Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 114(10): 2741-2746, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28223508

ABSTRACT

Intrinsically photosensitive retinal ganglion cells (ipRGCs) express the photopigment melanopsin and mediate several non-image-forming visual functions, including circadian photoentrainment and the pupillary light reflex (PLR). ipRGCs act as autonomous photoreceptors via the intrinsic melanopsin-based phototransduction pathway and as a relay for rod/cone input via synaptically driven responses. Under low light intensities, where only synaptically driven rod/cone input activates ipRGCs, the duration of the ipRGC response will be determined by the termination kinetics of the rod/cone circuits. Little is known, however, about the termination kinetics of the intrinsic melanopsin-based phototransduction pathway and its contribution to several melanopsin-mediated behaviors. Here, we show that C-terminal phosphorylation of melanopsin determines the recovery kinetics of the intrinsic melanopsin-based photoresponse in ipRGCs, the duration of the PLR, and the speed of reentrainment. In contrast, circadian phase alignment and direct effects of light on activity (masking) are not influenced by C-terminal phosphorylation of melanopsin. Electrophysiological measurements demonstrate that expression of a virally encoded melanopsin lacking all C-terminal phosphorylation sites (C terminus phosphonull) leads to a prolonged intrinsic light response. In addition, mice expressing the C terminus phosphonull in ipRGCs reentrain faster to a delayed light/dark cycle compared with mice expressing virally encoded WT melanopsin; however, the phase angle of entrainment and masking were indistinguishable. Importantly, a sustained PLR in the phosphonull animals is only observed at brighter light intensities that activate melanopsin phototransduction, but not at dimmer light intensities that activate only the rod/cone pathway. Taken together, our results highlight how the kinetics of the melanopsin photoresponse differentially regulate distinct light-mediated behaviors.


Subject(s)
Behavior, Animal , Light Signal Transduction/genetics , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Animals , Circadian Rhythm/genetics , Kinetics , Light , Light Signal Transduction/physiology , Mice , Patch-Clamp Techniques , Phosphorylation/genetics , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/physiology , Reflex, Pupillary/genetics , Reflex, Pupillary/physiology , Retina/metabolism , Retina/physiology , Retinal Ganglion Cells/physiology , Rod Opsins/chemistry , Rod Opsins/genetics , Synapses/genetics , Synapses/metabolism , Vision, Ocular/genetics , Vision, Ocular/physiology
2.
Endocrinology ; 156(8): 2807-20, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25978516

ABSTRACT

Hindbrain catecholamine neurons are required for elicitation of feeding responses to glucose deficit, but the forebrain circuitry required for these responses is incompletely understood. Here we examined interactions of catecholamine and orexin neurons in eliciting glucoprivic feeding. Orexin neurons, located in the perifornical lateral hypothalamus (PeFLH), are heavily innervated by hindbrain catecholamine neurons, stimulate food intake, and increase arousal and behavioral activation. Orexin neurons may therefore contribute importantly to appetitive responses, such as food seeking, during glucoprivation. Retrograde tracing results showed that nearly all innervation of the PeFLH from the hindbrain originated from catecholamine neurons and some raphe nuclei. Results also suggested that many catecholamine neurons project collaterally to the PeFLH and paraventricular hypothalamic nucleus. Systemic administration of the antiglycolytic agent, 2-deoxy-D-glucose, increased food intake and c-Fos expression in orexin neurons. Both responses were eliminated by a lesion of catecholamine neurons innervating orexin neurons using the retrogradely transported immunotoxin, anti-dopamine-ß-hydroxylase saporin, which is specifically internalized by dopamine-ß-hydroxylase-expressing catecholamine neurons. Using designer receptors exclusively activated by designer drugs in transgenic rats expressing Cre recombinase under the control of tyrosine hydroxylase promoter, catecholamine neurons in cell groups A1 and C1 of the ventrolateral medulla were activated selectively by peripheral injection of clozapine-N-oxide. Clozapine-N-oxide injection increased food intake and c-Fos expression in PeFLH orexin neurons as well as in paraventricular hypothalamic nucleus neurons. In summary, catecholamine neurons are required for the activation of orexin neurons during glucoprivation. Activation of orexin neurons may contribute to appetitive responses required for glucoprivic feeding.


Subject(s)
Catecholamines/metabolism , Deoxyglucose/pharmacology , Glucose/deficiency , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/physiology , Neuropeptides/metabolism , Rhombencephalon/metabolism , Animals , Eating/drug effects , Eating/physiology , Glucose/antagonists & inhibitors , Male , Neurons/drug effects , Neurons/metabolism , Orexins , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Rhombencephalon/drug effects
3.
Invest Ophthalmol Vis Sci ; 56(2): 1367-73, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25650413

ABSTRACT

PURPOSE: Administration of voriconazole, an antifungal triazole, causes transient visual disturbances in patients and attenuates the b-wave of the ERG. We sought to identify the retinal target of voriconazole underlying the effect on the ERG b-wave. METHODS: Electroretinograms were recorded from mice before and after intraperitoneal injection of voriconazole. The effect of voriconazole on ON-bipolar cells was tested by patch-clamp recordings of ON-bipolar cells in mouse retinal slices. Effects of voriconazole on mGluR6 and TRPM3 were assessed by patch-clamp recordings of Chinese hamster ovary (CHO) and HEK293 cells transfected with either TRPM3 or mGluR6 plus Kir3.1/Kir3.4. RESULTS: Voriconazole attenuated the ERG b-wave in mice, and inhibited ON-bipolar cell responses evoked by application of CPPG, an mGluR6 antagonist, onto the ON-bipolar cell dendrites, indicating that voriconazole blocks a step in the mGluR6-TRPM1 signal transduction pathway. Voriconazole almost completely blocked capsaicin-activated currents in ON-bipolar cells, which have been attributed to direct activation of the TRPM1 cation channel. Furthermore, application of voriconazole to CHO cells expressing TRPM3, a closely related channel to TRPM1, showed that voriconazole reversibly blocked pregnenolone sulfate-stimulated TRPM3 currents in transfected cells. In contrast, voriconazole only slightly inhibited mGluR6-mediated activation of G-protein activated inward rectifier potassium (GIRK) currents in cotransfected cells, suggesting that mGluR6 is not the primary target of voriconazole in ON-bipolar cells. CONCLUSIONS: The visual disturbances associated with voriconazole are likely due to block of TRPM1 channels in retinal ON-bipolar cells. Other neurological effects of voriconazole may be due to block of TRPM3 channels expressed in the brain.


Subject(s)
Retinal Diseases/metabolism , TRPM Cation Channels/antagonists & inhibitors , Visual Acuity/drug effects , Voriconazole/toxicity , Animals , Antifungal Agents/toxicity , Cells, Cultured , Cricetinae , Dark Adaptation/drug effects , Dark Adaptation/physiology , Disease Models, Animal , Electroretinography , Female , Mice , Retinal Diseases/chemically induced , Retinal Diseases/physiopathology , TRPM Cation Channels/metabolism
4.
PLoS One ; 10(2): e0117615, 2015.
Article in English | MEDLINE | ID: mdl-25679224

ABSTRACT

Transient receptor potential (TRP) channels constitute a large family of cation permeable ion channels that serve crucial functions in sensory systems by transducing environmental changes into cellular voltage and calcium signals. Within the retina, two closely related members of the melastatin TRP family, TRPM1 and TRPM3, are highly expressed. TRPM1 has been shown to be required for the depolarizing response to light of ON-bipolar cells, but the role of TRPM3 in the retina is unknown. Immunohistochemical staining of mouse retina with an antibody directed against the C-terminus of TRPM3 labeled the inner plexiform layer (IPL) and a subset of cells in the ganglion cell layer. Within the IPL, TRPM3 immunofluorescence was markedly stronger in the OFF sublamina than in the ON sublamina. Electroretinogram recordings showed that the scotopic and photopic a- and b-waves of TRPM3(-/-) mice are normal indicating that TRPM3 does not play a major role in visual processing in the outer retina. TRPM3 activity was measured by calcium imaging and patch-clamp recording of immunopurified retinal ganglion cells. Application of the TRPM3 agonist, pregnenolone sulfate (PS), stimulated increases in intracellular calcium in ~40% of cells from wild type and TRPM1(­/­) mice, and the PS-stimulated increases in calcium were blocked by co-application of mefenamic acid, a TRPM3 antagonist. No PS-stimulated changes in fluorescence were observed in ganglion cells from TRPM3(-/-) mice. Similarly, PS-stimulated currents that could be blocked by mefenamic acid were recorded from wild type retinal ganglion cells but were absent in ganglion cells from TRPM3-/- mice.


Subject(s)
Gene Expression , Retina/metabolism , TRPM Cation Channels/genetics , Animals , CHO Cells , Calcium Signaling/drug effects , Cricetinae , Cricetulus , Electroretinography , Mice , Mice, Transgenic , Pregnenolone/pharmacology , Protein Isoforms , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retina/drug effects , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , TRPM Cation Channels/metabolism
5.
Biochemistry ; 52(46): 8352-62, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24164424

ABSTRACT

Cyclic-nucleotide gated (CNG) channels are essential for phototransduction within retinal photoreceptors. We have demonstrated previously that the enzymatic activity of matrix metalloproteinase-2 and -9, members of the matrix metalloproteinase (MMP) family of extracellular, Ca(2+)- and Zn(2+)-dependent proteases, enhances the ligand sensitivity of both rod (CNGA1 and CNGB1) and cone (CNGA3 and CNGB3) CNG channels. Additionally, we have observed a decrease in the maximal CNG channel current (Imax) that begins late during MMP-directed gating changes. Here we demonstrate that CNG channels become nonconductive after prolonged MMP exposure. Concurrent with the loss of conductive channels is the increased relative contribution of channels exhibiting nonmodified gating properties, suggesting the presence of a subpopulation of channels that are protected from MMP-induced gating effects. CNGA subunits are known to possess one extracellular core glycosylation site, located at one of two possible positions within the turret loop near the pore-forming region. Our results indicate that CNGA glycosylation can impede MMP-dependent modification of CNG channels. Furthermore, the relative position of the glycosylation site within the pore turret influences the extent of MMP-dependent proteolysis. Glycosylation at the site found in CNGA3 subunits was found to be protective, while glycosylation at the bovine CNGA1 site was not. Relocating the glycosylation site in CNGA1 to the position found in CNGA3 recapitulated CNGA3-like protection from MMP-dependent processing. Taken together, these data indicate that CNGA glycosylation may protect CNG channels from MMP-dependent proteolysis, consistent with MMP modification of channel function having a requirement for physical access to the extracellular face of the channel.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/metabolism , Ion Channel Gating/drug effects , Matrix Metalloproteinase 9/metabolism , Amino Acid Sequence , Animals , Cattle , Cyclic Nucleotide-Gated Cation Channels/chemistry , Glycosylation , Humans , Matrix Metalloproteinase 2/metabolism , Oocytes/metabolism , Sequence Alignment , Xenopus laevis
6.
PLoS One ; 7(9): e45387, 2012.
Article in English | MEDLINE | ID: mdl-23049792

ABSTRACT

The visual pigment melanopsin is expressed in intrinsically photosensitive retinal ganglion cells (ipRGCs) in the mammalian retina, where it is involved in non-image forming light responses including circadian photoentrainment, pupil constriction, suppression of pineal melatonin synthesis, and direct photic regulation of sleep. It has recently been shown that the melanopsin-based light response in ipRGCs is attenuated by the neurotransmitter dopamine. Here, we use a heterologous expression system to demonstrate that mouse melanopsin can be phosphorylated by protein kinase A, and that phosphorylation can inhibit melanopsin signaling in HEK cells. Site-directed mutagenesis experiments revealed that this inhibitory effect is primarily mediated by phosphorylation of sites T186 and S287 located in the second and third intracellular loops of melanopsin, respectively. Furthermore, we show that this phosphorylation can occur in vivo using an in situ proximity-dependent ligation assay (PLA). Based on these data, we suggest that the attenuation of the melanopsin-based light response by dopamine is mediated by direct PKA phosphorylation of melanopsin, rather than phosphorylation of a downstream component of the signaling cascade.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Retinal Ganglion Cells/enzymology , Rod Opsins/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Dopamine/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , HEK293 Cells , Humans , Light , Light Signal Transduction/drug effects , Light Signal Transduction/radiation effects , Mice , Models, Molecular , Mutagenesis, Site-Directed , Phosphorylation , Phylogeny , Protein Structure, Secondary , Protein Structure, Tertiary , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/radiation effects , Rod Opsins/classification , Rod Opsins/genetics , Serine/genetics , Serine/metabolism , Threonine/genetics , Threonine/metabolism , Transfection
7.
Channels (Austin) ; 6(3): 181-96, 2012.
Article in English | MEDLINE | ID: mdl-22699690

ABSTRACT

Photoreceptor cyclic nucleotide-gated (CNG) channels are the principal ion channels responsible for transduction of the light-induced change in cGMP concentration into an electrical signal. The ligand sensitivity of photoreceptor CNG channels is subject to regulation by intracellular signaling effectors, including calcium-calmodulin, tyrosine kinases and phosphoinositides. Little is known, however, about regulation of channel activity by modification to extracellular regions of CNG channel subunits. Extracellular proteases MMP9 and -2 are present in the interphotoreceptor matrix adjacent to photoreceptor outer segments. Given that MMPs have been implicated in retinal dysfunction and degeneration, we hypothesized that MMP activity may alter the functional properties of photoreceptor CNG channels. For heterologously expressed rod and cone CNG channels, extracellular exposure to MMPs dramatically increased the apparent affinity for cGMP and the efficacy of cAMP. These changes to ligand sensitivity were not prevented by destabilization of the actin cytoskeleton or by disruption of integrin mediated cell adhesion, but could be attenuated by inhibition of MMP catalytic activity. MMP-mediated gating changes exhibited saturable kinetic properties consistent with enzymatic processing of the CNG channels. In addition, exposure to MMPs decreased the abundance of full-length expressed CNGA3 subunits, with a concomitant increase in putative degradation products. Similar gating effects and apparent proteolysis were observed also for native rod photoreceptor CNG channels. Furthermore, constitutive apparent proteolysis of retinal CNGA1 and retinal MMP9 levels were both elevated in aged mice compared with young mice. Together, these results provide evidence that MMP-mediated proteolysis can regulate the ligand sensitivity of CNG channels.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Age Factors , Allosteric Site , Animals , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cytoskeletal Proteins/metabolism , Humans , Ion Channel Gating , Ligands , Mice , Mice, Inbred C57BL , Oocytes , Protein Subunits/metabolism , Proteolysis , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Xenopus
8.
Biochem J ; 441(2): 685-96, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21992067

ABSTRACT

Mutations that perturb the function of photoreceptor CNG (cyclic nucleotide-gated) channels are associated with several human retinal disorders, but the molecular and cellular mechanisms leading to photoreceptor dysfunction and degeneration remain unclear. Many loss-of-function mutations result in intracellular accumulation of CNG channel subunits. Accumulation of proteins in the ER (endoplasmic reticulum) is known to cause ER stress and trigger the UPR (unfolded protein response), an evolutionarily conserved cellular programme that results in either adaptation via increased protein processing capacity or apoptotic cell death. We hypothesize that defective trafficking of cone photoreceptor CNG channels can induce UPR-mediated cell death. To test this idea, CNGA3 subunits bearing the R563H and Q655X mutations were expressed in photoreceptor-derived 661W cells with CNGB3 subunits. Compared with wild-type, R563H and Q655X subunits displayed altered degradation rates and/or were retained in the ER. ER retention was associated with increased expression of UPR-related markers of ER stress and with decreased cell viability. Chemical and pharmacological chaperones {TUDCA (tauroursodeoxycholate sodium salt), 4-PBA (sodium 4-phenylbutyrate) and the cGMP analogue CPT-cGMP [8-(4-chlorophenylthio)-cGMP]} differentially reduced degradation and/or promoted plasma-membrane localization of defective subunits. Improved subunit maturation was concordant with reduced expression of ER-stress markers and improved viability of cells expressing localization-defective channels. These results indicate that ER stress can arise from expression of localization-defective CNG channels, and may represent a contributing factor for photoreceptor degeneration.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/genetics , Endoplasmic Reticulum/physiology , Retinal Cone Photoreceptor Cells/physiology , Unfolded Protein Response/drug effects , Animals , Cell Death/drug effects , Cell Line , Cell Membrane/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Mice , Molecular Chaperones/pharmacology , Protein Transport/drug effects , Transcription Factor CHOP/metabolism
9.
Behav Brain Funct ; 6: 36, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20604961

ABSTRACT

BACKGROUND: In mammals, the brain clock responsible for generating circadian rhythms is located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Light entrainment of the clock occurs through intrinsically photosensitive retinal ganglion cells (ipRGCs) whose axons project to the SCN via the retinohypothalamic tract. Although ipRGCs are sufficient for photoentrainment, rod and cone photoreceptors also contribute. Adult CBA/J mice, which exhibit loss of rod and cone photoreceptors during early postnatal development, have greater numbers of ipRGCs compared to CBA/N control mice. A greater number of photosensitive cells might argue for enhanced light responses, however, these mice exhibit attenuated phase shifting behaviors. To reconcile these findings, we looked for potential differences in SCN neurons of CBA/J mice that might underly the altered circadian behaviors. We hypothesized that CBA/J mice have differences in the expression of neuropeptides in the SCN, where ipRGCs synapse. The neuropeptides vasoactive intestinal peptide (VIP) and vasopressin (VP) are expressed by many SCN neurons and play an important role in the generation of circadian rhythms and photic entrainment. METHODS: Using immunohistochemistry, we looked for differences in the expression of VIP and VP in the SCN of CBA/J mice, and using a light-induced FOS assay, we also examined the degree of retinal innervation of the SCN by ipRGCs. RESULTS: Our data demonstrate greater numbers of VIP-and VP-positive cells in the SCN of CBA/J mice and a greater degree of light-induced FOS expression. CONCLUSIONS: These results implicate changes in neuropeptide expression in the SCN which may underlie the altered circadian responses to light in these animals.


Subject(s)
Retinal Degeneration/metabolism , Suprachiasmatic Nucleus/metabolism , Vasoactive Intestinal Peptide/metabolism , Vasopressins/metabolism , Age Factors , Animals , Cell Count , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Immunohistochemistry , Male , Mice , Mice, Inbred CBA , Mice, Mutant Strains , Photic Stimulation , Proto-Oncogene Proteins c-fos/metabolism , Retina/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Visual Pathways/pathology
10.
Vis Neurosci ; 27(1-2): 9-17, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20100392

ABSTRACT

The rate-limiting step in the recovery of the photoreceptor light response is the hydrolysis of GTP by transducin, a reaction that is accelerated by the RGS9-Gbeta5 complex, and its membrane anchor, R9AP. Similar complexes, including RGS7, RGS11, and Gbeta5, are found in retinal ON-bipolar cell dendrites. Here, we present evidence that R9AP is also expressed in the dendritic tips of ON-bipolar cells. Immunofluorescent staining for R9AP revealed a punctate pattern of labeling in the outer plexiform layer, where it colocalized with mGluR6. In photoreceptors, R9AP is required for proteolytic stability of the entire regulator of G protein signaling complex, and we found that genetic deletion of R9AP also results in a marked reduction in the levels of RGS11 and Gbeta5 in the bipolar cell dendrites; the level of RGS7 was unaffected, suggesting the presence of another interaction partner to stabilize RGS7. To determine the effect of R9AP deletion on the response kinetics of ON-bipolar cells, we compared the electroretinogram (ERG) between wild-type and R9AP-deficient mice. The ERG b-wave, reflecting ON-bipolar cell activity, was delayed and larger in the R9AP-deficient mice. Our data indicate that R9AP is required for stable expression of RGS11-Gbeta5 in ON-bipolar cell dendrites. Furthermore, they suggest that the RGS11-Gbeta5-R9AP complex accelerates the initial ON-bipolar cell response to light.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , Gene Expression Regulation/genetics , Light , Membrane Proteins/physiology , Retina/cytology , Retinal Bipolar Cells/physiology , Animals , Cell Line, Transformed , Dendrites/metabolism , Dendrites/ultrastructure , Electroretinography/methods , Evoked Potentials/genetics , Humans , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission/methods , Reaction Time/genetics , Retinal Bipolar Cells/ultrastructure , Synapses/metabolism , Synapses/ultrastructure , Transfection/methods
11.
Invest Ophthalmol Vis Sci ; 51(2): 1121-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19797214

ABSTRACT

PURPOSE: The retinal ON-bipolar cell (ON-BPC) light response is initiated upon deactivation of the metabotropic glutamate receptor mGluR6 and the G protein Go. G protein-based signaling cascades are typically accelerated by interaction of the G protein alpha subunit with a member of the regulator of G protein signaling (RGS) protein family. The goal of this study was to determine whether RGS7 and/or -11 serve this function in retinal ON-BPCs. METHODS: Retinas from mice lacking RGS11 (RGS11(-/-)), or with a deletion mutation in RGS7 (RGS7(Delta/Delta)), or both, were compared to wild-type (WT) by immunofluorescence confocal microscopy. The retinal light response was measured with the electroretinogram (ERG). The kinetics of simulated light responses from individual rod bipolar cells were recorded by whole-cell patch-clamp electrophysiology. RESULTS: Levels of the R7 RGS interaction partners, Gbeta5 and R9AP, were reduced in the outer plexiform layer of the RGS11(-/-) and RGS7(Delta/Delta)/RGS11(-/-) mice. ERG recordings demonstrated a delay in the rising phase of the ERG b-wave, larger photopic b-wave amplitudes, and increased scotopic threshold response sensitivity in the RGS11(-/-) and RGS7(Delta/Delta)/RGS11(-/-) mice. The ERG measured from the RGS7(Delta/Delta) retina was normal. Patch-clamp recordings of chemically simulated light responses of rod BPCs revealed a 25-ms delay in the onset of the ON-BPC response in the RGS7(Delta/Delta)/RGS11(-/-) mouse compared with the WT. CONCLUSIONS: RGS11 plays a role in the deactivation of Galphao, which precedes activation of the depolarizing current in ON-BPCs. RGS7 must also serve a role as changes in RGS7(Delta/Delta)/RGS11(-/-) mice were greater than those in RGS11(-/-) mice.


Subject(s)
Light , RGS Proteins/physiology , Retinal Bipolar Cells/radiation effects , Signal Transduction/physiology , Animals , Blotting, Western , Electrophysiology , Electroretinography , Genotype , Guanosine Triphosphate/metabolism , Hydrolysis , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Patch-Clamp Techniques , Photic Stimulation , Receptors, Metabotropic Glutamate/metabolism , Retinal Bipolar Cells/metabolism
12.
Proc Natl Acad Sci U S A ; 106(45): 19174-8, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19861548

ABSTRACT

The ON pathway of the visual system, which detects increases in light intensity, is established at the first retinal synapse between photoreceptors and ON-bipolar cells. Photoreceptors hyperpolarize in response to light and reduce the rate of glutamate release, which in turn causes the depolarization of ON-bipolar cells. This ON-bipolar cell response is mediated by the metabotropic glutamate receptor, mGluR6, which controls the activity of a depolarizing current. Despite intensive research over the past two decades, the molecular identity of the channel that generates this depolarizing current has remained elusive. Here, we present evidence indicating that TRPM1 is necessary for the depolarizing light response of ON-bipolar cells, and further that TRPM1 is a component of the channel that generates this light response. Gene expression profiling revealed that TRPM1 is highly enriched in ON-bipolar cells. In situ hybridization experiments confirmed that TRPM1 mRNA is found in cells of the retinal inner nuclear layer, and immunofluorescent confocal microscopy showed that TRPM1 is localized in the dendrites of ON-bipolar cells in both mouse and macaque retina. The electroretinogram (ERG) of TRPM1-deficient (TRPM1(-/-)) mice had a normal a-wave, but no b-wave, indicating a loss of bipolar cell response. Finally, whole-cell patch-clamp recording from ON-bipolar cells in mouse retinal slices demonstrated that genetic deletion of TRPM1 abolished chemically simulated light responses from rod bipolar cells and dramatically altered the responses of cone ON-bipolar cells. Identification of TRPM1 as a mGluR6-coupled cation channel reveals a key step in vision, expands the role of the TRP channel family in sensory perception, and presents insights into the evolution of vertebrate vision.


Subject(s)
Light , Photoreceptor Cells, Vertebrate/metabolism , Retinal Bipolar Cells/metabolism , TRPM Cation Channels/metabolism , Vision, Ocular/physiology , Animals , Dendrites/metabolism , Electroretinography , Gene Expression Profiling , In Situ Hybridization , Mice , Mice, Transgenic , Microscopy, Fluorescence , Patch-Clamp Techniques , Photoreceptor Cells, Vertebrate/physiology , Retinal Bipolar Cells/physiology
13.
Eur J Neurosci ; 29(2): 359-67, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19200239

ABSTRACT

In mammals, the neuronal pathways by which rod and cone photoreceptors mediate vision have been well documented. The roles that classical photoreceptors play in photoentrainment, however, have been less clear. In mammals, intrinsically photosensitive retinal ganglion cells (ipRGCs) that express the photopigment melanopsin project directly to the suprachiasmatic nucleus of the hypothalamus, the site of the circadian clock, and thereby contribute to non-image-forming responses to light. Classical photoreceptors are not necessary for photoentrainment as loss of rods and cones does not eliminate light entrainment. Conflicting evidence arose, however, when attenuated phase-shifting responses were observed in the retinal-degenerate CBA/J mouse. In this study, we examined the time course of retinal degeneration in CBA/J mice and used these animals to determine if maturation of the outer retina regulates the morphology, number and distribution of ipRGCs. We also examined whether degeneration during the early development of the outer retina can alter the function of the adult circadian system. We report that dendritic stratification and distribution of ipRGCs was unaltered in mice with early retinal degeneration, suggesting that normal development of the outer retina was not necessary for these processes. We found, however, that adult CBA/J mice have greater numbers of ipRGCs than controls, implicating a role for the outer retinal photoreceptors in regulating developmental cell death of ipRGCs.


Subject(s)
Cell Differentiation/genetics , Retinal Degeneration/metabolism , Retinal Ganglion Cells/metabolism , Rod Opsins/metabolism , Animals , Animals, Newborn , Apoptosis/physiology , Cell Shape/physiology , Circadian Rhythm/physiology , Dendrites/physiology , Dendrites/ultrastructure , Disease Models, Animal , Male , Mice , Mice, Inbred CBA , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurogenesis/physiology , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/physiology , Retina/cytology , Retina/growth & development , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology , Retinal Ganglion Cells/cytology , Rod Opsins/genetics
14.
Acta Crystallogr D Biol Crystallogr ; 64(Pt 10): 1034-42, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18931410

ABSTRACT

Cyclic nucleotide-gated (CNG) ion channels play pivotal roles in sensory transduction by retinal photoreceptors and olfactory neurons. The elapid snake toxins pseudechetoxin (PsTx) and pseudecin (Pdc) are the only known protein blockers of CNG channels. These toxins belong to a cysteine-rich secretory protein (CRISP) family containing an N-terminal pathogenesis-related proteins of group 1 (PR-1) domain and a C-terminal cysteine-rich domain (CRD). PsTx and Pdc are highly homologous proteins, but their blocking affinities on CNG channels are different: PsTx blocks both the olfactory and retinal channels with approximately 15-30-fold higher affinity than Pdc. To gain further insights into their structure and function, the crystal structures of PsTx, Pdc and Zn2+-bound Pdc were determined. The structures revealed that most of the amino-acid-residue differences between PsTx and Pdc are located around the concave surface formed between the PR-1 domain and the CRD, suggesting that the concave surface is functionally important for CNG-channel binding and inhibition. A structural comparison in the presence and absence of Zn2+ ion demonstrated that the concave surface can open and close owing to movement of the CRD upon Zn2+ binding. The data suggest that PsTx and Pdc occlude the pore entrance and that the dynamic motion of the concave surface facilitates interaction with the CNG channels.


Subject(s)
Cysteine/chemistry , Ion Channel Gating , Ion Channels/chemistry , Snake Venoms/chemistry , Amino Acid Sequence , Cell Line , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid
15.
Proc Natl Acad Sci U S A ; 105(26): 8861-5, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18579788

ABSTRACT

In mammals, melanopsin is exclusively expressed in intrinsically photosensitive retinal ganglion cells (ipRGCs), which play an important role in circadian photoentrainment and other nonimage-forming functions. These ipRGCs reside in the inner retina, far removed from the pigment epithelium, which synthesizes the 11-cis retinal chromophore used by rod and cone photoreceptors to regenerate opsin for light detection. There has been considerable interest in the identification of the melanopsin chromophore and in understanding the process of photopigment regeneration in photoreceptors that are not in proximity to the classical visual cycle. We have devised an immuno-magnetic purification protocol that allows melanopsin-expressing retinal ganglion cells to be isolated and collected from multiple mouse retinas. Using this technique, we have demonstrated that native melanopsin in vivo exclusively binds 11-cis retinal in the dark and that illumination causes isomerization to the all-trans isoform. Furthermore, spectral analysis of the melanopsin photoproduct shows the formation of a protonated metarhodopsin with a maximum absorbance between 520 and 540 nm. These results indicate that even if melanopsin functions as a bistable photopigment with photo-regenerative activity native melanopsin must also use some other light-independent retinoid regeneration mechanism to return to the dark state, where all of the retinal is observed to be in the 11-cis form.


Subject(s)
Photochemistry , Retina/metabolism , Rod Opsins/metabolism , Animals , Cell Separation , Chromatography, High Pressure Liquid , Gene Expression Regulation , Immunohistochemistry , Immunomagnetic Separation , Mice , Retina/cytology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Rod Opsins/genetics , Spectrum Analysis
16.
Eur J Neurosci ; 26(10): 2899-905, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18001285

ABSTRACT

The time course of G-protein-coupled responses is largely determined by the kinetics of GTP hydrolysis by the G protein alpha subunit, which is accelerated by interaction with regulator of G-protein signaling (RGS) proteins. Light responses of ON-bipolar cells of the vertebrate retina require rapid inactivation of the G protein Galphao, which is activated in the dark by metabotropic glutamate receptor, mGluR6, in their dendritic tips. It is not yet known, however, which RGS protein(s) might be responsible for rapid inactivation kinetics. By immunofluorescence and co-immunoprecipitation, we have identified complexes of the Galphao-selective RGS proteins RGS7 and RGS11, with their obligate binding partner, Gbeta5, that are localized to the dendritic tips of murine rod and cone ON-bipolar cells, along with mGluR6. Experiments using pre- and post-synaptic markers, and a dissociated bipolar cell preparation, clearly identified the location of these complexes as the ON-bipolar cell dendritic tips and not the adjacent photoreceptor terminals or horizontal cell dendrites. In mice lacking mGluR6, the distribution of RGS11, RGS7 and Gbeta5 shifts away from the dendritic tips, implying a functional relationship with mGluR6. The precise co-localization of Gbeta5-RGS7 and Gbeta5-RGS11 with mGluR6, and the dependence of localization on the presence of mGluR6, suggests that Gbeta5-RGS7 and Gbeta5-RGS11 function specifically in the mGluR6 signal transduction pathway, where they may stimulate the GTPase activity of Galphao, thus accelerating the ON-bipolar cell light response, in a manner analogous to the acceleration of photoreceptor light responses by the Gbeta5-RGS9-1 complex.


Subject(s)
GTP-Binding Protein Regulators/metabolism , GTP-Binding Protein beta Subunits/metabolism , Receptors, Metabotropic Glutamate/metabolism , Retina/cytology , Retinal Bipolar Cells/metabolism , Animals , Immunoprecipitation/methods , Mice , Protein Kinase C-alpha/metabolism , Retinal Bipolar Cells/ultrastructure
17.
Curr Pharm Des ; 12(28): 3597-613, 2006.
Article in English | MEDLINE | ID: mdl-17073662

ABSTRACT

Cyclic nucleotide-gated (CNG) ion channels play a central role in vision and olfaction, generating the electrical responses to light in photoreceptors and to odorants in olfactory receptors. These channels have been detected in many other tissues where their functions are largely unclear. The use of gene knockouts and other methods have yielded some information, but there is a pressing need for potent and specific pharmacological agents directed at CNG channels. To date there has been very little systematic effort in this direction - most of what can be termed CNG channel pharmacology arose from testing reagents known to target protein kinases or other ion channels, or by accident when researchers were investigating other intracellular pathways that may regulate the activity of CNG channels. Predictably, these studies have not produced selective agents. However, taking advantage of emerging structural information and the increasing knowledge of the biophysical properties of these channels, some promising compounds and strategies have begun to emerge. In this review we discuss progress on two fronts, cyclic nucleotide analogs as both activators and competitive inhibitors, and inhibitors that target the pore or gating machinery of the channel. We also discuss the potential of these compounds for treating certain forms of retinal degeneration.


Subject(s)
Ion Channels/antagonists & inhibitors , Ion Channels/metabolism , Cyclic Nucleotide-Gated Cation Channels , Retinal Degeneration
18.
Proc Natl Acad Sci U S A ; 103(42): 15635-40, 2006 Oct 17.
Article in English | MEDLINE | ID: mdl-17032767

ABSTRACT

Phosphatidylinositol-3,4,5-trisphosphate (PIP3) has been proposed to modulate the odorant sensitivity of olfactory sensory neurons by inhibiting activation of cyclic nucleotide-gated (CNG) channels in the cilia. When applied to the intracellular face of excised patches, PIP3 has been shown to inhibit activation of heteromeric olfactory CNG channels, composed of CNGA2, CNGA4, and CNGB1b subunits, and homomeric CNGA2 channels. In contrast, we discovered that channels formed by CNGA3 subunits from cone photoreceptors were unaffected by PIP3. Using chimeric channels and a deletion mutant, we determined that residues 61-90 within the N terminus of CNGA2 are necessary for PIP3 regulation, and a biochemical "pulldown" assay suggests that PIP3 directly binds this region. The N terminus of CNGA2 contains a previously identified calcium-calmodulin (Ca2+/CaM)-binding domain (residues 68-81) that mediates Ca2+/CaM inhibition of homomeric CNGA2 channels but is functionally silent in heteromeric channels. We discovered, however, that this region is required for PIP3 regulation of both homomeric and heteromeric channels. Furthermore, PIP3 occluded the action of Ca2+/CaM on both homomeric and heteromeric channels, in part by blocking Ca2+/CaM binding. Our results establish the importance of the CNGA2 N terminus for PIP3 inhibition of olfactory CNG channels and suggest that PIP3 inhibits channel activation by disrupting an autoexcitatory interaction between the N and C termini of adjacent subunits. By dramatically suppressing channel currents, PIP3 may generate a shift in odorant sensitivity that does not require prior channel activity.


Subject(s)
Calmodulin/metabolism , Ion Channels/metabolism , Olfactory Receptor Neurons/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Subunits/metabolism , Animals , Calcium/metabolism , Cattle , Cell Line , Cyclic GMP/metabolism , Cyclic Nucleotide-Gated Cation Channels , Humans , Ion Channels/genetics , Patch-Clamp Techniques , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Subunits/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Cone Photoreceptor Cells/metabolism
19.
Eur J Neurosci ; 24(4): 1117-23, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16930437

ABSTRACT

Melanopsin-containing retinal ganglion cells (RGCs) project to the suprachiasmatic nuclei (SCN) and mediate photoentrainment of the circadian system. Melanopsin is a novel retinal-based photopigment that renders these cells intrinsically photosensitive (ip). Although genetic ablation of melanopsin abolishes the intrinsic light response, it has a surprisingly minor effect on circadian photoentrainment. This and other non-visual responses to light are lost only when the melanopsin deficiency is coupled with mutations that disable classical rod and cone photoreceptors, suggesting that melanopsin-containing RGCs also receive rod- and cone-driven synaptic inputs. Using whole-cell patch-clamp recording, we demonstrate that light triggers synaptic currents in ipRGCs via activation of ionotropic glutamate and gamma-aminobutyric acid (GABA) receptors. Miniature postsynaptic currents (mPSCs) were clearly observed in ipRGCs, although they were less robust and were seen less frequently than those seen in non-ip cells. Pharmacological treatments revealed that the majority of ipRGCs receive excitatory glutamatergic inputs that were blocked by DNQX and/or kynurenic acid, as well as inhibitory GABAergic inputs that were blocked by bicuculline. Other ipRGCs received either glutamatergic or GABAergic inputs nearly exclusively. Although strychnine (Strych)-sensitive mPSCs were evident on many non-ipRGCs, indicating the presence of glycinergic inputs, we saw no evidence of Strych-sensitive events in ipRGCs. Based on these results, it is clear that SCN-projecting RGCs can respond to light both via an intrinsic melanopsin-based signaling cascade and via a synaptic pathway driven by classical rod and/or cone photoreceptors. It remains to be determined how the ipRGCs integrate these temporally distinct inputs to generate the signals that mediate circadian photoentrainment and other non-visual responses to light.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Neural Pathways , Retinal Ganglion Cells/metabolism , Synapses/metabolism , Animals , Bicuculline/metabolism , Excitatory Amino Acid Antagonists/metabolism , GABA Antagonists/metabolism , Glycine Agents/metabolism , Kynurenic Acid/metabolism , Light , Neural Pathways/metabolism , Neural Pathways/physiology , Patch-Clamp Techniques , Quinoxalines/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA/metabolism , Receptors, Glutamate/metabolism , Retinal Ganglion Cells/cytology , Rod Opsins/metabolism , Strychnine/metabolism , Synaptic Transmission/physiology , Tetrodotoxin/metabolism
20.
Curr Biol ; 16(12): 1211-6, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16782012

ABSTRACT

Nonmotile cilia on olfactory sensory neurons (OSNs) compartmentalize signaling molecules, including odorant receptors and cyclic nucleotide-gated (CNG) channels, allowing for efficient, spatially confined responses to sensory stimuli . Little is known about the mechanisms of the ciliary targeting of olfactory CNG channels, composed of three subunits: CNGA2, CNGA4, and CNGB1b . Recent reports suggest that subunit composition of the retinal CNG channel influences localization, leading to disease . However, the mechanistic role of subunits in properly targeting native olfactory CNG channels remains unclear. Here, we show that heteromeric assembly with CNGB1b, containing a critical carboxy-terminal motif (RVxP), is required for ciliary trafficking of olfactory CNG channels. Movement of proteins within the cilia is governed by intraflagellar transport (IFT), a process that facilitates bidirectional movement of cargo along microtubules. Work in C. elegans has established that heterotrimeric and homodimeric kinesin-2 family members play a critical role in anterograde transport . In mammalian systems, the heterotrimeric KIF3a/KIF3b/KAP-3 complex plays a clear role in IFT; however, no role has been established for KIF17, the mammalian homolog of OSM-3 . Here, we demonstrate that KIF17 is required for olfactory CNG channel targeting, providing novel insights into mechanisms of mammalian ciliary transport.


Subject(s)
Cilia/metabolism , Ion Channels/metabolism , Kinesins/metabolism , Molecular Motor Proteins/metabolism , Protein Subunits/metabolism , Amino Acid Sequence , Animals , Cell Line , Cilia/ultrastructure , Cyclic Nucleotide-Gated Cation Channels , Dogs , Ion Channels/chemistry , Luminescent Proteins/analysis , Molecular Sequence Data , Protein Transport , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...