 mg/kg). The therapeutic effect and adverse reactions were observed, and the changes in blood glucose (GLU), blood lipid, serum inflammatory cytokines and oxidative stress indexes before and after treatment were detected. Moreover, the serum VEGF level was examined, the central macular thickness (CMT), retinal neovascularization (RNV) leakage area and best corrected visual acuity (BCVA) of retinopathy were observed, and the vascular endothelial function indexes such as nitric oxide (NO), endothelin-1 (ET-1) and intercellular adhesion molecule-1 (ICAM-1) were detected. The therapeutic effect in the observation group was significantly better than that in control group (P<0.05). In observation group after treatment, the levels of GLU, total cholesterol (TC), triglyceride (TG), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), myeloperoxidase (MPO), malondialdehyde (MDA) and catalase (CAT) in the serum, VEGF, CMT and RNV, as well as NO, ET-1 and ICAM-1 were remarkably decreased (P<0.05), while those of superoxide dismutase (SOD) and BCVA were evidently increased (P<0.05). The changes were more significant at 3 months after treatment than those at 1 month. TA combined with AG can inhibit inflammation and oxidative stress, improve vascular endothelial function and retinal function and reduce VEGF expression in DR patients. The overall effect of the combined therapy is good, and its application is worth popularizing.
Collapse
Affiliation(s)
- Kai Xu
- Department of Ophthalmology, Taizhou Second People's Hospital Affiliated to Yangzhou University, Taizhou, Zhejiang 225500, P.R. China
| | - Hanliang Qian
- Department of Ophthalmology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Meibo Zou
- Department of Ophthalmology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| |
Collapse
|
11
|
Jacoby J, Nath A, Jessen ZF, Schwartz GW. A Self-Regulating Gap Junction Network of Amacrine Cells Controls Nitric Oxide Release in the Retina. Neuron 2018; 100:1149-1162.e5. [PMID: 30482690 PMCID: PMC6317889 DOI: 10.1016/j.neuron.2018.09.047] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/28/2018] [Accepted: 09/25/2018] [Indexed: 01/31/2023]
Abstract
Neuromodulators regulate circuits throughout the nervous system, and revealing the cell types and stimulus conditions controlling their release is vital to understanding their function. The effects of the neuromodulator nitric oxide (NO) have been studied in many circuits, including in the vertebrate retina, where it regulates synaptic release, gap junction coupling, and blood vessel dilation, but little is known about the cells that release NO. We show that a single type of amacrine cell (AC) controls NO release in the inner retina, and we report its light responses, electrical properties, and calcium dynamics. We discover that this AC forms a dense gap junction network and that the strength of electrical coupling in the network is regulated by light through NO. A model of the network offers insights into the biophysical specializations leading to auto-regulation of NO release within the network.
Collapse
Affiliation(s)
- Jason Jacoby
- Department of Ophthalmology, Northwestern University, Chicago, IL, USA
| | - Amurta Nath
- Interdepartmental Neuroscience Program, Northwestern University, Chicago, IL, USA; Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, USA
| | - Zachary F Jessen
- Medical Scientist Training Program, Northwestern University, Chicago, IL, USA
| | - Gregory W Schwartz
- Department of Ophthalmology, Northwestern University, Chicago, IL, USA; Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
12
|
Dunn VK, Gleason E. Inhibition of endocytosis suppresses the nitric oxide-dependent release of Cl- in retinal amacrine cells. PLoS One 2018; 13:e0201184. [PMID: 30044876 PMCID: PMC6059450 DOI: 10.1371/journal.pone.0201184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/10/2018] [Indexed: 11/18/2022] Open
Abstract
Our lab has previously shown that nitric oxide (NO) can alter the synaptic response properties of amacrine cells by releasing Cl- from internal acidic compartments. This alteration in the Cl- gradient brings about a positive shift in the reversal potential of the GABA-gated current, which can convert inhibitory synapses into excitatory synapses. Recently, we have shown that the cystic fibrosis transmembrane regulator (CFTR) Cl- channel is involved in the Cl- release. Here, we test the hypothesis that (acidic) synaptic vesicles are a source of NO-releasable Cl- in chick retinal amacrine cells. If SVs are a source of Cl-, then depleting synaptic vesicles should decrease the nitric oxide-dependent shift in the reversal potential of the GABA-gated current. The efficacy of four inhibitors of dynamin (dynasore, Dyngo 4a, Dynole 34-2, and MiTMAB) were evaluated. In order to deplete synaptic vesicles, voltage-steps were used to activate V-gated Ca2+ channels and stimulate the synaptic vesicle cycle either under control conditions or after treatment with the dynamin inhibitors. Voltage-ramps were used to measure the NO-dependent shift in the reversal potential of the GABA-gated currents under both conditions. Our results reveal that activating the synaptic vesicle cycle in the presence of dynasore or Dyngo 4a blocked the NO-dependent shift in EGABA. However, we also discovered that some dynamin inhibitors reduced Ca2+ signaling and L-type Ca2+ currents. Conversely, dynasore also increased neurotransmitter release at autaptic sites. To further resolve the mechanism underlying the inhibition of the NO-dependent shift in the reversal potential for the GABA-gated currents, we also tested the effects of the clathrin assembly inhibitor Pitstop 2 and found that this compound also inhibited the shift. These data provide evidence that dynamin inhibitors have multiple effects on amacrine cell synaptic transmission. These data also suggest that inhibition of endocytosis disrupts the ability of NO to elicit Cl- release from internal stores which may in part be due to depletion of synaptic vesicles.
Collapse
Affiliation(s)
- Vernon K. Dunn
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Evanna Gleason
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States of America
| |
Collapse
|
13
|
Maddox JW, Khorsandi N, Gleason E. TRPC5 is required for the NO-dependent increase in dendritic Ca 2+ and GABA release from chick retinal amacrine cells. J Neurophysiol 2017; 119:262-273. [PMID: 28978766 DOI: 10.1152/jn.00500.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
GABAergic signaling from amacrine cells (ACs) is a fundamental aspect of visual signal processing in the inner retina. We have previously shown that nitric oxide (NO) can elicit release of GABA independently from activation of voltage-gated Ca2+ channels in cultured retinal ACs. This voltage-independent quantal GABA release relies on a Ca2+ influx mechanism with pharmacological characteristics consistent with the involvement of the transient receptor potential canonical (TRPC) channels TRPC4 and/or TRPC5. To determine the identity of these channels, we evaluated the ability of NO to elevate dendritic Ca2+ and to stimulate GABA release from cultured ACs under conditions known to alter the function of TRPC4 and 5. We found that these effects of NO are phospholipase C dependent, have a biphasic dependence on La3+, and are unaffected by moderate concentrations of the TRPC4-selective antagonist ML204. Together, these results suggest that NO promotes GABA release by activating TRPC5 channels in AC dendrites. To confirm a role for TRPC5, we knocked down the expression of TRPC5 using CRISPR/Cas9-mediated gene knockdown and found that both the NO-dependent Ca2+ elevations and increase in GABA release are dependent on the expression of TRPC5. These results demonstrate a novel NO-dependent mechanism for regulating neurotransmitter output from retinal ACs. NEW & NOTEWORTHY Elucidating the mechanisms regulating GABAergic synaptic transmission in the inner retina is key to understanding the flexibility of retinal ganglion cell output. Here, we demonstrate that nitric oxide (NO) can activate a transient receptor potential canonical 5 (TRPC5)-mediated Ca2+ influx, which is sufficient to drive vesicular GABA release from retinal amacrine cells. This NO-dependent mechanism can bypass the need for depolarization and may have an important role in processing the visual signal by enhancing retinal amacrine cell GABAergic inhibitory output.
Collapse
Affiliation(s)
- J Wesley Maddox
- Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| | - Nikka Khorsandi
- Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| | - Evanna Gleason
- Department of Biological Sciences, Louisiana State University , Baton Rouge, Louisiana
| |
Collapse
|
14
|
Retinal exposure to high glucose condition modifies the GABAergic system: Regulation by nitric oxide. Exp Eye Res 2017; 162:116-125. [PMID: 28734674 DOI: 10.1016/j.exer.2017.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/16/2017] [Accepted: 07/18/2017] [Indexed: 11/20/2022]
Abstract
Diabetic retinopathy is a severe retinal complication that diabetic patients are susceptible to present. Although this disease is currently characterized as a microvascular disease, there is growing evidence that neural changes occur and maybe precede vascular impairments. Using chicken retina, an avascular tissue with no direct contact with blood vessels and neural retina, this study aimed to evaluate the influence of acute exposure to high glucose concentration in the retinal GABAergic system, and the role of nitric oxide (NO) in this modulation. Therefore, in ex vivo experiments, retinas were incubated in control (10 mM glucose) or high glucose condition (35 mM) for 30 min. By using DAF-FM to evaluate NO production, it was possible to show that high glucose (HG) significantly increased NO levels in the outer nuclear layer, inner nuclear layer (outer and inner portion), and inner plexiform layer. It was also observed that HG increased GABA immunoreactivity (IR) in amacrine and horizontal cells. HG did not change glutamic acid decarboxylase-IR, whereas it decreased GABA Transporter (GAT) 1-IR and increased GAT-3-IR. The co-treatment with 7-NI, an inhibitor of neuronal nitric oxide synthase (nNOS), blocked all changes stimulated by HG exposure. The concomitant exposure with SNAP-5114, a GAT-2/3 inhibitor, blocked the increase in GABA-IR caused by HG incubation. Therefore, our data suggest that hyperglycemia induces GABA accumulation in the cytosol by modulating GABA transporters. This response is dependent on NO production and signaling.
Collapse
|
15
|
Agurto A, Vielma AH, Cadiz B, Couve E, Schmachtenberg O. NO signaling in retinal bipolar cells. Exp Eye Res 2017; 161:30-35. [PMID: 28579034 DOI: 10.1016/j.exer.2017.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/28/2017] [Accepted: 05/30/2017] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) is a neuromodulator involved in physiological and pathological processes in the retina. In the inner retina, a subgroup of amacrine cells have been shown to synthesize NO, but bipolar cells remain controversial as NO sources. This study correlates NO synthesis in dark-adapted retinas, through labeling with the NO marker DAF-FM, with neuronal nitric oxide synthase (nNOS) and inducible NOS expression, and presence of the NO receptor soluble guanylate cyclase in bipolar cells. NO containing bipolar cells were morphologically identified by dialysis of DAF fluorescent cells with intracellular dyes, or by DAF labeling followed by immunohistochemistry for nNOS and other cellular markers. DAF fluorescence was observed in all types of bipolar cells that could be identified, but the most intense DAF fluorescence was observed in bipolar cells with severed processes, supporting pathological NO signaling. Among nNOS expressing bipolar cells, type 9 was confirmed unequivocally, while types 2, 3a, 3b, 4, 5, 7, 8 and the rod bipolar cell were devoid of this enzyme. These results establish specific bipolar cell types as NO sources in the inner retina, and support the involvement of NO signaling in physiological and pathological processes in the inner retina.
Collapse
Affiliation(s)
- A Agurto
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - A H Vielma
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - B Cadiz
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Instituto de Biología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - E Couve
- Instituto de Biología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - O Schmachtenberg
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
16
|
Maddox JW, Gleason E. Nitric oxide promotes GABA release by activating a voltage-independent Ca 2+ influx pathway in retinal amacrine cells. J Neurophysiol 2017; 117:1185-1199. [PMID: 28053242 DOI: 10.1152/jn.00803.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/30/2016] [Accepted: 12/30/2016] [Indexed: 11/22/2022] Open
Abstract
Retinal amacrine cells express nitric oxide (NO) synthase and produce NO, making NO available to regulate the function of amacrine cells. Here we test the hypothesis that NO can alter the GABAergic synaptic output of amacrine cells. We investigate this using whole cell voltage clamp recordings and Ca2+ imaging of cultured chick retinal amacrine cells. When recording from amacrine cells receiving synaptic input from other amacrine cells, we find that NO increases GABAergic spontaneous postsynaptic current (sPSC) frequency. This increase in sPSC frequency does not require the canonical NO receptor, soluble guanylate cyclase, or presynaptic action potentials. However, removal of extracellular Ca2+ and buffering of cytosolic Ca2+ both inhibit the response to NO. In Ca2+ imaging experiments, we confirm that NO increases cytosolic Ca2+ in amacrine cell processes by activating a Ca2+ influx pathway. Neither the increase in sPSC frequency nor the cytosolic Ca2+ elevations are dependent upon Ca2+ release from stores. NO also enhances evoked GABAergic responses. Because voltage-gated Ca2+ channel function is not altered by NO, the increased evoked response is likely due to the combined effect of voltage-dependent Ca2+ influx adding to the NO-dependent, voltage-independent, Ca2+ influx. Insight into the identity of the Ca2+ influx pathway is provided by the transient receptor potential canonical (TRPC) channel inhibitor clemizole, which prevents the NO-dependent increase in sPSC frequency and cytosolic Ca2+ elevations. These data suggest that NO production in the inner retina will enhance Ca2+-dependent GABA release from amacrine cells by activating TRPC channel(s).NEW & NOTEWORTHY Our research provides evidence that nitric oxide (NO) promotes GABAergic output from retinal amacrine cells by activating a likely transient receptor potential canonical-mediated Ca2+ influx pathway. This NO-dependent mechanism promoting GABA release can be voltage independent, suggesting that, in the retina, local NO production can bypass the formal retinal circuitry and increase local inhibition.
Collapse
Affiliation(s)
- J Wesley Maddox
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - Evanna Gleason
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
17
|
Gupta V, Gupta VB, Chitranshi N, Gangoda S, Vander Wall R, Abbasi M, Golzan M, Dheer Y, Shah T, Avolio A, Chung R, Martins R, Graham S. One protein, multiple pathologies: multifaceted involvement of amyloid β in neurodegenerative disorders of the brain and retina. Cell Mol Life Sci 2016; 73:4279-4297. [PMID: 27333888 PMCID: PMC11108534 DOI: 10.1007/s00018-016-2295-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 01/18/2023]
Abstract
Accumulation of amyloid β (Aβ) and its aggregates in the ageing central nervous system is regarded synonymous to Alzheimer's disease (AD) pathology. Despite unquestionable advances in mechanistic and diagnostic aspects of the disease understanding, the primary cause of Aβ accumulation as well as its in vivo roles remains elusive; nonetheless, the majority of the efforts to address pathological mechanisms for therapeutic development are focused towards moderating Aβ accumulation in the brain. More recently, Aβ deposition has been identified in the eye and is linked with distinct age-related diseases including age-related macular degeneration, glaucoma as well as AD. Awareness of the Aβ accumulation in these markedly different degenerative disorders has led to an increasing body of work exploring overlapping mechanisms, a prospective biomarker role for Aβ and the potential to use retina as a model for brain related neurodegenerative disorders. Here, we present an integrated view of current understanding of the retinal Aβ deposition discussing the accumulation mechanisms, anticipated impacts and outlining ameliorative approaches that can be extrapolated to the retina for potential therapeutic benefits. Further longitudinal investigations in humans and animal models will determine retinal Aβ association as a potential pathognomonic, diagnostic or prognostic biomarker.
Collapse
Affiliation(s)
- Vivek Gupta
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Veer B Gupta
- School of Medical Sciences, Edith Cowan University, Perth, Australia.
| | - Nitin Chitranshi
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Sumudu Gangoda
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Roshana Vander Wall
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Mojdeh Abbasi
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Mojtaba Golzan
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Yogita Dheer
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Tejal Shah
- School of Medical Sciences, Edith Cowan University, Perth, Australia
| | - Alberto Avolio
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Roger Chung
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Ralph Martins
- School of Medical Sciences, Edith Cowan University, Perth, Australia
| | - Stuart Graham
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
- Save Sight Institute, Sydney University, Sydney, Australia
| |
Collapse
|
18
|
Rey-Funes M, Larrayoz IM, Fernández JC, Contartese DS, Rolón F, Inserra PIF, Martínez-Murillo R, López-Costa JJ, Dorfman VB, Martínez A, Loidl CF. Methylene blue prevents retinal damage in an experimental model of ischemic proliferative retinopathy. Am J Physiol Regul Integr Comp Physiol 2016; 310:R1011-9. [PMID: 26984891 DOI: 10.1152/ajpregu.00266.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 03/11/2016] [Indexed: 11/22/2022]
Abstract
Perinatal asphyxia induces retinal lesions, generating ischemic proliferative retinopathy, which may result in blindness. Previously, we showed that the nitrergic system was involved in the physiopathology of perinatal asphyxia. Here we analyze the application of methylene blue, a well-known soluble guanylate cyclase inhibitor, as a therapeutic strategy to prevent retinopathy. Male rats (n = 28 per group) were treated in different ways: 1) control group comprised born-to-term animals; 2) methylene blue group comprised animals born from pregnant rats treated with methylene blue (2 mg/kg) 30 and 5 min before delivery; 3) perinatal asphyxia (PA) group comprised rats exposed to perinatal asphyxia (20 min at 37°C); and 4) methylene blue-PA group comprised animals born from pregnant rats treated with methylene blue (2 mg/kg) 30 and 5 min before delivery, and then the pups were subjected to PA as above. For molecular studies, mRNA was obtained at different times after asphyxia, and tissue was collected at 30 days for morphological and biochemical analysis. Perinatal asphyxia produced significant gliosis, angiogenesis, and thickening of the inner retina. Methylene blue treatment reduced these parameters. Perinatal asphyxia resulted in a significant elevation of the nitrergic system as shown by NO synthase (NOS) activity assays, Western blotting, and (immuno)histochemistry for the neuronal isoform of NOS and NADPH-diaphorase activity. All these parameters were also normalized by the treatment. In addition, methylene blue induced the upregulation of the anti-angiogenic peptide, pigment epithelium-derived factor. Application of methylene blue reduced morphological and biochemical parameters of retinopathy. This finding suggests the use of methylene blue as a new treatment to prevent or decrease retinal damage in the context of ischemic proliferative retinopathy.
Collapse
Affiliation(s)
- Manuel Rey-Funes
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Ignacio M Larrayoz
- Angiogenesis Study Group, Center for Biomedical Research of La Rioja, Logroño, Spain;
| | - Juan C Fernández
- Primera Cátedra de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniela S Contartese
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Federico Rolón
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Pablo I F Inserra
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico, Universidad Maimónides, Buenos Aires, Argentina
| | - Ricardo Martínez-Murillo
- Neurovascular Research Group, Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, Consejo Superior Investigaciones Científicas, Madrid, Spain; and
| | - Juan J López-Costa
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Verónica B Dorfman
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico, Universidad Maimónides, Buenos Aires, Argentina
| | - Alfredo Martínez
- Angiogenesis Study Group, Center for Biomedical Research of La Rioja, Logroño, Spain
| | - César F Loidl
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina; Laboratorio de Neurociencia, Facultad de Ciencias Médicas, Universidad Católica de Cuyo, San Juan, Argentina
| |
Collapse
|
19
|
Tooker RE, Vigh J. Light-evoked S-nitrosylation in the retina. J Comp Neurol 2015; 523:2082-110. [PMID: 25823749 DOI: 10.1002/cne.23780] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/05/2015] [Accepted: 03/23/2015] [Indexed: 01/14/2023]
Abstract
Nitric oxide (NO) synthesis in the retina is triggered by light stimulation. NO has been shown to modulate visual signal processing at multiple sites in the vertebrate retina, via activation of the most sensitive target of NO signaling, soluble guanylate cyclase. NO can also alter protein structure and function and exert biological effects directly by binding to free thiol groups of cysteine residues in a chemical reaction called S-nitrosylation. However, in the central nervous system, including the retina, this reaction has not been considered to be significant under physiological conditions. Here we provide immunohistochemical evidence for extensive S-nitrosylation that takes place in the goldfish and mouse retinas under physiologically relevant light intensities, in an intensity-dependent manner, with a strikingly similar pattern in both species. Pretreatment with N-ethylmaleimide (NEM), which occludes S-nitrosylation, or with 1-(2-trifluromethylphenyl)imidazole (TRIM), an inhibitor of neuronal NO synthase, eliminated the light-evoked increase in S-nitrosylated protein immunofluorescence (SNI) in the retinas of both species. Similarly, light did not increase SNI, above basal levels, in retinas of transgenic mice lacking neuronal NO synthase. Qualitative analysis of the light-adapted mouse retina with mass spectrometry revealed more than 300 proteins that were S-nitrosylated upon illumination, many of which are known to participate directly in retinal signal processing. Our data strongly suggest that in the retina light-evoked NO production leads to extensive S-nitrosylation and that this process is a significant posttranslational modification affecting a wide range of proteins under physiological conditions.
Collapse
Affiliation(s)
- Ryan E Tooker
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, 80523
| | - Jozsef Vigh
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, 80523
| |
Collapse
|
20
|
Walter LT, Higa GSV, Schmeltzer C, Sousa E, Kinjo ER, Rüdiger S, Hamassaki DE, Cerchiaro G, Kihara AH. Functional regulation of neuronal nitric oxide synthase expression and activity in the rat retina. Exp Neurol 2014; 261:510-7. [PMID: 25116452 DOI: 10.1016/j.expneurol.2014.07.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/16/2014] [Accepted: 07/29/2014] [Indexed: 11/19/2022]
Abstract
In the nervous system within physiological conditions, nitric oxide (NO) production depends on the activity of nitric oxide synthases (NOSs), and particularly on the expression of the neuronal isoform (nNOS). In the sensory systems, the role of NO is poorly understood. In this study, we identified nNOS-positive cells in the inner nuclear layer (INL) of the rat retina, with distinct characteristics such as somata size, immunolabeling level and location. Employing mathematical cluster analysis, we determined that nNOS amacrine cells are formed by two distinct populations. We next investigated the molecular identity of these cells, which did not show colocalization with calbindin (CB), choline acetyltransferase (ChAT), parvalbumin (PV) or protein kinase C (PKC), and only partial colocalization with calretinin (CR), revealing the accumulation of nNOS in specific amacrine cell populations. To access the functional, circuitry-related roles of these cells, we performed experiments after adaptation to different ambient light conditions. After 24h of dark-adaptation, we detected a subtle, yet statistically significant decrease in nNOS transcript levels, which returned to steady-state levels after 24h of normal light-dark cycle, revealing that nNOS expression is governed by ambient light conditions. Employing electron paramagnetic resonance (EPR), we demonstrated that dark-adaptation decreases NO production in the retina. Furthermore, nNOS accumulation changed in the dark-adapted retinas, with a general reduction in the inner plexiform layer. Finally, computational analysis based on clustering techniques revealed that dark-adaptation differently affected both types of nNOS-positive amacrine cells. Taken together, our data disclosed functional regulation of nNOS expression and activity, disclosing new circuitry-related roles of nNOS-positive cells. More importantly, this study indicated unsuspected roles for NO in the sensory systems, particularly related to adaptation to ambient demands.
Collapse
Affiliation(s)
- Lais Takata Walter
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Brazil
| | - Guilherme Shigueto Vilar Higa
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Brazil; Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| | | | - Erica Sousa
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Brazil
| | - Erika Reime Kinjo
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Brazil
| | - Sten Rüdiger
- Institute of Physics, Humboldt University at Berlin, Germany
| | - Dânia Emi Hamassaki
- Departamento de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil
| | - Giselle Cerchiaro
- Núcleo de Cognição e Sistemas Complexos, Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Brazil
| | - Alexandre Hiroaki Kihara
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Brazil; Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil.
| |
Collapse
|
21
|
Dhingra A, Tummala SR, Lyubarsky A, Vardi N. PDE9A is expressed in the inner retina and contributes to the normal shape of the photopic ERG waveform. Front Mol Neurosci 2014; 7:60. [PMID: 25018695 PMCID: PMC4073215 DOI: 10.3389/fnmol.2014.00060] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 06/09/2014] [Indexed: 11/19/2022] Open
Abstract
The ubiquitous second messenger cGMP is synthesized by guanylyl cyclase and hydrolyzed by phosphodiesterase (PDE). cGMP mediates numerous signaling pathways in multiple tissues. In the retina, cGMP regulates signaling in nearly every cell class including photoreceptors, bipolar cells, amacrine cells, and ganglion cells. In order to understand the specific role of cGMP and its regulating enzymes in different cell types, it is first necessary to localize these components and dissect their influence on the circuits. Here we tested the contribution of PDE9A to retinal processing by recording the electroretinograms (ERG) of PDE9A™/™ (KO) mice and by localizing the enzyme. We found that while the scotopic ERG of KO was the same as that of wild type (WT) in both amplitude and kinetics, the photopic ERG was greatly affected. The greatest effect was on the recovery of the b-wave; the falling phase and the b-wave duration were significantly longer in the KO mice for all photopic stimuli (UV, green, or saturating white flashes). The rising phase was slower in KO than in WT for UV and green stimuli. For certain stimuli, amplitudes of both the a- and b-waves were smaller than in WT. Using Lac-Z expression in KO retinas as a reporter for PDE9A expression pattern, we found that PDE9A is localized to GABA-positive and GABA-negative amacrine cells, and likely also to certain types of ganglion cells. Our results indicate that PDE9A, by controlling the level of cGMP, modulates inhibitory processes within the cone pathway. We speculate that these circuits involve NO/cGMP signaling pathways.
Collapse
Affiliation(s)
- Anuradha Dhingra
- Retina Lab, Department of Neuroscience, University of Pennsylvania Philadelphia, PA, USA
| | - Shanti R Tummala
- Retina Lab, Department of Neuroscience, University of Pennsylvania Philadelphia, PA, USA
| | - Arkady Lyubarsky
- Department of Ophthalmology, University of Pennsylvania Philadelphia, PA, USA
| | - Noga Vardi
- Retina Lab, Department of Neuroscience, University of Pennsylvania Philadelphia, PA, USA
| |
Collapse
|
22
|
Tooker RE, Lipin MY, Leuranguer V, Rozsa E, Bramley JR, Harding JL, Reynolds MM, Vigh J. Nitric oxide mediates activity-dependent plasticity of retinal bipolar cell output via S-nitrosylation. J Neurosci 2013; 33:19176-93. [PMID: 24305814 PMCID: PMC3850041 DOI: 10.1523/jneurosci.2792-13.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/23/2013] [Accepted: 10/31/2013] [Indexed: 11/21/2022] Open
Abstract
Coding a wide range of light intensities in natural scenes poses a challenge for the retina: adaptation to bright light should not compromise sensitivity to dim light. Here we report a novel form of activity-dependent synaptic plasticity, specifically, a "weighted potentiation" that selectively increases output of Mb-type bipolar cells in the goldfish retina in response to weak inputs but leaves the input-output ratio for strong stimuli unaffected. In retinal slice preparation, strong depolarization of bipolar terminals significantly lowered the threshold for calcium spike initiation, which originated from a shift in activation of voltage-gated calcium currents (ICa) to more negative potentials. The process depended upon glutamate-evoked retrograde nitric oxide (NO) signaling as it was eliminated by pretreatment with an NO synthase blocker, TRIM. The NO-dependent ICa modulation was cGMP independent but could be blocked by N-ethylmaleimide (NEM), indicating that NO acted via an S-nitrosylation mechanism. Importantly, the NO action resulted in a weighted potentiation of Mb output in response to small (≤-30 mV) depolarizations. Coincidentally, light flashes with intensity ≥ 2.4 × 10(8) photons/cm(2)/s lowered the latency of scotopic (≤ 2.4 × 10(8) photons/cm(2)/s) light-evoked calcium spikes in Mb axon terminals in an NEM-sensitive manner, but light responses above cone threshold (≥ 3.5 × 10(9) photons/cm(2)/s) were unaltered. Under bright scotopic/mesopic conditions, this novel form of Mb output potentiation selectively amplifies dim retinal inputs at Mb → ganglion cell synapses. We propose that this process might counteract decreases in retinal sensitivity during light adaptation by preventing the loss of visual information carried by dim scotopic signals.
Collapse
Affiliation(s)
| | | | | | - Eva Rozsa
- Department of Biomedical Sciences and
| | | | | | - Melissa M. Reynolds
- Department of Chemistry, Colorado State University, Ft. Collins, Colorado 80523
| | | |
Collapse
|
23
|
Lima MG, Maximino C, Matos Oliveira KR, Brasil A, Crespo-Lopez ME, Batista EDJO, Rocha FADF, Picanço-Diniz DLW, Herculano AM. Nitric oxide as a regulatory molecule in the processing of the visual stimulus. Nitric Oxide 2013; 36:44-50. [PMID: 24275015 DOI: 10.1016/j.niox.2013.10.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 10/18/2013] [Accepted: 10/30/2013] [Indexed: 01/02/2023]
Abstract
Nitric oxide (NO) is a highly reactive gas with considerable diffusion power that is produced pre- and post synaptically in the central nervous system (CNS). In the visual system, it is involved in the processing of the visual information from the retina to superior visual centers. In this review we discuss the main mechanisms through which nitric oxide acts, in physiological levels, on the retina, lateral geniculate nucleus (LGN) and primary visual cortex. In the retina, the cGMP-dependent nitric oxide activity initially amplifies the signal, subsequently increasing the inhibitory activity, suggesting that the signal is "filtered". In the thalamus, on dLGN, neuronal activity is amplified by NO derived from brainstem cholinergic cells, in a cGMP-independent mechanism; the result is the amplification of the signal arriving from retina. Finally, on the visual cortex (V1), NO acts through changes on the cGMP levels, increasing signal detection. These observations suggest that NO works like a filter, modulating the signal along the visual pathways.
Collapse
Affiliation(s)
- Monica Gomes Lima
- Laboratory of Neuroendocrinology, Biological Sciences Institute, Federal University of Pará, Av. Augusto Correa, 01 Guamá, Belém, Pará 66075-110, Brazil.
| | - Caio Maximino
- Laboratory of Neuroendocrinology, Biological Sciences Institute, Federal University of Pará, Av. Augusto Correa, 01 Guamá, Belém, Pará 66075-110, Brazil; Zebrafish Neuroscience Research Consortium (ZNRC) Slidell, Louisiana 70458, USA.
| | - Karen Renata Matos Oliveira
- Laboratory of Neuroendocrinology, Biological Sciences Institute, Federal University of Pará, Av. Augusto Correa, 01 Guamá, Belém, Pará 66075-110, Brazil.
| | - Alódia Brasil
- Laboratory of Neuroendocrinology, Biological Sciences Institute, Federal University of Pará, Av. Augusto Correa, 01 Guamá, Belém, Pará 66075-110, Brazil.
| | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Biological Sciences Institute, Federal University of Pará, Av. Augusto Correa, 01 Guamá, Belém, Pará CEP 66075-110, Brazil.
| | - Evander de Jesus Oliveira Batista
- Laboratory of Neuroendocrinology, Biological Sciences Institute, Federal University of Pará, Av. Augusto Correa, 01 Guamá, Belém, Pará 66075-110, Brazil.
| | - Fernando Allan de Farias Rocha
- Laboratory of Neuroscience Dr. Eduardo Oswaldo Cruz, Biological Sciences Institute, Federal University of Pará, Av. Augusto Correa, 01 Guamá, Belém, Pará CEP 66075-110, Brazil
| | - Domingos Luiz Wanderley Picanço-Diniz
- Nucleus Oriximiná, Federal University of Western Pará, University Campus Oriximiná, Rodovia PA-254, n° 257 Bairro Santíssimo, Oriximiná, Pará CEP 68270-000, Brazil.
| | - Anderson Manoel Herculano
- Laboratory of Neuroendocrinology, Biological Sciences Institute, Federal University of Pará, Av. Augusto Correa, 01 Guamá, Belém, Pará 66075-110, Brazil; Zebrafish Neuroscience Research Consortium (ZNRC) Slidell, Louisiana 70458, USA.
| |
Collapse
|
24
|
Guthrie MJ, Kang-Mieler JJ. Dual electroretinogram/nitric oxide carbon fiber microelectrode for direct measurement of nitric oxide in the in vivo retina. IEEE Trans Biomed Eng 2013; 61:611-9. [PMID: 24043366 DOI: 10.1109/tbme.2013.2281541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nitric oxide (NO) plays an important physiological role in normal and pathological retinas. Intraretinal NO concentrations have not been directly measured due to lack of NO electrodes capable of determining their location in the retina. The microelectrodes described here allow recording of the intraretinal electroretinogram (ERG) and NO concentration from the same location, with ERGs used to determine retinal depth. Double-barreled electrodes were constructed with one barrel serving as a reference/voltage recording barrel and the other containing a Nafion-coated carbon fiber used to detect NO amperometrically. Nafion coating imparted a high selectivity for NO versus ascorbic acid (2000:1). In vivo rodent experiments demonstrated that the electrodes could record intraretinal ERGs and NO current with minimal retinal thickness deformation (9%), allowing for retinal NO depth profile measurements. Comparison of NO depth profiles under control conditions and under nitric oxide synthase (NOS) inhibition by 5 mM L-NG-Nitroarginine methyl ester (L-NAME) verified that the recorded current was attributable to NO. NO concentrations from control profiles ( n = 4) were 2.37 ± 0.34 μM at the choroid and 1.12 ± 0.14 μM at the retinal surface. NO concentrations from L-NAME profiles ( n = 4) were significantly lower at 0.83 ± 0.15 μM at the choroid ( p = 0.006) and 0.27 ± 0.04 μM at the retinal surface ( p = 0.001). Localized regions of increased NO (100-400 nM) were seen in the inner retina under control conditions but not after L-NAME. The dual ERG-NO electrode may be a valuable tool in evaluating the role of NO in normal and diseased retinas.
Collapse
|
25
|
Nitric oxide production and the expression of two nitric oxide synthases in the avian retina. Vis Neurosci 2013; 30:91-103. [PMID: 23721886 DOI: 10.1017/s0952523813000126] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nitric oxide (NO) is known to exert multiple effects on the function of many retinal neurons and their synapses. Therefore, it is equally important to understand the potential sources of NO within the retina. To explore this, we employ a combination of 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF-FM) based NO detection and immunohistochemistry for the NO synthetic enzymes, neuronal and endothelial nitric oxide synthase (nNOS and eNOS). We find DAF signals in photoreceptors, horizontal cells, amacrine cells, efferent synapses, Müller cells, and cells in the ganglion cell layer (GCL). nNOS immunoreactivity was consistent with the DAF signal with the exception that horizontal cells and Müller cells were not clearly labeled. eNOS-like immunoreactivity (eNOS-LI) was more widespread with photoreceptors, horizontal cells, occasional bipolar cells, amacrine cells, Müller cells, and cells in the GCL all showing labeling. Double labeling with antibodies raised against calretinin, syntaxin, and glutamine synthetase confirmed that horizontal cells, amacrine cells, and Müller cells (respectively) were expressing eNOS-LI. Although little or no nNOS labeling is observed in horizontal cells or Müller cells, the expression of eNOS-LI is consistent with the ability of these cells to produce NO. Together these results suggest that the capability to produce NO is widespread in the chicken retina. We propose that multiple forms of regulation for nNOS and eNOS play a role in the patterning of NO production in the chicken retina.
Collapse
|
26
|
Blom J, Giove T, Deshpande M, Eldred WD. Characterization of nitric oxide signaling pathways in the mouse retina. J Comp Neurol 2013; 520:4204-17. [PMID: 22592770 DOI: 10.1002/cne.23148] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Nitric oxide (NO) is a gaseous neuromodulator with physiological functions in every retinal cell type. NO is synthesized by several nitric oxide synthases (NOS) and often functions through its second messenger, cyclic guanosine monophosphate (cGMP), and protein kinase G (PKG). This study combined NO imaging, immunocytochemistry, biochemistry, and molecular biology to localize NO and its downstream signaling pathways in the mouse retina. Neuronal NOS (nNOS) was localized primarily in puncta in the inner plexiform layer, in amacrine cells, and in somata in the ganglion cell layer. Endothelial NOS was in blood vessels. Light-stimulated NO production imaged with diaminofluorescein was present in somata in the inner nuclear layer and in synaptic boutons in the inner plexiform layer. The downstream target of NO, soluble guanylate cyclase (sGC), was in somata in the inner and outer nuclear layers and in both plexiform layers. Cyclic GMP immunocytochemistry was used functionally to localize sGC that was activated by an NO donor in amacrine, bipolar, and ganglion cells. Cyclic GMP-dependent protein kinase (PKG) Iα was found in bipolar cells, ganglion cells, and both plexiform layers, whereas PKG II was found in the outer plexiform layer, amacrine cells, and somata in the ganglion cell layer. This study shows that the NO/cGMP/PKG signaling pathway is functional and widely distributed in specific cell types in the outer and inner mouse retina. A better understanding of these signaling pathways in normal retina will provide a firm basis for targeting their roles in retinal pathology.
Collapse
Affiliation(s)
- Jan Blom
- Department of Biology, Boston University, Boston, Massachusetts 02215, USA
| | | | | | | |
Collapse
|
27
|
Gasulla J, Beltrán González AN, Calvo DJ. Nitric oxide potentiation of the homomeric ρ1 GABA(C) receptor function. Br J Pharmacol 2013; 167:1369-77. [PMID: 22747884 DOI: 10.1111/j.1476-5381.2012.02087.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND PURPOSE NO is a highly diffusible and reactive gas produced in the nervous system, which acts as a neuronal signal mediating physiological or pathological mechanisms. NO can modulate the activity of neurotransmitter receptors and ion channels, including NMDA and GABA(A) receptors. In the present work, we examined whether GABA(C) receptor function can also be regulated by NO. EXPERIMENTAL APPROACH Homomeric ρ1 GABA(C) receptors were expressed in oocytes and GABA-evoked responses electrophysiologically recorded in the presence or absence of the NO donor DEA. Chemical protection of cysteines by selective sulfhydryl reagents and site-directed mutagenesis were used to determine the protein residues involved in the actions of NO. KEY RESULTS GABAρ1 receptor responses were significantly enhanced in a dose-dependent, fast and reversible manner by DEA and the specific NO scavenger CPTIO prevented these potentiating effects. The ρ1 subunits contain only three cysteine residues, two extracellular at the Cys-loop (C177 and C191) and one intracellular (C364). Mutations of C177 and C191 render the ρ1 GABA receptors non-functional, but C364 can be safely exchanged by alanine (C364A). NEM, N-ethyl maleimide and (2-aminoethyl) methanethiosulfonate prevented the effects of DEA on GABAρ1 receptors. Meanwhile, the potentiating effects of DEA on mutant GABAρ1(C364A) receptors were similar to those observed on wild-type receptors. CONCLUSIONS AND IMPLICATIONS Our results suggest that the function of GABA(C) receptors can be enhanced by NO acting at the extracellular Cys-loop.
Collapse
Affiliation(s)
- J Gasulla
- Laboratorio de Neurobiología Celular y Molecular, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | | | | |
Collapse
|
28
|
Blom JJ, Giove TJ, Favazza TL, Akula JD, Eldred WD. Inhibition of the adrenomedullin/nitric oxide signaling pathway in early diabetic retinopathy. J Ocul Biol Dis Infor 2012; 4:70-82. [PMID: 23316263 DOI: 10.1007/s12177-011-9072-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 12/05/2011] [Indexed: 12/18/2022] Open
Abstract
The nitric oxide (NO) signaling pathway is integrally involved in visual processing and changes in the NO pathway are measurable in eyes of diabetic patients. The small peptide adrenomedullin (ADM) can activate a signaling pathway to increase the enzyme activity of neuronal nitric oxide synthase (nNOS). ADM levels are elevated in eyes of diabetic patients and therefore, ADM may play a role in the pathology of diabetic retinopathy. The goal of this research was to test the effects of inhibiting the ADM/NO signaling pathway in early diabetic retinopathy. Inhibition of this pathway decreased NO production in high-glucose retinal cultures. Treating diabetic mice with the PKC β inhibitor ruboxistaurin for 5 weeks lowered ADM mRNA levels and ADM-like immunoreactivity and preserved retinal function as assessed by electroretinography. The results of this study indicate that inhibiting the ADM/NO signaling pathway prevents neuronal pathology and functional losses in early diabetic retinopathy.
Collapse
Affiliation(s)
- Jan J Blom
- Department of Biology, Boston University, Boston, MA USA
| | | | | | | | | |
Collapse
|
29
|
Vielma AH, Retamal MA, Schmachtenberg O. Nitric oxide signaling in the retina: what have we learned in two decades? Brain Res 2011; 1430:112-25. [PMID: 22133309 DOI: 10.1016/j.brainres.2011.10.045] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 10/14/2011] [Accepted: 10/27/2011] [Indexed: 01/21/2023]
Abstract
Two decades after its first detection in the retina, nitric oxide (NO) continues to puzzle visual neuroscientists. While its liberation by photoreceptors remains controversial, recent evidence supports three subtypes of amacrine cells as main sources of NO in the inner retina. NO synthesis was shown to depend on light stimulation, and mounting evidence suggests that NO is a regulator of visual adaptation at different signal processing levels. NO modulates light responses in all retinal neuron classes, and specific ion conductances are activated by NO in rods, cones, bipolar and ganglion cells. Light-dependent gap junction coupling in the inner and outer plexiform layers is also affected by NO. The vast majority of these effects were shown to be mediated by activation of the NO receptor soluble guanylate cyclase and resultant cGMP elevation. This review analyzes the current state of knowledge on physiological NO signaling in the retina.
Collapse
Affiliation(s)
- Alex H Vielma
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | | | | |
Collapse
|
30
|
Narayanan SP, Suwanpradid J, Saul A, Xu Z, Still A, Caldwell RW, Caldwell RB. Arginase 2 deletion reduces neuro-glial injury and improves retinal function in a model of retinopathy of prematurity. PLoS One 2011; 6:e22460. [PMID: 21811615 PMCID: PMC3141070 DOI: 10.1371/journal.pone.0022460] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 06/26/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Retinopathy of prematurity (ROP) is a major cause of vision impairment in low birth weight infants. While previous work has focused on defining the mechanisms of vascular injury leading to retinal neovascularization, recent studies show that neurons are also affected. This study was undertaken to determine the role of the mitochondrial arginine/ornithine regulating enzyme arginase 2 (A2) in retinal neuro-glial cell injury in the mouse model of ROP. METHODS AND FINDINGS Studies were performed using wild type (WT) and A2 knockout (A2-/-) mice exposed to Oxygen Induced Retinopathy (OIR). Neuronal injury and apoptosis were assessed using immunohistochemistry, TUNEL (terminal deoxynucleotidyl transferase dUTP nick end) labeling and Western blotting. Electroretinography (ERG) was used to assess retinal function. Neuro-glial injury in WT ROP mice was evident by TUNEL labeling, retinal thinning, decreases in number of rod bipolar cells and glial cell activation as compared with room air controls. Significant reduction in numbers of TUNEL positive cells, inhibition of retinal thinning, preservation of the rod bipolar cells and prevention of glial activation were observed in the A2-/- retinas. Retinal function was markedly impaired in the WT OIR mice as shown by decreases in amplitude of the b-wave of the ERG. This defect was significantly reduced in A2-/- mice. Levels of the pro-apoptotic proteins p53, cleaved caspase 9, cytochrome C and the mitochondrial protein Bim were markedly increased in WT OIR retinas compared to controls, whereas the pro-survival Mitochondrial protein BCL-xl was reduced. These alterations were largely blocked in the A2-/- OIR retina. CONCLUSIONS Our data implicate A2 in neurodegeneration during ROP. Deletion of A2 significantly improves neuronal survival and function, possibly through the regulation of mitochondrial membrane permeability mediated apoptosis during retinal ischemia. These molecular events are associated with decreased activation of glial cells, suggesting a rescue effect on macroglia as well.
Collapse
Affiliation(s)
- Subhadra P. Narayanan
- Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia, United States of America
- Department of Cellular Biology and Anatomy, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Jutamas Suwanpradid
- Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Alan Saul
- Department of Ophthalmology, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Zhimin Xu
- Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Amber Still
- Department of Ophthalmology, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Robert W. Caldwell
- Department of Pharmacology and Toxicology, Georgia Health Sciences University, Augusta, Georgia, United States of America
| | - Ruth B. Caldwell
- Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia, United States of America
- Department of Cellular Biology and Anatomy, Georgia Health Sciences University, Augusta, Georgia, United States of America
- Department of Ophthalmology, Georgia Health Sciences University, Augusta, Georgia, United States of America
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
31
|
McMains E, Gleason E. Role of pH in a nitric oxide-dependent increase in cytosolic Cl- in retinal amacrine cells. J Neurophysiol 2011; 106:641-51. [PMID: 21593387 DOI: 10.1152/jn.00057.2011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Nitric oxide (NO) synthase-expressing neurons are found throughout the vertebrate retina. Previous work by our laboratory has shown that NO can transiently convert inhibitory GABAergic synapses onto cultured retinal amacrine cells into excitatory synapses by releasing Cl(-) from an internal store in the postsynaptic cell. The mechanism underlying this Cl(-) release is currently unknown. Because transport of Cl(-) across internal membranes can be coupled to proton flux, we asked whether protons could be involved in the NO-dependent release of internal Cl(-). Using pH imaging and whole cell voltage-clamp recording, we addressed the relationship between cytosolic pH and cytosolic Cl(-) in cultured retinal amacrine cells. We found that NO reliably produces a transient decrease in cytosolic pH. A physiological link between cytosolic pH and cytosolic Cl(-) was established by demonstrating that shifting cytosolic pH in the absence of NO altered cytosolic Cl(-) concentrations. Strong buffering of cytosolic pH limited the ability of NO to increase cytosolic Cl(-), suggesting that cytosolic acidification is involved in generating the NO-dependent elevation in cytosolic Cl(-). Furthermore, disruption of internal proton gradients also reduced the effects of NO on cytosolic Cl(-). Taken together, these results suggest a cytosolic environment where proton and Cl(-) fluxes are coupled in a dynamic and physiologically meaningful way.
Collapse
Affiliation(s)
- Emily McMains
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | | |
Collapse
|
32
|
Abstract
Using both NADPH diaphorase and anti-nNOS antibodies, we have identified-from retinal flatmounts-neuronal types in the inner retina of the chicken that are likely to be nitrergic. The two methods gave similar results and yielded a total of 15 types of neurons, comprising 9 amacrine cells, 5 ganglion cells, and 1 centrifugal midbrain neuron. Six of these 15 cell types are ubiquitously distributed, comprising 3 amacrine cells, 2 displaced ganglion cells, and a presumed orthotopic ganglion cell. The remaining nine cell types are regionally restricted within the retina. As previously reported, efferent fibers of midbrain neurons and their postsynaptic partners, the unusual axon-bearing target amacrine cells, are entirely confined to the ventral retina. Also confined to the ventral retina, though with somewhat different distributions, are the "bullwhip" amacrine cells thought to be involved in eye growth, an orthotopic ganglion cell, and two types of large axon-bearing amacrine cells whose dendrites and axons lie in stratum 1 of the inner plexiform layer (IPL). Intracellular fills of these two cell types showed that only a minority of otherwise morphologically indistinguishable neurons are nitrergic. Two amacrine cells that branch throughout the IPL are confined to an equatorial band, and one small-field orthotopic ganglion cell that branches in the proximal IPL is entirely dorsal. These findings suggest that the retina uses different processing on different regions of the visual image, though the benefit of this is presently obscure.
Collapse
|
33
|
Rey-Funes M, Ibarra ME, Dorfman VB, Serrano J, Fernández AP, Martínez-Murillo R, Martínez A, Coirini H, Rodrigo J, Loidl CF. Hypothermia prevents nitric oxide system changes in retina induced by severe perinatal asphyxia. J Neurosci Res 2011; 89:729-43. [DOI: 10.1002/jnr.22556] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 10/19/2010] [Accepted: 10/20/2010] [Indexed: 11/06/2022]
|
34
|
The light-induced reduction of horizontal cell receptive field size in the goldfish retina involves nitric oxide. Vis Neurosci 2011; 28:137-44. [PMID: 21324227 DOI: 10.1017/s0952523810000490] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Horizontal cells of the vertebrate retina have large receptive fields as a result of extensive gap junction coupling. Increased ambient illumination reduces horizontal cell receptive field size. Using the isolated goldfish retina, we have assessed the contribution of nitric oxide to the light-dependent reduction of horizontal cell receptive field size. Horizontal cell receptive field size was assessed by comparing the responses to centered spot and annulus stimuli and from the responses to translated slit stimuli. A period of steady illumination decreased the receptive field size of horizontal cells, as did treatment with the nitric oxide donor (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate (100 μM). Blocking the endogenous production of nitric oxide with the nitric oxide synthase inhibitor, N(G)-nitro-L-arginine methyl ester (1 mM), decreased the light-induced reduction of horizontal cell receptive field size. These findings suggest that nitric oxide is involved in light-induced reduction of horizontal cell receptive field size.
Collapse
|
35
|
Nitric oxide amplifies the rat electroretinogram. Exp Eye Res 2010; 91:700-9. [DOI: 10.1016/j.exer.2010.08.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 07/26/2010] [Accepted: 08/12/2010] [Indexed: 12/30/2022]
|
36
|
Pang JJ, Gao F, Wu SM. Light responses and morphology of bNOS-immunoreactive neurons in the mouse retina. J Comp Neurol 2010; 518:2456-74. [PMID: 20503422 DOI: 10.1002/cne.22347] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Nitric oxide (NO), produced by NO synthase (NOS), modulates the function of all retinal neurons and ocular blood vessels and participates in the pathogenesis of ocular diseases. To further understand the regulation of ocular NO release, we systematically studied the morphology, topography, and light responses of NOS-containing amacrine cells (NOACs) in dark-adapted mouse retina. Immunohistological staining for neuronal NOS (bNOS), combined with retrograde labeling of ganglion cells (GCs) with Neurobiotin (NB, a gap junction permeable dye) and Lucifer yellow (LY, a less permeable dye), was used to identify NOACs. The light responses of ACs were recorded under whole-cell voltage clamp conditions and cell morphology was examined with a confocal microscope. We found that in dark-adapted conditions bNOS-immunoreactivity (IR) was present primarily in the inner nuclear layer and the ganglion cell layer. bNOS-IR somas were negative for LY, thus they were identified as ACs; nearly 6% of the cells were labeled by NB but not by LY, indicating that they were dye-coupled with GCs. Three morphological subtypes of NOACs (NI, NII, and displaced) were identified. The cell density, intercellular distance, and the distribution of NOACs were studied in whole retinas. Light evoked depolarizing highly sensitive ON-OFF responses in NI cells and less sensitive OFF responses in NII cells. Frequent (1-2 Hz) or abrupt change of light intensity evoked larger peak responses. The possibility for light to modify NO release from NOACs is discussed.
Collapse
Affiliation(s)
- Ji-Jie Pang
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
37
|
Opposite effects of nitric oxide on rod and cone photoreceptors of rat retina in situ. Neurosci Lett 2010; 473:62-6. [DOI: 10.1016/j.neulet.2010.02.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 02/09/2010] [Accepted: 02/11/2010] [Indexed: 02/02/2023]
|
38
|
Giove TJ, Deshpande MM, Eldred WD. Identification of alternate transcripts of neuronal nitric oxide synthase in the mouse retina. J Neurosci Res 2009; 87:3134-42. [PMID: 19479987 DOI: 10.1002/jnr.22133] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Nitric oxide (NO) is a major signaling molecule in the retina and CNS, with physiological roles in every cell type in the retina. Previous work shows that neuronal nitric oxide synthase (nNOS) is an important source of NO in the vertebrate retina. There are distinct, active alternative transcripts of nNOS observed in many tissues, including testes and brain, that may differ in both localization and enzyme kinetics. The present study characterized nNOS and the NO production from nNOS in the mouse retina in terms of its alternate transcripts, namely, nNOS alpha, nNOS beta, and nNOS gamma. We examined both basal and light-stimulated NO production as imaged using the NO-sensitive dye 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate-FM (DAF-FM), and we compared the NO production with the immunocytochemical localization of nNOS using antisera that recognize nNOS alpha/beta or nNOS alpha/beta/gamma. Western blots suggested the presence of NOS alpha/gamma protein in retina, but not nNOS beta, and we confirmed this at the message level by using a combination of RT-PCR and quantitative real-time PCR. Our findings indicated that the primary source of NO in the mammalian retina is nNOS alpha and that nNOS gamma may contribute to NO production as well.
Collapse
Affiliation(s)
- Thomas J Giove
- Laboratory of Visual Neurobiology, Department of Biology, Boston University, Boston, Massachusetts 02215, USA
| | | | | |
Collapse
|
39
|
Increased neuronal nitric oxide synthase activity in retinal neurons in early diabetic retinopathy. Mol Vis 2009; 15:2249-58. [PMID: 19936028 PMCID: PMC2776346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Accepted: 11/03/2009] [Indexed: 11/11/2022] Open
Abstract
PURPOSE There are increased levels of nitric oxide (NO) in diabetic retinas. The purpose of this study was to determine the extent that neuronal nitric oxide synthase (nNOS) contributes to the increased levels of retinal NO in early diabetic retinopathy by examining the expression and activity of nNOS in retinal neurons after 5 weeks of diabetes. METHODS Changes in NO levels were measured using NO imaging of retinal neurons in mice with streptozotocin-induced diabetes for five weeks. NO imaging was compared to nNOS localization using immunocytochemistry, and nNOS message and protein levels were measured using quantitative real-time PCR and western blots. RESULTS There was a close anatomic correlation between the localization of the increased NO production and the nNOS immunoreactivity in the retinal plexiform layers of diabetic retinas. There was no change in nNOS message, but nNOS protein was decreased and its subcellular localization was altered. Treatment with insulin or aminoguanidine partially ameliorated the increase in NO in diabetic retinas. CONCLUSIONS These results suggest that increased nNOS activity is responsible for the majority of increased NO in retinal neurons in early diabetic retinopathy. This supports a role for increased nNOS activity in the early neuronal dysfunction in the diabetic retina.
Collapse
|
40
|
Tummala SR, Benac S, Tran H, Vankawala A, Zayas-Santiago A, Appel A, Kang Derwent JJ. Effects of inhibition of neuronal nitric oxide synthase on basal retinal blood flow regulation. Exp Eye Res 2009; 89:801-9. [DOI: 10.1016/j.exer.2009.07.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 07/08/2009] [Accepted: 07/12/2009] [Indexed: 12/16/2022]
|
41
|
Nemargut JP, Wang GY. Inhibition of nitric oxide synthase desensitizes retinal ganglion cells to light by diminishing their excitatory synaptic currents under light adaptation. Vision Res 2009; 49:2936-47. [PMID: 19772868 DOI: 10.1016/j.visres.2009.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 09/11/2009] [Accepted: 09/16/2009] [Indexed: 10/20/2022]
Abstract
The effect of inhibiting nitric oxide synthase (NOS) on the visual responses of mouse retinal ganglion cells (RGCs) was studied under light adaptation by using patch-clamp recordings. The results demonstrated that NOS inhibitor, l-NAME, reduced the sensitivity of RGCs to light under light adaptation at different ambient light conditions. These observations were seen in all cells that recordings were made from. l-NAME diminished the excitatory synaptic currents (EPSCs), rather than increasing the inhibitory synaptic currents, of RGCs to reduce the sensitivity of RGCs to light. Cones may be the sites that l-NAME acted to diminish the EPSCs of RGCs.
Collapse
Affiliation(s)
- Joseph P Nemargut
- Department of Structural and Cellular Biology, School of Medicine, Tulane University, New Orleans, LA 70112, United States
| | | |
Collapse
|
42
|
Hong H, Sun J, Cai W. Multimodality imaging of nitric oxide and nitric oxide synthases. Free Radic Biol Med 2009; 47:684-98. [PMID: 19524664 DOI: 10.1016/j.freeradbiomed.2009.06.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 05/28/2009] [Accepted: 06/10/2009] [Indexed: 01/27/2023]
Abstract
Nitric oxide (NO) and NO synthases (NOSs) are crucial factors in many pathophysiological processes such as inflammation, vascular/neurological function, and many types of cancer. Noninvasive imaging of NO or NOS can provide new insights in understanding these diseases and facilitate the development of novel therapeutic strategies. In this review, we will summarize the current state-of-the-art multimodality imaging in detecting NO and NOSs, including optical (fluorescence, chemiluminescence, and bioluminescence), electron paramagnetic resonance (EPR), magnetic resonance (MR), and positron emission tomography (PET). With continued effort over the last several years, these noninvasive imaging techniques can now reveal the biodistribution of NO or NOS in living subjects with high fidelity which will greatly facilitate scientists/clinicians in the development of new drugs and/or patient management. Lastly, we will also discuss future directions/applications of NO/NOS imaging. Successful development of novel NO/NOS imaging agents with optimal in vivo stability and desirable pharmacokinetics for clinical translation will enable the maximum benefit in patient management.
Collapse
Affiliation(s)
- Hao Hong
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705-2275, USA
| | | | | |
Collapse
|
43
|
Abstract
Nitric oxide (NO) is a gaseous neuromodulator that has physiological functions in every cell type in the retina. Evidence indicates that NO often plays a role in the processing of visual information in the retina through the second messenger cyclic guanosine monophosphate (cGMP). Despite numerous structural and functional studies of this signaling pathway in the retina, none have examined many of the elements of this pathway within a single study in a single species. In this study, the NO/cGMP pathway was localized to specific regions and cell types within the inner and outer retina. We have immunocytochemically localized nitric oxide synthase, the enzyme that produces NO, in photoreceptor ellipsoids, four distinct classes of amacrine cells, Müller and bipolar cells, somata in the ganglion cell layer, as well as in processes within both plexiform layers. Additionally, we localized NO production in specific cell types using the NO-sensitive dye diaminofluorescein. cGMP immunocytochemistry was used to functionally localize soluble guanylate cyclase that was activated by an NO donor in select amacrine and bipolar cell classes. Analysis of cGMP and its downstream target, cGMP-dependent protein kinase II (PKGII), showed colocalization within processes in the outer retina as well as in somata in the inner retina. The results of this study showed that the NO/cGMP signaling pathway was functional and its components were widely distributed throughout specific cell types in the outer and inner salamander retina.
Collapse
|
44
|
Petzold GC, Haack S, von Bohlen und Halbach O, Priller J, Lehmann TN, Heinemann U, Dirnagl U, Dreier JP. Nitric Oxide Modulates Spreading Depolarization Threshold in the Human and Rodent Cortex. Stroke 2008; 39:1292-9. [DOI: 10.1161/strokeaha.107.500710] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Gabor C. Petzold
- From the Departments of Experimental Neurology (G.C.P., S.H., J.P., U.D., J.P.D.), Neurology (G.C.P., U.D., J.P.D.), Psychiatry (J.P.), and Neurosurgery (T.-N.L.), and the Johannes Müller Institute of Physiology (U.H.), Charité University Medicine Berlin, Berlin, and the Interdisciplinary Center for Neuroscience (O.v.B.u.H.), Department of Neuroanatomy, University of Heidelberg, Heidelberg, Germany
| | - Stephan Haack
- From the Departments of Experimental Neurology (G.C.P., S.H., J.P., U.D., J.P.D.), Neurology (G.C.P., U.D., J.P.D.), Psychiatry (J.P.), and Neurosurgery (T.-N.L.), and the Johannes Müller Institute of Physiology (U.H.), Charité University Medicine Berlin, Berlin, and the Interdisciplinary Center for Neuroscience (O.v.B.u.H.), Department of Neuroanatomy, University of Heidelberg, Heidelberg, Germany
| | - Oliver von Bohlen und Halbach
- From the Departments of Experimental Neurology (G.C.P., S.H., J.P., U.D., J.P.D.), Neurology (G.C.P., U.D., J.P.D.), Psychiatry (J.P.), and Neurosurgery (T.-N.L.), and the Johannes Müller Institute of Physiology (U.H.), Charité University Medicine Berlin, Berlin, and the Interdisciplinary Center for Neuroscience (O.v.B.u.H.), Department of Neuroanatomy, University of Heidelberg, Heidelberg, Germany
| | - Josef Priller
- From the Departments of Experimental Neurology (G.C.P., S.H., J.P., U.D., J.P.D.), Neurology (G.C.P., U.D., J.P.D.), Psychiatry (J.P.), and Neurosurgery (T.-N.L.), and the Johannes Müller Institute of Physiology (U.H.), Charité University Medicine Berlin, Berlin, and the Interdisciplinary Center for Neuroscience (O.v.B.u.H.), Department of Neuroanatomy, University of Heidelberg, Heidelberg, Germany
| | - Thomas-Nicolas Lehmann
- From the Departments of Experimental Neurology (G.C.P., S.H., J.P., U.D., J.P.D.), Neurology (G.C.P., U.D., J.P.D.), Psychiatry (J.P.), and Neurosurgery (T.-N.L.), and the Johannes Müller Institute of Physiology (U.H.), Charité University Medicine Berlin, Berlin, and the Interdisciplinary Center for Neuroscience (O.v.B.u.H.), Department of Neuroanatomy, University of Heidelberg, Heidelberg, Germany
| | - Uwe Heinemann
- From the Departments of Experimental Neurology (G.C.P., S.H., J.P., U.D., J.P.D.), Neurology (G.C.P., U.D., J.P.D.), Psychiatry (J.P.), and Neurosurgery (T.-N.L.), and the Johannes Müller Institute of Physiology (U.H.), Charité University Medicine Berlin, Berlin, and the Interdisciplinary Center for Neuroscience (O.v.B.u.H.), Department of Neuroanatomy, University of Heidelberg, Heidelberg, Germany
| | - Ulrich Dirnagl
- From the Departments of Experimental Neurology (G.C.P., S.H., J.P., U.D., J.P.D.), Neurology (G.C.P., U.D., J.P.D.), Psychiatry (J.P.), and Neurosurgery (T.-N.L.), and the Johannes Müller Institute of Physiology (U.H.), Charité University Medicine Berlin, Berlin, and the Interdisciplinary Center for Neuroscience (O.v.B.u.H.), Department of Neuroanatomy, University of Heidelberg, Heidelberg, Germany
| | - Jens P. Dreier
- From the Departments of Experimental Neurology (G.C.P., S.H., J.P., U.D., J.P.D.), Neurology (G.C.P., U.D., J.P.D.), Psychiatry (J.P.), and Neurosurgery (T.-N.L.), and the Johannes Müller Institute of Physiology (U.H.), Charité University Medicine Berlin, Berlin, and the Interdisciplinary Center for Neuroscience (O.v.B.u.H.), Department of Neuroanatomy, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
45
|
Cimini BA, Strang CE, Wotring VE, Keyser KT, Eldred WD. Role of acetylcholine in nitric oxide production in the salamander retina. J Comp Neurol 2008; 507:1952-63. [PMID: 18273886 DOI: 10.1002/cne.21655] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Although acetylcholine is one of the most widely studied neurotransmitters in the retina, many questions remain about its downstream signaling mechanisms. In this study we initially characterized the cholinergic neurotransmitter system in the salamander retina by localizing a variety of cholinergic markers. We then examined the link between both muscarinic and nicotinic receptor activation and nitric oxide production by using immunocytochemistry for cyclic guanosine monophosphate (cGMP) as an indicator. We found a large increase in cGMP-like immunoreactivity (cGMP-LI) in the inner retina in response to muscarinic (but not nicotinic) receptor activation. Based on the amplification of mRNA transcripts, receptor immunocytochemistry, and the use of selective antagonists, we identified these receptors as M2 muscarinic receptors. Using double-labeling techniques, we established that these increases in cGMP-LI were seen in GABAergic but not cholinergic amacrine cells, and that the increases were blocked by inhibitors of nitric oxide production. The creation of nitric oxide in response to cholinergic receptor activation may provide a mechanism for modulating the well-known mutual interactions of acetylcholine-glycine-GABA in the inner retina. As GABA and glycine are the primary inhibitory neurotransmitters in the retina, signaling pathways that modulate their levels or release will have major implications for the processing of complex stimuli by the retina.
Collapse
Affiliation(s)
- Beth A Cimini
- Department of Biology, Boston University, Boston, Massachusetts 02215, USA
| | | | | | | | | |
Collapse
|
46
|
|
47
|
Ichinose T, Lukasiewicz PD. Ambient light regulates sodium channel activity to dynamically control retinal signaling. J Neurosci 2007; 27:4756-64. [PMID: 17460088 PMCID: PMC3232015 DOI: 10.1523/jneurosci.0183-07.2007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The retinal network increases its sensitivity in low-light conditions to detect small visual inputs and decreases its sensitivity in bright-light conditions to prevent saturation. However, the cellular mechanisms that adjust visual signaling in the retinal network are not known. Here, we show that voltage-gated sodium channels in bipolar cells dynamically control retinal light sensitivity. In dim conditions, sodium channels amplified light-evoked synaptic responses mediated by cone pathways. Conversely, in bright conditions, sodium channels were inactivated by dopamine released from amacrine cells, and they did not amplify synaptic inputs, minimizing signal saturation. Our findings demonstrate that bipolar cell sodium channels mediate light adaptation by controlling retinal signaling gain.
Collapse
Affiliation(s)
| | - Peter D. Lukasiewicz
- Departments of Ophthalmology and Visual Sciences and
- Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
48
|
Abstract
The nitric oxide (NO)-cGMP pathway is implicated in modulation of visual information processing in the retina. Despite numerous functional studies of this pathway, information about the retinal distribution of the major downstream effector of NO, soluble guanylyl cyclase (sGC), is very limited. In the present work, we have used immunohistochemistry and multiple labeling to determine the distribution of sGC in rat retina. sGC was present at high levels in inner retina but barely detectable in outer retina. Photoreceptors and horizontal cells, as well as Müller cells, were immunonegative, whereas retinal ganglion cells exhibited moderate staining for sGC. Strong immunostaining was found in subpopulations of bipolar and amacrine cells, but staining was weak in rod bipolar cells, and AII amacrine cells were immunonegative. Double labeling of sGC with neuronal nitric oxide synthase showed that the two proteins are generally located in adjacent puncta in inner plexiform layer, implying paracrine interactions. Our results suggest that the NO-cGMP pathway modulates the neural circuitry in inner retina, preferentially within the cone pathway.
Collapse
Affiliation(s)
- Jin-Dong Ding
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | |
Collapse
|
49
|
Vidal L, Díaz F, Villena A, Moreno M, Campos JG, de Vargas IP. Nitric oxide synthase in retina and optic nerve head of rat with increased intraocular pressure and effect of timolol. Brain Res Bull 2006; 70:406-13. [PMID: 17027776 DOI: 10.1016/j.brainresbull.2006.07.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Revised: 06/23/2006] [Accepted: 07/14/2006] [Indexed: 11/16/2022]
Abstract
We investigated the expression of nitric oxide synthase (NOS) isoforms -1, -2 and -3 in the retina and optic nerve head (ONH) in an experimental rat model of elevated intraocular pressure (IOP) before and after treatment with timolol, to assess whether its neuroprotective action is associated with the activity of these enzymes. Episcleral vein cauterization in unilateral eyes of Wistar rats was performed to produce elevated IOP. Histological sections of retina and ONH from animals with normal IOP, with elevated IOP, and elevated IOP treated with timolol, were studied by immunohistochemistry with antibodies to NOS-1, NOS-2, and NOS-3. In the control rats, NOS-1 was localized to photoreceptor inner segments, amacrine cells and bipolar cells in the retina, and in astrocytes, pericytes and vascular nitrergic terminals in the ONH. NOS-3 immunostaining localized to the endothelial cells. The rats with elevated IOP showed increased expression of NOS-1 in the plexiform layers of the retina and reactive astrocytes in the ONH. These cells also showed NOS-2 positivity. The rats treated with timolol showed reduced expression of NOS-1 in the retina and ONH. NOS-2 was only detected in a few groups of astrocytes in the ONH. NOS-3 was unchanged in both elevated IOP and timolol-treated groups. These results show that excessive levels of NO synthesized by the NOS-1 and -2 isoforms, considered neurotoxic, might contribute to the progressive lesions of retinal ganglion cell axons. Their reduction after treatment suggests a possible neuroprotective effect of timolol in neurons exposed to excessive amounts of NO.
Collapse
Affiliation(s)
- Lourdes Vidal
- Department of Histology and Histopathology, School of Medicine, University of Malaga, Boulevard Louis Pasteur 32, 29071 Malaga, Spain.
| | | | | | | | | | | |
Collapse
|
50
|
Hoffpauir B, McMains E, Gleason E. Nitric oxide transiently converts synaptic inhibition to excitation in retinal amacrine cells. J Neurophysiol 2006; 95:2866-77. [PMID: 16467419 DOI: 10.1152/jn.01317.2005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nitric oxide (NO) is generated by multiple cell types in the vertebrate retina, including amacrine cells. We investigate the role of NO in the modulation of synaptic function using a culture system containing identified retinal amacrine cells. We find that moderate concentrations of NO alter GABA(A) receptor function to produce an enhancement of the GABA-gated current. Higher concentrations of NO also enhance GABA-gated currents, but this enhancement is primarily due to a substantial positive shift in the reversal potential of the current. Several pieces of evidence, including a similar effect on glycine-gated currents, indicate that the positive shift is due to an increase in cytosolic Cl-. This change in the chloride distribution is especially significant because it can invert the sign of GABA- and glycine-gated voltage responses. Furthermore, current- and voltage-clamp recordings from synaptic pairs of GABAergic amacrine cells demonstrate that NO transiently converts signaling at GABAergic synapses from inhibition to excitation. Persistence of the NO-induced shift in E(Cl-) in the absence of extracellular Cl- indicates that the increase in cytosolic Cl- is due to release of Cl- from an internal store. An NO-dependent release of Cl- from an internal store is also demonstrated for rat hippocampal neurons indicating that this mechanism is not restricted to the avian retina. Thus signaling in the CNS can be fundamentally altered by an NO-dependent mobilization of an internal Cl- store.
Collapse
Affiliation(s)
- Brian Hoffpauir
- West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | | | | |
Collapse
|