Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Neuroscience ; 512: 1-15, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36572172

ABSTRACT

Currently, there is a lack of treatments for retinal neurotrauma. To address this issue, this study uses an alpha7 nAChR agonist, PNU-282987, to determine it effects on functional activity in the retina shortly after a traumatic blast exposure. The objectives of this research include: (1) examination of the cellular and functional damage associated with ocular blast exposure, and (2) evaluation of structural and functional changes that occur post PNU-282987 treatment. Significant ocular blast damage was induced in adult mice after exposure to a single blast of 35 psi to the left eye. Blast-exposed transgenic mice expressing tdTomato Müller glia were treated daily with eyedrops containing PNU-282987 for 4 weeks following the blast exposure. Antibody staining studies in these transgenic mice was conducted to examine lineage tracing and electroretinograms (ERGs) were obtained to examine functional changes. Blast exposure caused a significant loss of cells in all retinal layers after 4 weeks. Immunohistochemical analysis demonstrated tdTomato-positive labeled photoreceptors and retinal ganglion cells in blast-exposed mice treated with PNU-282987. ERG recordings were taken from control animals, from blast-damaged animals and from animals exposed to blast followed by 4 weeks of PNU-282987 treatment. Scotopic ERG recordings from blast-exposed mice had significantly decreased amplitudes of a-wave, b-wave, oscillatory potentials and flicker frequencies, which were prevented after PNU-282987 treatment. In photopic experiments, the PhNR response was reduced significantly after blast exposure but the decrease was prevented after treatment with PNU-282987. These are the first experiments that demonstrate preservation of retinal function after blast exposure using an alpha7 nAChR agonist.


Subject(s)
Receptors, Nicotinic , alpha7 Nicotinic Acetylcholine Receptor , Mice , Animals , Bridged Bicyclo Compounds/pharmacology , Nicotinic Agonists/pharmacology , Retina , Mice, Transgenic
2.
J Mol Neurosci ; 72(9): 1990-2010, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35867327

ABSTRACT

PNU-282987, a selective alpha7 nicotinic acetylcholine receptor agonist, has previously been shown to have both neurogenic and broad regenerative effects in the adult murine retina. The objective of this study was to assay the molecular mechanism by which PNU-282987 promotes the production of Muller-derived progenitor cells through signaling via the resident retinal pigment epithelium. These Muller-derived progenitor cells generate a myriad of differentiated neurons throughout the retina that have previously been characterized by morphology. Herein, we demonstrate that topical application of PNU-282987 stimulates production of functional neurons as measured by electroretinograms. Further, we examine the mechanism of how this phenomenon occurs through activation of this atypical receptor using a transcriptomic approach isolated retinal pigment epithelium activated by PNU-282987 and in whole retina. We provide evidence that PNU-282987 causes a bi-modal signaling event in which early activation primes the retina with an inflammatory response and developmental signaling cues, followed by an inhibition of gliotic mechanisms and a decrease in the immune response, ending with upregulation of genes associated with specific retinal neuron generation. Taken together, these data provide evidence that PNU-282987 activates the retinal pigment epithelium to signal to Muller glia to produce Muller-derived progenitor cells, which can differentiate into new, functional neurons in adult mice. These data not only increase our understanding of how adult mammalian retinal regeneration can occur, but also provide therapeutic promise for treating functional vision loss.


Subject(s)
Retinal Neurons , Retinal Pigment Epithelium , Animals , Benzamides , Bridged Bicyclo Compounds , Mammals/genetics , Mice , Neurogenesis , Neuroglia , Retina , Transcriptome
3.
Exp Eye Res ; 210: 108717, 2021 09.
Article in English | MEDLINE | ID: mdl-34348130

ABSTRACT

The adult mammal lacks the ability to regenerate neurons lost to retinal damage or disease in a meaningful capacity. However, previous studies from this laboratory have demonstrated that PNU-282987, an α7 nicotinic acetylcholine receptor agonist, elicits a robust neurogenic response in the adult murine retina. With eye drop application of PNU-282987, Müller glia cells re-enter the cell cycle and produce progenitor-like cells that can differentiate into various types of retinal neurons. In this study, we analyzed the regenerative capability of PNU-282987 in two retinal disease models and identified the source of newly regenerated neurons. Wild-type mice and mice with a transgenic Müller-glia lineage tracer were manipulated to mimic loss of retinal cells associated with glaucoma or photoreceptor degeneration. Following treatment with PNU-282987, the regenerative response of retinal neurons was quantified and characterized. After onset of photoreceptor degeneration, PNU-282987 was able to successfully regenerate both rod and cone photoreceptors. Quantification of this response demonstrated significant regeneration, restoring photoreceptors to near wild-type density. In mice that had glaucoma-like conditions induced, PNU-282987 treatment led to a significant increase in retinal ganglion cells. Retrograde labeling of optic nerve axon fibers demonstrated that newly regenerated axons projected into the optic nerve. Lineage tracing analysis demonstrated that these new neurons were derived from Müller glia. These results demonstrate that PNU-282987 can induce retinal regeneration in adult mice following onset of retinal damage. The ability of PNU-282987 to regenerate retinal neurons in a robust manner offers a new direction for developing novel and potentially transformative treatments to combat neurodegenerative disease.


Subject(s)
Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , Disease Models, Animal , Nerve Regeneration/physiology , Retinal Degeneration/drug therapy , Retinal Ganglion Cells/physiology , Retinal Neurons/physiology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Cell Cycle , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Glaucoma/physiopathology , Intraocular Pressure/physiology , Mice , Mice, Inbred Strains , Mice, Transgenic , Neurogenesis , Nicotinic Agonists/pharmacology , Retinal Degeneration/metabolism
4.
Front Mol Biosci ; 7: 200, 2020.
Article in English | MEDLINE | ID: mdl-32923455

ABSTRACT

Previous studies from this lab have determined that dedifferentiation of Müller glia occurs after eye drop application of an α7 nicotinic acetylcholine receptor (nAChR) agonist, PNU-282987, to the adult rodent eye. PNU-282987 acts on α7 nAChRs on retinal pigment epithelial cells to stimulate production of Müller-derived progenitor cells (MDPCs) and ultimately lead to neurogenesis. This current study was designed to test the hypothesis that the activation of genes involved in the HB-EGF/Ascl1/Lin28a signaling pathway in Müller glia leads to the genesis of MDPCs. RNA-seq was performed on a Müller glial cell line (rMC-1) following contact with supernatant collected from a retinal pigment epithelial (RPE) cell line treated with PNU-282987. Differentially regulated genes were compared with published literature of Müller glia dedifferentiation that occurs in lower vertebrate regeneration and early mammalian development. HB-EGF was significantly up-regulated by 8 h and expression increased through 12 h. By 48 h, up-regulation of Ascl1 and Lin28a was observed, two genes known to be rapidly induced in dedifferentiating zebrafish Müller glia. Up-regulation of other genes known to be involved in mammalian development and zebrafish regeneration were also observed, as well as down-regulation of some factors necessary for Müller glia cell identity. RNA-seq results were verified using qRT-PCR. Using immunocytochemistry, the presence of markers associated with MDCP identity, Otx2, Nestin, and Vsx2, were found to be expressed in the 48 h treatment group cultures. This study is novel in its demonstration that Müller glia in adult rodents can be induced into regenerative activity by stimulating genes involved in the HB-EGF/Ascl1/Lin28a pathway that leads to MDPCs after introducing conditioned media from PNU-282987 treated RPE. This study furthers our understanding of the mechanism by which Müller glia dedifferentiate in response to PNU-282987 in the adult mammalian retina.

5.
Invest Ophthalmol Vis Sci ; 60(2): 570-579, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30721274

ABSTRACT

Purpose: The adult mammalian retina is typically incapable of regeneration when damaged by disease or trauma. Restoration of function would require generation of new adult neurons, something that until recently, mammals were thought to be incapable of doing. However, previous studies from this laboratory have shown that the α7 nicotinic acetylcholine receptor (α7 nAChR) agonist, PNU-282987, induces cell cycle reentry of Müller glia and generation of mature retinal neurons in adult rats, in the absence of detectible injury. This study analyzes how PNU-282987 treatment in RPE leads to robust BrdU incorporation in Müller glia in adult mice and leads to generation of Müller-derived retinal progenitors and neuronal differentiation. Methods: Retinal BrdU incorporation was examined after eye drop application of PNU-282987 in adult wild-type and transgenic mice that contain tamoxifen-inducible tdTomato Müller glia, or after intraocular injection of conditioned medium from PNU-282987-treated cultured RPE cells. Results: PNU-282987 induced robust incorporation of BrdU in all layers of the adult mouse retina. The α7 nAChR agonist was found to stimulate cell cycle reentry of Müller glia and their generation of new retinal progenitors indirectly, via the RPE, in an α7 nAChR-dependent fashion. Conclusions: The results from this study point to RPE as a contributor to Müller glial neurogenic responses. The manipulation of the RPE to stimulate retinal neurogenesis offers a new direction for developing novel and potentially transformative treatments to reverse the loss of neurons associated with neurodegenerative disease, traumatic injury, or aging.


Subject(s)
Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , Ependymoglial Cells/physiology , Neurogenesis/physiology , Neuroglia/physiology , Nicotinic Agonists/pharmacology , Retinal Pigment Epithelium/drug effects , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Bromodeoxyuridine/metabolism , Cell Cycle/physiology , Cells, Cultured , Ependymoglial Cells/cytology , Female , Male , Mice , Mice, Transgenic , PAX6 Transcription Factor/metabolism , Rats , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Stem Cells/physiology
6.
Bio Protoc ; 8(21)2018 Nov 05.
Article in English | MEDLINE | ID: mdl-30687771

ABSTRACT

Eye drop treatments are typically used to apply drugs to the anterior structures of the eye. Recently, however, studies have demonstrated that eye drops can reach the retina in the back of the eye if pharmacological agents are carried in appropriate vehicles. Here, we introduce an eye drop procedure to deliver a drug (PNU-282987), in combination with BrdU, to stimulate cell cycle re-entry and label dividing cells in the retinas of adult rodents. This procedure avoids potential systemic complications of repeated intraperitoneal injections, as well as the retinal damage that is induced by repeated intravitreal injections. Although the delivery of PNU-282987 and BrdU is the focus of this article, many different proliferating compounds could be delivered to the retina using this procedure.

7.
Neuroscience ; 346: 437-446, 2017 03 27.
Article in English | MEDLINE | ID: mdl-28147247

ABSTRACT

Irreversible vision loss due to disease or age is responsible for a reduced quality of life. The experiments in this study test the hypothesis that the α7 nicotinic acetylcholine receptor agonist, PNU-282987, leads to the generation of retinal neurons in an adult mammalian retina in the absence of retinal injury or exogenous growth factors. Using antibodies against BrdU, retinal ganglion cells, progenitor cells and Müller glia, the results of this study demonstrate that multiple types of retinal cells and neurons are generated after eye drop application of PNU-282987 in adult Long Evans rats in a dose-dependent manner. The results of this study provide evidence that progenitor cells, derived from Müller glia after treatment with PNU-282987, differentiate and migrate to the photoreceptor and retinal ganglion cell layers. If retinas were treated with the alpha7 nAChR antagonist, methyllycaconitine, before agonist treatment, BrdU-positive cells were significantly reduced. As adult mammalian neurons do not typically regenerate or proliferate, these results have implications for reversing vision loss due to neurodegenerative disease or the aging process to improve the quality of life for millions of patients.


Subject(s)
Benzamides/administration & dosage , Bridged Bicyclo Compounds/administration & dosage , Retinal Neurons/drug effects , Retinal Neurons/physiology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Apoptosis/drug effects , Bromodeoxyuridine , Cell Movement/drug effects , Ependymoglial Cells/drug effects , Ependymoglial Cells/physiology , Female , Male , Neural Stem Cells/drug effects , Neural Stem Cells/physiology , Rats , Rats, Long-Evans , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/physiology
9.
Curr Pharm Des ; 22(14): 2178-92, 2016.
Article in English | MEDLINE | ID: mdl-26818878

ABSTRACT

BACKGROUND: Glaucoma is characterized as a neuropathic disease that causes progressive degeneration of retinal ganglion cells (RGCs) in the retina, resulting in irreversible loss of vision. All conventional treatments for glaucoma are focused on reducing intraocular pressure (IOP) in the anterior chamber of the eye. However, these treatments alone are insufficient to halt the progression of the disease. As a result, neuroprotective strategies have been developed that prevent retinal neuron loss and disease progression. METHODS: The goal of this review is to summarize and discuss neuroprotective strategies in glaucoma at the level of the retina and the ganglion cell layer instead of treatments targeting IOP. Recent and past neuroprotective therapies used to prevent the loss of retinal ganglion cells, the loss of axons in the optic nerve and the loss of vision and function associated with glaucoma are presented. RESULTS: Pharmacological approaches have targeted specific receptors, signaling cascades and neurotrophic factors to induce neuroprotection in the retina, while others have focused on the mechanism of cellular loss associated with glaucoma, including excitotoxicity, oxidative stress and apoptotic processes. In addition to neuroprotective pharmacological treatments, stem cell, gene therapy and viral research have demonstrated neuroprotection against the loss of RGCs in glaucomatous conditions. CONCLUSION: It is likely that future development for glaucoma treatment will include a combination of these treatments to prevent the pathophysiology of glaucoma.


Subject(s)
Glaucoma/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Glaucoma/pathology , Humans , Intraocular Pressure/drug effects , Neuroprotective Agents/chemistry , Retina/drug effects , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology
10.
Neuropharmacology ; 99: 337-46, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26239818

ABSTRACT

The α7nAChR agonist, PNU-282987, has previously been shown to have a neuroprotective effect against loss of retinal ganglion cells (RGCs) in an in vivo glaucoma model when the agent was injected into the vitreous chamber of adult Long Evans rat eyes. Here, we characterized the neuroprotective effect of PNU-282987 at the nerve fiber and retinal ganglion cell layer, determined that neuroprotection occurred when the agonist was applied as eye drops and verified detection of the agonist in the retina, using LC/MS/MS. To induce glaucoma-like conditions in adult Long Evans rats, hypertonic saline was injected into the episcleral veins to induce scar tissue and increase intraocular pressure. Within one month, this procedure produced significant loss of RGCs compared to untreated conditions. RGCs were quantified after immunostaining with an antibody against Thy 1.1 and imaged using a confocal microscope. In dose-response studies, concentrations of PNU-282987 were applied to the animal's right eye two times each day, while the left eye acted as an internal control. Eye drops of PNU-282987 resulted in neuroprotection against RGC loss in a dose-dependent manner using concentrations between 100 µM and 2 mM PNU-282987. LC/MS/MS results demonstrated that PNU-282987 was detected in the retina when applied as eye drops, relatively small amounts of PNU-282987 were measured in blood plasma and no PNU-282987 was detected in cardiac tissue. These results support the hypothesis that eye drop application of PNU-282987 can prevent loss of RGCs associated with glaucoma, which can lead to neuroprotective treatments for diseases that involve α7nAChRs.


Subject(s)
Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , Glaucoma/drug therapy , Neuroprotective Agents/pharmacology , Retinal Ganglion Cells/drug effects , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Benzamides/pharmacokinetics , Bridged Bicyclo Compounds/pharmacokinetics , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Glaucoma/metabolism , Glaucoma/pathology , Heart/drug effects , Male , Microscopy, Confocal , Myocardium/metabolism , Neuroprotective Agents/pharmacokinetics , Ophthalmic Solutions/pharmacokinetics , Ophthalmic Solutions/pharmacology , Rats, Long-Evans , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Saline Solution, Hypertonic , alpha7 Nicotinic Acetylcholine Receptor/metabolism
11.
Invest Ophthalmol Vis Sci ; 55(2): 1078-87, 2014 Feb 24.
Article in English | MEDLINE | ID: mdl-24458148

ABSTRACT

PURPOSE: The purpose of this study was to analyze the neuroprotective effect of an α7 nAChR agonist, PNU-282987, using an in vivo model of glaucoma in Long Evans rats. METHODS: One eye in each animal was surgically manipulated to induce glaucoma in control untreated animals and in animals that were treated with intravitreal injections of PNU-282987. To induce glaucoma-like conditions, 0.05 mL of 2 M NaCl was injected into the episcleral veins of right eyes in each rat to create scar tissue and increase intraocular pressure. The left eye in each rat acted as an internal control. One month following NaCl injection, rats were euthanized, retinas were removed, flatmounted, fixed, and nuclei were stained with cresyl violet or RGCs were immunostained with an antibody against Thy 1.1 or against Brn3a. Stained nuclei in the RGC layer and labeled RGCs in NaCl-injected retinas were counted and compared with cell counts from untreated retinas in the same animal. RESULTS: NaCl injections into the episcleral veins caused a significant loss of cells by an average of 27.35% (± 2.12 SEM) in the RGC layer within 1 month after NaCl injection, which corresponded to a significant loss of RGCs. This loss of RGCs was eliminated if 5 µL of 100 µM PNU-282987 was injected into the right eye an hour before NaCl injection. CONCLUSIONS: The results from this study support the hypothesis that the α7 agonist, PNU-282987, has a neuroprotective effect in the rat retina. PNU-282987 may be a viable candidate for future therapeutic treatments of glaucoma.


Subject(s)
Benzamides/therapeutic use , Bridged Bicyclo Compounds/therapeutic use , Glaucoma/prevention & control , Nicotinic Agonists/therapeutic use , Optic Nerve Diseases/prevention & control , Retinal Degeneration/prevention & control , Retinal Ganglion Cells/drug effects , Animals , Cell Count , Chromatography, High Pressure Liquid , Disease Models, Animal , Female , Glaucoma/chemically induced , Glaucoma/pathology , Intravitreal Injections , Male , Neuroprotective Agents/therapeutic use , Optic Nerve Diseases/chemically induced , Optic Nerve Diseases/pathology , Rats , Rats, Long-Evans , Retinal Degeneration/chemically induced , Retinal Degeneration/pathology , Retinal Ganglion Cells/pathology , Tandem Mass Spectrometry , Tonometry, Ocular , alpha7 Nicotinic Acetylcholine Receptor/agonists
12.
Neuropharmacology ; 73: 111-21, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23727438

ABSTRACT

The objective of this study was to determine the neuroprotective role of tropisetron on retinal ganglion cells (RGCs) as well as to explore the possible mechanisms associated with alpha7 nAChR-induced neuroprotection. Adult pig RGCs were isolated from all other retinal tissue using a two-step panning technique. Once isolated, RGCs were cultured for 3 days under control untreated conditions, in the presence of 500 µM glutamate to induce excitotoxicity, and when tropisetron was applied before glutamate to induce neuroprotection. 500 µM glutamate decreased RGC survival by an average of 62% compared to control conditions. However, RGCs pretreated with 100 nM tropisetron before glutamate increased cell survival to an average of 105% compared to controls. Inhibition studies using the alpha7 nAChR antagonist, MLA (10 nM), support the hypothesis that tropisetron is an effective neuroprotective agent against glutamate-induced excitotoxicity; mediated by α7 nAChR activation. ELISA studies were performed to determine if signaling cascades normally associated with excitotoxicity and neuroprotection were up- or down-regulated after tropisetron treatment. Tropisetron had no discernible effects on pAkt levels but significantly decreased p38 MAPK levels associated with excitotoxicity from an average of 15 ng/ml to 6 ng/ml. Another mechanism shown to be associated with neuroprotection involves internalization of NMDA receptors. Double-labeled immunocytochemistry and electrophysiology studies provided further evidence that tropisetron caused internalization of NMDA receptor subunits. The findings of this study suggest that tropisetron could be an effective therapeutic agent for the treatment of degenerative disorders of the central nervous system that involves excitotoxicity.


Subject(s)
Glutamic Acid/toxicity , Indoles/pharmacology , Neuroprotective Agents/pharmacology , Nicotinic Agonists/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , Aconitine/analogs & derivatives , Aconitine/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Indoles/antagonists & inhibitors , Nicotinic Antagonists/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Signal Transduction/drug effects , Swine , Tropisetron , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Neurochem ; 112(1): 214-26, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19845831

ABSTRACT

Previous studies have reported that activation of nicotinic acetylcholine (ACh) receptors (nAChRs) on cultured pig retinal ganglion cells (RGCs) has a neuroprotective effect against glutamate-induced excitotoxicity. However, the mechanism linking nAChRs to neuroprotection is unknown. Here, we tested the hypothesis that signaling cascades involving p38 mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) --> Akt are involved in linking activation of nAChRs to neuroprotection in isolated pig RGCs. In ELISA studies, regulation of phosphorylated p38 MAPK and Akt were analyzed after inducing excitotoxicity or neuroprotection in the presence and absence of specific inhibitors for p38 MAPK and PI3K. ELISA results demonstrated that ACh significantly increased phosphorylated Akt and decreased p38 MAPK. Glutamate increased phosphorylated p38 MAPK but had no significant effect on phosphorylated Akt. Other ELISA studies using p38 MAPK and PI3K inhibitors also supported the hypothesis that ACh up-regulated Bcl-2 levels downstream from PI3K and Akt, whereas glutamate down-regulated Bcl-2 levels downstream from p38 MAPK. RGC survival was subsequently assessed by culturing RGCs in conditions to induce excitotoxicity or neuroprotection in the presence or absence of specific inhibitors of p38 MAPK or PI3K. The p38 MAPK inhibitor significantly decreased the number of RGCs that died by glutamate-induced excitotoxicity but had no effect on the number of cells that survived because of ACh-induced neuroprotection. PI3K inhibitors significantly decreased cell survival caused by ACh-induced neuroprotection but had no effect on cell death caused by glutamate-induced excitotoxicity. These results demonstrate that glutamate mediates excitotoxicity through the p38 MAPK signaling pathway and that ACh provides neuroprotection by stimulating the PI3K --> Akt --> Bcl-2 signaling pathway and inhibiting the p38 MAPK --> Bcl-2 pathway.


Subject(s)
Excitatory Amino Acid Agonists/toxicity , Glutamic Acid/toxicity , Receptors, Cholinergic/physiology , Retinal Ganglion Cells/physiology , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Receptors, Cholinergic/chemistry , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Signal Transduction/drug effects , Swine
14.
J Neurophysiol ; 94(5): 3134-42, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16222072

ABSTRACT

In the frog, vestibular efferent fibers innervate only type-II vestibular hair cells. Through this direct contact with hair cells, efferent neurons are capable of modifying transmitter release from hair cells onto primary vestibular afferents. The major efferent transmitter, acetylcholine (ACh), is known to produce distinct pharmacological actions involving several ACh receptors. Previous studies have implicated the presence of muscarinic ACh receptors on vestibular hair cells, although, surprisingly, a muscarinic-mediated electrical response has not been demonstrated in solitary vestibular hair cells. This study demonstrates that muscarinic receptors can evoke transmitter release from vestibular hair cells. Detection of this release was obtained through patch-clamp recordings from catfish cone horizontal cells, serving as glutamate detectors after pairing them with isolated frog semicircular canal hair cells in a two-cell preparation. Although horizontal cells alone failed to respond to carbachol, application of 20 microM carbachol to the two-cell preparation resulted in a horizontal cell response that could be mimicked by exogenous application of glutamate. All of the horizontal cells in the two-cell preparation responded to 20 microM CCh. Furthermore, this presumed transmitter release persisted in the presence of d-tubocurarine at concentrations that block all known hair cell nicotinic ACh receptors. The effect on the detector cell, imparted by the carbachol application to the hair cell-horizontal cell preparation, was blocked both by 2-amino-5-phosphonopentanoic acid, a selective N-methyl-D-aspartate antagonist, and the muscarinic antagonist, atropine. Thus vestibular hair cells from the frog semicircular canal can be stimulated to release transmitter by activating their muscarinic receptors.


Subject(s)
Action Potentials/physiology , Glutamic Acid/metabolism , Hair Cells, Vestibular/physiology , Long-Term Potentiation/physiology , Neurotransmitter Agents/metabolism , Receptors, Muscarinic/metabolism , Synaptic Transmission/physiology , Animals , Catfishes , Cells, Cultured , Ranidae
15.
Invest Ophthalmol Vis Sci ; 45(5): 1531-43, 2004 May.
Article in English | MEDLINE | ID: mdl-15111612

ABSTRACT

PURPOSE: To determine which glutamate receptor (GluR) subtypes are responsible for glutamate-induced excitotoxicity in cultured adult pig retinal ganglion cells (RGCs) and to characterize the neuroprotective effect of acetylcholine (ACh) on pig RGCs. METHODS: Adult pig RGCs were isolated from other retinal tissue by a modified panning technique using Thy 1.1 antibody. Isolated RGCs were cultured in control media and media containing: glutamate, NMDA, or KA; glutamate and CNQX, MK-801, or AP-7; ACh, nicotine or muscarine; ACh and alpha-bungarotoxin (Bgt) or methyllycaconitine (MLA); and glutamate and choline or glutamate, choline, and MLA. To determine cell viability, cells were loaded with calcein and counted. RESULTS: Ninety-eight percent of isolated cells were immunolabeled with Thy 1.1 antibody. Chronic exposure to 500 microM glutamate decreased the number of surviving large and small RGCs, compared to control conditions. This glutamate-induced excitotoxicity was mediated through both NMDA and non-NMDA GluRs. In neuroprotective studies, ACh, nicotine, and choline significantly reduced glutamate-induced excitotoxicity in adult pig RGCs through alpha-Bgt-sensitive nicotinic ACh receptors (nAChRs). DISCUSSION: This was the first report of a modified panning technique to isolate adult pig RGCs. Cell viability was relatively high using this method, and both large and small RGCs grew extensive neurites in culture. The finding that both NMDA and non-NMDA GluRs were involved in glutamate-induced excitotoxicity suggests that isolated pig RGCs provide a good model for glaucoma. In addition, activation of AChRs may be useful in protecting RGC from excitotoxic insults occurring in neurodegenerative diseases such as glaucoma.


Subject(s)
Acetylcholine/pharmacology , Glutamic Acid/toxicity , Neuroprotective Agents/pharmacology , Retinal Ganglion Cells/drug effects , Animals , Bungarotoxins/pharmacology , Cell Separation/methods , Cell Survival/drug effects , Cells, Cultured , Choline/pharmacology , Cytoprotection , Excitatory Amino Acid Agonists/toxicity , Excitatory Amino Acid Antagonists/pharmacology , Fluoresceins/metabolism , Nicotine/pharmacology , Receptors, Nicotinic/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Swine
16.
Am J Physiol Cell Physiol ; 284(5): C1193-204, 2003 May.
Article in English | MEDLINE | ID: mdl-12676655

ABSTRACT

In this study, we investigated the mechanism that links activation of N-methyl-D-aspartate (NMDA) receptors to inhibition of voltage-gated sodium channels in isolated catfish cone horizontal cells. NMDA channels were activated in voltage-clamped cells incubated in low-calcium saline or dialyzed with the calcium chelator BAPTA to determine that calcium influx through NMDA channels is required for sodium channel modulation. To determine whether calcium influx through NMDA channels triggers calcium-induced calcium release (CICR), cells were loaded with the calcium-sensitive dye calcium green 2 and changes in relative fluorescence were measured in response to NMDA. Responses were compared with measurements obtained when caffeine depleted stores. Voltage-clamp studies demonstrated that CICR modulated sodium channels in a manner similar to that of NMDA. Blocking NMDA receptors with AP-7, blocking CICR with ruthenium red, depleting stores with caffeine, or dialyzing cells with calmodulin antagonists W-5 or peptide 290-309 all prevented sodium channel modulation. These results support the hypothesis that NMDA modulation of voltage-gated sodium channels in horizontal cells requires CICR and activation of a calmodulin-dependent signaling pathway.


Subject(s)
Ion Channel Gating , Receptors, N-Methyl-D-Aspartate/physiology , Retina/metabolism , Sodium Channels/physiology , Animals , Calcium/metabolism , Calmodulin/physiology , Electric Conductivity , Electrophysiology , Ictaluridae , Intracellular Membranes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...