Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
J Neurosci ; 35(20): 8021-34, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-25995484

RESUMO

Under physiological conditions, the voltage-gated sodium channel Nav1.8 is expressed almost exclusively in primary sensory neurons. The mechanism restricting Nav1.8 expression is not entirely clear, but we have previously described a 3.7 kb fragment of the Scn10a promoter capable of recapitulating the tissue-specific expression of Nav1.8 in transfected neurons and cell lines (Puhl and Ikeda, 2008). To validate these studies in vivo, a transgenic mouse encoding EGFP under the control of this putative sensory neuron specific promoter was generated and characterized in this study. Approximately 45% of dorsal root ganglion neurons of transgenic mice were EGFP-positive (mean diameter = 26.5 µm). The majority of EGFP-positive neurons bound isolectin B4, although a small percentage (∼10%) colabeled with markers of A-fiber neurons. EGFP expression correlated well with the presence of Nav1.8 transcript (95%), Nav1.8-immunoreactivity (70%), and TTX-R INa (100%), although not all Nav1.8-expressing neurons expressed EGFP. Several cranial sensory ganglia originating from neurogenic placodes, such as the nodose ganglion, failed to express EGFP, suggesting that additional regulatory elements dictate Scn10a expression in placodal-derived sensory neurons. EGFP was also detected in discrete brain regions of transgenic mice. Quantitative PCR and Nav1.8-immunoreactivity confirmed Nav1.8 expression in the amygdala, brainstem, globus pallidus, lateral and paraventricular hypothalamus, and olfactory tubercle. TTX-R INa recorded from EGFP-positive hypothalamic neurons demonstrate the usefulness of this transgenic line to study novel roles of Nav1.8 beyond sensory neurons. Overall, Scn10a-EGFP transgenic mice recapitulate the majority of the Nav1.8 expression pattern in neural crest-derived sensory neurons.


Assuntos
Linhagem da Célula , Proteínas de Fluorescência Verde/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Crista Neural/metabolismo , Regiões Promotoras Genéticas , Animais , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Masculino , Camundongos , Camundongos Transgênicos , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Fibras Nervosas Mielinizadas/metabolismo , Crista Neural/citologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese , Especificidade de Órgãos , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo
2.
Mol Pharmacol ; 88(2): 238-44, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26013543

RESUMO

6-Alkoxy-5-aryl-3-pyridincarboxamides, including the brain-penetrant compound 14G: [5-(4-chlorophenyl)-6-(cyclopropylmethoxy)-N-[(1R,2R)-2-hydroxy-cyclohexyl]-3-pyridinecarboxamide] and its peripherally restricted analog 14H: [5-(4-chlorophenyl)-N-[(1R,2R)-2-hydroxycyclohexyl]-6-(2-methoxyethoxy)-3-pyridinecarboxamide], have been recently introduced as selective, high-affinity antagonists of the human cannabinoid-1 receptor (hCB1R). Binding analyses revealed two orders of magnitude lower affinity of these compounds for mouse and rat versus human CB1R, whereas the affinity of rimonabant is comparable for all three CB1Rs. Modeling of ligand binding to CB1R and binding assays with native and mutant (Ile105Met) hCB1Rs indicate that the Ile105 to Met mutation in rodent CB1Rs accounts for the species-dependent affinity of 14G: and 14H: . Our work identifies Ile105 as a new pharmacophore component for developing better hCB1R antagonists and invalidates rodent models for assessing the antiobesity efficacy of 14G: and 14H: .


Assuntos
Encéfalo/metabolismo , Antagonistas de Receptores de Canabinoides/síntese química , Antagonistas de Receptores de Canabinoides/farmacologia , Niacinamida/análogos & derivados , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/genética , Animais , Antagonistas de Receptores de Canabinoides/química , Células HEK293 , Humanos , Isoleucina/metabolismo , Camundongos , Modelos Moleculares , Niacinamida/síntese química , Niacinamida/química , Niacinamida/farmacologia , Piperidinas/química , Pirazóis/química , Ratos , Receptor CB1 de Canabinoide/metabolismo , Rimonabanto , Especificidade da Espécie , Relação Estrutura-Atividade , Difração de Raios X
3.
J Neurosci ; 33(49): 19314-25, 2013 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-24305827

RESUMO

Free fatty acids receptor 3 (FFA3, GPR41) and 2 (FFA2, GPR43), for which the short-chain fatty acids (SCFAs) acetate and propionate are agonist, have emerged as important G-protein-coupled receptors influenced by diet and gut flora composition. A recent study (Kimura et al., 2011) demonstrated functional expression of FFA3 in the rodent sympathetic nervous system (SNS) providing a potential link between nutritional status and autonomic function. However, little is known of the source of endogenous ligands, signaling pathways, or effectors in sympathetic neurons. In this study, we found that FFA3 and FFA2 are unevenly expressed in the rat SNS with higher transcript levels in prevertebral (e.g., celiac-superior mesenteric and major pelvic) versus paravertebral (e.g., superior cervical and stellate) ganglia. FFA3, whether heterologously or natively expressed, coupled via PTX-sensitive G-proteins to produce voltage-dependent inhibition of N-type Ca(2+) channels (Cav2.2) in sympathetic neurons. In addition to acetate and propionate, we show that ß-hydroxybutyrate (BHB), a metabolite produced during ketogenic conditions, is also an FFA3 agonist. This contrasts with previous interpretations of BHB as an antagonist at FFA3. Together, these results indicate that endogenous BHB levels, especially when elevated under certain conditions, such as starvation, diabetic ketoacidosis, and ketogenic diets, play a potentially important role in regulating the activity of the SNS through FFA3.


Assuntos
Ácido 3-Hidroxibutírico/farmacologia , Canais de Cálcio Tipo N/efeitos dos fármacos , Neurônios/fisiologia , Receptores Acoplados a Proteínas G/agonistas , Sistema Nervoso Simpático/fisiologia , Animais , DNA Complementar/biossíntese , DNA Complementar/genética , Fenômenos Eletrofisiológicos/fisiologia , Transferência Ressonante de Energia de Fluorescência , Gânglios Simpáticos/citologia , Gânglios Simpáticos/efeitos dos fármacos , Gânglios Simpáticos/fisiologia , Células HeLa , Humanos , Hibridização In Situ , Corpos Cetônicos/farmacologia , Ligantes , Masculino , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Sistema Nervoso Simpático/citologia , Transfecção
4.
Mol Pharmacol ; 83(1): 267-82, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23104136

RESUMO

Recent studies propose that N-arachidonyl glycine (NAGly), a carboxylic analogue of anandamide, is an endogenous ligand of the Gα(i/o) protein-coupled receptor 18 (GPR18). However, a high-throughput ß-arrestin-based screen failed to detect activation of GPR18 by NAGly (Yin et al., 2009; JBC, 18:12328). To address this inconsistency, this study investigated GPR18 coupling in a native neuronal system with endogenous signaling pathways and effectors. GPR18 was heterologously expressed in rat sympathetic neurons, and the modulation of N-type (Ca(v)2.2) calcium channels was examined. Proper expression and trafficking of receptor were confirmed by the "rim-like" fluorescence of fluorescently tagged receptor and the positive staining of external hemagglutinin-tagged GPR18-expressing cells. Application of NAGly on GPR18-expressing neurons did not inhibit calcium currents but instead potentiated currents in a voltage-dependent manner, similar to what has previously been reported (Guo et al., 2008; J Neurophysiol, 100:1147). Other proposed agonists of GPR18, including anandamide and abnormal cannabidiol, also failed to induce inhibition of calcium currents. Mutants of GPR18, designed to constitutively activate receptors, did not tonically inhibit calcium currents, indicating a lack of GPR18 activation or coupling to endogenous G proteins. Other downstream effectors of Gα(i/o)-coupled receptors, G protein-coupled inwardly rectifying potassium channels and adenylate cyclase, were not modulated by GPR18 signaling. Furthermore, GPR18 did not couple to other G proteins tested: Gα(s), Gα(z), and Gα(15). These results suggest NAGly is not an agonist for GPR18 or that GPR18 signaling involves noncanonical pathways not examined in these studies.


Assuntos
Ácidos Araquidônicos/farmacologia , Glicina/análogos & derivados , Receptores Acoplados a Proteínas G/agonistas , Animais , Canais de Cálcio Tipo N/fisiologia , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Glicina/farmacologia , Células HEK293 , Células HeLa , Humanos , Técnicas In Vitro , Masculino , Mutação , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Gânglio Cervical Superior/citologia
5.
J Neurophysiol ; 105(1): 442-53, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20962070

RESUMO

Electrically excitable cells have voltage-dependent ion channels on the plasma membrane that regulate membrane permeability to specific ions. Voltage-gated Ca(2+) channels (VGCCs) are especially important as Ca(2+) serves as both a charge carrier and second messenger. Zebrafish (Danio rerio) are an important model vertebrate for studies of neuronal excitability, circuits, and behavior. However, electrophysiological properties of zebrafish VGCCs remain largely unexplored because a suitable preparation for whole cell voltage-clamp studies is lacking. Rohon-Beard (R-B) sensory neurons represent an attractive candidate for this purpose because of their relatively large somata and functional homology to mammalian dorsal root ganglia (DRG) neurons. Transgenic zebrafish expressing green fluorescent protein in R-B neurons, (Isl2b:EGFP)(ZC7), were used to identify dissociated neurons suitable for whole cell patch-clamp experiments. Based on biophysical and pharmacological properties, zebrafish R-B neurons express both high- and low-voltage-gated Ca(2+) current (HVA- and LVA-I(Ca), respectively). Ni(+)-sensitive LVA-I(Ca) occur in the minority of R-B neurons (30%) and ω-conotoxin GVIA-sensitive Ca(V)2.2 (N-type) Ca(2+) channels underlie the vast majority (90%) of HVA-I(Ca). To identify G protein coupled receptors (GPCRs) that modulate HVA-I(Ca), a panel of neurotransmitters was screened. Application of GABA/baclofen or serotonin produced a voltage-dependent inhibition while application of the mu-opioid agonist DAMGO resulted in a voltage-independent inhibition. Unlike in mammalian neurons, GPCR-mediated voltage-dependent modulation of I(Ca) appears to be transduced primarily via a cholera toxin-sensitive Gα subunit. These results provide the basis for using the zebrafish model system to understanding Ca(2+) channel function, and in turn, how Ca(2+) channels contribute to mechanosensory function.


Assuntos
Canais de Cálcio/fisiologia , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/fisiologia , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Baclofeno/farmacologia , Canais de Cálcio/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Proteínas de Fluorescência Verde/genética , Modelos Animais , Neurotransmissores/farmacologia , Técnicas de Patch-Clamp , Ácido gama-Aminobutírico/farmacologia
6.
Neuropharmacology ; 184: 108415, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33275959

RESUMO

Several studies have shown that ethanol (EtOH) can enhance the activity of GABAergic synapses via presynaptic mechanisms, including in hippocampal CA1 neurons. The serotonin type 3 receptor (5-HT3-R) has been implicated in the neural actions of ethanol (EtOH) and in modulation of GABA release from presynaptic terminals. In the present study, we investigated EtOH modulation of GABA release induced by 5-HT3-R activation using the mechanically isolated neuron/bouton preparation from the rat CA1 hippocampal subregion. EtOH application before and during exposure to the selective 5-HT3 receptor agonist, m-chlorophenylbiguanide (mCPBG) potentiated the mCPBG-induced increases in the peak frequency and charge transfer of spontaneous GABAergic inhibitory postsynaptic currents. Interestingly, the potentiation was maintained even after EtOH was removed from the preparation. A protein kinase A inhibitor reduced the magnitude of EtOH potentiation. Fluorescent Ca2+ imaging showed that Ca2+ transients in the presynaptic terminals increased during EtOH exposure. These findings indicate that EtOH produces long-lasting potentiation of 5-HT3-induced GABA release by modulating calcium levels, via a process involving cAMP-mediated signaling in presynaptic terminals.


Assuntos
Região CA1 Hipocampal/metabolismo , Etanol/administração & dosagem , Neurônios/metabolismo , Receptores 5-HT3 de Serotonina/metabolismo , Sinapses/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Animais Recém-Nascidos , Biguanidas/administração & dosagem , Região CA1 Hipocampal/efeitos dos fármacos , Cálcio/metabolismo , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Masculino , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Agonistas do Receptor de Serotonina/administração & dosagem , Sinapses/efeitos dos fármacos
7.
J Neurosci ; 29(43): 13603-12, 2009 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-19864572

RESUMO

Endocannabinoids (eCB) such as 2-arachidonylglycerol (2-AG) are lipid metabolites that are synthesized in a postsynaptic neurons and act upon CB(1) cannabinoid receptors (CB(1)R) in presynaptic nerve terminals. This retrograde transmission underlies several forms of short and long term synaptic plasticity within the CNS. Here, we constructed a model system based on isolated rat sympathetic neurons, in which an eCB signaling cascade could be studied in a reduced, spatially compact, and genetically malleable system. We constructed a complete eCB production/mobilization pathway by sequential addition of molecular components. Heterologous expression of four components was required for eCB production and detection: metabotropic glutamate receptor 5a (mGluR5a), Homer 2b, diacylglycerol lipase alpha, and CB(1)R. In these neurons, application of l-glutamate produced voltage-dependent modulation of N-type Ca(2+) channels mediated by activation of CB(1)R. Using both molecular dissection and pharmacological agents, we provide evidence that activation of mGluR5a results in rapid enzymatic production of 2-AG followed by activation of CB(1)R. These experiments define the critical elements required to recapitulate retrograde eCB production and signaling in a single peripheral neuron. Moreover, production/mobilization of eCB can be detected on a physiologically relevant time scale using electrophysiological techniques. The system provides a platform for testing candidate molecules underlying facilitation of eCB transport across the plasma membrane.


Assuntos
Ácidos Araquidônicos/metabolismo , Glicerídeos/metabolismo , Modelos Neurológicos , Neurônios/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Gânglio Cervical Superior/metabolismo , Animais , Cálcio/metabolismo , Proteínas de Transporte/metabolismo , Endocanabinoides , Ácido Glutâmico/metabolismo , Proteínas de Arcabouço Homer , Humanos , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Masculino , Técnicas de Patch-Clamp , Nervos Periféricos/metabolismo , Ratos , Ratos Wistar , Receptor CB1 de Canabinoide/metabolismo , Receptor de Glutamato Metabotrópico 5 , Transdução de Sinais , Fatores de Tempo
8.
Front Neurosci ; 14: 615362, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33424545

RESUMO

Interoceptive and exteroceptive signals, and the corresponding coordinated control of internal organs and sensory functions, including pain, are received and orchestrated by multiple neurons within the peripheral, central and autonomic nervous systems. A central aim of the present report is to obtain a molecularly informed basis for analgesic drug development aimed at peripheral rather than central targets. We compare three key peripheral ganglia: nodose, sympathetic (superior cervical), and dorsal root ganglia in the rat, and focus on their molecular composition using next-gen RNA-Seq, as well as their neuroanatomy using immunocytochemistry and in situ hybridization. We obtained quantitative and anatomical assessments of transmitters, receptors, enzymes and signaling pathways mediating ganglion-specific functions. Distinct ganglionic patterns of expression were observed spanning ion channels, neurotransmitters, neuropeptides, G-protein coupled receptors (GPCRs), transporters, and biosynthetic enzymes. The relationship between ganglionic transcript levels and the corresponding protein was examined using immunohistochemistry for select, highly expressed, ganglion-specific genes. Transcriptomic analyses of spinal dorsal horn and intermediolateral cell column (IML), which form the termination of primary afferent neurons and the origin of preganglionic innervation to the SCG, respectively, disclosed pre- and post-ganglionic molecular-level circuits. These multimodal investigations provide insight into autonomic regulation, nodose transcripts related to pain and satiety, and DRG-spinal cord and IML-SCG communication. Multiple neurobiological and pharmacological contexts can be addressed, such as discriminating drug targets and predicting potential side effects, in analgesic drug development efforts directed at the peripheral nervous system.

9.
Neuron ; 35(3): 411-2, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12165463

RESUMO

The identity of signaling elements that couple muscarinic acetylcholine receptor (mAChR) activation to M current (KCNQ K(+) channels) modulation has remained unknown despite decades of study. Suh and Hille (in this issue of Neuron) demonstrate that activation of phospholipase C (PLC) initiates M current modulation and that recovery requires ATP and phosphoinositide 4-kinase (PI 4-K). These data suggest that breakdown of phosphotidylinositol 4,5-bisphosphate (PIP(2)) is a crucial determinant of M channel modulation.


Assuntos
Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Receptores Muscarínicos/metabolismo , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Fosfolipases Tipo C/metabolismo , 1-Fosfatidilinositol 4-Quinase/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Humanos , Canais de Potássio KCNQ , Canal de Potássio KCNQ1
10.
J Neurochem ; 106(3): 1209-24, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18466327

RESUMO

Voltage-gated sodium channels (VGSC) are critical membrane components that participate in the electrical activity of excitable cells. The type one VGSC family includes the tetrodotoxin insensitive sodium channel, Na(V)1.8, encoded by the Scn10a gene. Na(V)1.8 expression is restricted to small and medium diameter nociceptive sensory neurons of the dorsal root ganglia and cranial sensory ganglia. To understand the stringent transcriptional regulation of the Scn10a gene, the sensory neuron specific promoter was functionally identified. While identifying the mRNA 5'-end, alternative splicing within the 5'-UTR was observed to create heterogeneity in the RNA transcript. Four kilobases of upstream genomic DNA was cloned and the presence of tissue specific promoter activity was tested by microinjection and adenoviral infection of fluorescent protein reporter constructs into primary mouse and rat neurons, and cell lines. The region contained many putative transcription factor-binding sites and strong homology with the predicted rat ortholog. Homology to the predicted human ortholog was limited to the proximal end and several conserved cis elements were noted. Two regulatory modules were identified by microinjection of reporter constructs into dorsal root ganglia and superior cervical ganglia neurons: a neuron specific proximal promoter region between -1.6 and -0.2 kb of the transcription start site cluster, and a distal sensory neuron switch region beyond -1.6 kb that restricted fluorescent protein expression to a subset of primary sensory neurons.


Assuntos
Neurônios Aferentes/fisiologia , Sequências Reguladoras de Ácido Nucleico/genética , Canais de Sódio/genética , Regiões 5' não Traduzidas/genética , Animais , Sequência de Bases , Linhagem Celular , Linhagem Celular Tumoral , Gânglios Espinais/fisiologia , Células HeLa , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Canal de Sódio Disparado por Voltagem NAV1.8 , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Canais de Sódio/fisiologia
11.
J Pharmacol Exp Ther ; 324(1): 342-51, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17940199

RESUMO

GPR35 is a G protein-coupled receptor recently "de-orphanized" using high-throughput intracellular calcium measurements in clonal cell lines expressing a chimeric G-protein alpha-subunit. From these screens, kynurenic acid, an endogenous metabolite of tryptophan, and zaprinast, a synthetic inhibitor of cyclic guanosine monophosphate-specific phosphodiesterase, emerged as potential agonists for GPR35. To investigate the coupling of GPR35 to natively expressed neuronal signaling pathways and effectors, we heterologously expressed GPR35 in rat sympathetic neurons and examined the modulation of N-type (Ca(V)2.2) calcium channels. In neurons expressing GPR35, calcium channels were inhibited in the absence of overt agonists, indicating a tonic receptor activity. Application of kynurenic acid or zaprinast resulted in robust voltage-dependent calcium current inhibition characteristic of Gbetagamma-mediated modulation. Both agonist-independent and -dependent effects of GPR35 were blocked by Bordetella pertussis toxin pretreatment indicating the involvement of G(i/o) proteins. In neurons expressing GPR35a, a short splice variant of GPR35, zaprinast was more potent (EC(50) = 1 microM) than kynurenic acid (58 microM) but had a similar efficacy (approximately 60% maximal calcium current inhibition). Expression of GPR35b, which has an additional 31 residues at the N terminus, produced similar results but with much greater variability. Both GPR35a and GPR35b appeared to have similar expression patterns when fused to fluorescent proteins. These results suggest a potential role for GPR35 in regulating neuronal excitability and synaptic release.


Assuntos
Canais de Cálcio Tipo N/fisiologia , Neurônios/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Gânglio Cervical Superior/fisiologia , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , DNA Complementar/genética , Células HeLa , Humanos , Ácido Cinurênico/farmacologia , Masculino , Toxina Pertussis , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Purinonas/farmacologia , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética
12.
Sci Rep ; 8(1): 17379, 2018 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-30478340

RESUMO

Activation of short-chain free fatty acid receptors 3 (FFAR3) has been suggested to promote sympathetic outflow in postganglionic sympathetic neurons or hamper it by a negative coupling to N-type calcium (CaV2.2) channels. Heterogeneity of FFAR3 expression in sympathetic neurons, however, renders single neurons studies extremely time-consuming in wild-type mice. Previous studies demonstrated large variability of the degree of CaV2.2 channel inhibition by FFAR3 in a global population of rat sympathetic neurons. Therefore, we focused on a small subpopulation of mouse sympathetic neurons using an FFAR3 antibody and an Ffar3 reporter mouse to perform immunofluorescent and electrophysiological studies. Whole-cell patch-clamp recordings of identified FFAR3-expressing neurons from reporter mice revealed a 2.5-fold decrease in the CaV2.2-FFAR3 inhibitory coupling variability and 1.5-fold increase in the mean ICa2+ inhibition, when compared with unlabeled neurons from wild-type mice. Further, we found that the ablation of Ffar3 gene expression in two knockout mouse models led to a complete loss-of-function. Subpopulations of sympathetic neurons are associated with discrete functional pathways. However, little is known about the neural pathways of the FFAR3-expressing subpopulation. Our data indicate that FFAR3 is expressed primarily in neurons with a vasoconstrictor phenotype. Thus, fine-tuning of chemically-coded neurotransmitters may accomplish an adequate outcome.


Assuntos
Fibras Adrenérgicas/metabolismo , Canais de Cálcio Tipo N/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Feminino , Masculino , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Neurônios , Técnicas de Patch-Clamp/métodos , Transdução de Sinais/fisiologia
13.
J Biomed Opt ; 12(5): 054011, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17994899

RESUMO

We have previously demonstrated that Forster resonance energy transfer (FRET) efficiency and the relative concentration of donor and acceptor fluorophores can be determined in living cells using three-cube wide-field fluorescence microscopy. Here, we extend the methodology to estimate the effective equilibrium dissociation constant (Kd) and the intrinsic FRET efficiency (Emax) of an interacting donor-acceptor pair. Assuming bimolecular interaction, the predicted FRET efficiency is a function of donor concentration, acceptor concentration, Kd, and Emax. We estimate Kd and Emax by minimizing the sum of the squared error (SSE) between the predicted and measured FRET efficiency. This is accomplished by examining the topology of SSE values for a matrix of hypothetical Kd and Emax values. Applying an F-test, the 95% confidence contour of Kd and Emax is calculated. We test the method by expressing an inducible FRET fusion pair consisting of FKBP12-Cerulean and Frb-Venus in HeLa cells. As the Kd for FKBP12-rapamycin and Frb has been analytically determined, the relative Kd (in fluorescence units) could be calibrated with a value based on protein concentration. The described methodology should be useful for comparing protein-protein interaction affinities in living cells.


Assuntos
Algoritmos , Transferência Ressonante de Energia de Fluorescência/métodos , Interpretação de Imagem Assistida por Computador/métodos , Microscopia de Fluorescência/métodos , Proteínas de Neoplasias/metabolismo , Mapeamento de Interação de Proteínas/métodos , Células HeLa , Humanos
14.
J Neurosci ; 25(42): 9762-72, 2005 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-16237180

RESUMO

Rad, Gem/Kir, Rem, and Rem2 are members of the Ras-related RGK (Rad, Gem, and Kir) family of small GTP-binding proteins. Heterologous expression of RGK proteins interferes with de novo calcium channel assembly/trafficking and dramatically decreases the amplitude of currents arising from preexisting high-voltage-activated calcium channels. These effects probably result from the direct interaction of RGK proteins with calcium channel beta subunits. Among the RGK family, Rem2 is the only member abundantly expressed in neuronal tissues. Here, we examined the ability of Rem2 to modulate endogenous voltage-activated calcium channels in rat sympathetic and dorsal root ganglion neurons. Heterologous expression of Rem2 nearly abolished calcium currents arising from preexisting high-voltage-activated calcium channels without affecting low-voltage-activated calcium channels. Rem2 inhibition of N-type calcium channels required both the Ras homology (core) domain and the polybasic C terminus. Mutation of a putative GTP/Mg2+ binding motif in Rem2 did not affect suppression of calcium currents. Loading neurons with GDP-beta-S via the patch pipette did not reverse Rem2-mediated calcium channel inhibition. Finally, [(125)I]Tyr22-omega-conotoxin GVIA cell surface binding in tsA201 cells stably expressing N-type calcium channels was not altered by Rem2 expression at a time when calcium current was totally abolished. Together, our results support a model in which Rem2 localizes to the plasma membrane via a C-terminal polybasic motif and interacts with calcium channel beta subunits in the preassembled N-type channel, thereby forming a nonconducting species.


Assuntos
Bloqueadores dos Canais de Cálcio/metabolismo , Canais de Cálcio Tipo N/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Proteínas Monoméricas de Ligação ao GTP/biossíntese , Animais , Células COS , Bloqueadores dos Canais de Cálcio/farmacologia , Contagem de Células/métodos , Membrana Celular/enzimologia , Células Cultivadas , Chlorocebus aethiops , Masculino , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Neurônios/enzimologia , Ratos , Ratos Wistar , Propriedades de Superfície
16.
Cold Spring Harb Protoc ; 2016(5)2016 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-27140920

RESUMO

Sympathetic neurons isolated from adult rat superior cervical ganglia (SCG) are a well-established model to study G-protein modulation of voltage-gated Ca(2+) channels (VGCCs). SCG neurons can be easily dissociated and are amendable to heterologous expression of genes, including genetic tools to study G-protein signaling pathways, within a time frame to maintain good spatial voltage-clamp control of membrane potential during electrophysiological recordings (8-36 h postdissociation). This protocol focuses on examining G-protein modulation of VGCCs; however, the procedures and experimental setup for acute application of agonists can be applied to study modulation of other ion channels (e.g., M-current, G-protein-coupled inwardly rectifying K(+) channels). We also discuss some common sources of artifacts that can arise during acute drug application onto dissociated neurons, which can mislead interpretation of results.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Neurônios/fisiologia , Técnicas de Patch-Clamp/métodos , Gânglio Cervical Superior/citologia , Animais , Canais de Cálcio Tipo N/genética , Proteínas de Ligação ao GTP/genética , Ratos
17.
Cold Spring Harb Protoc ; 2016(5)2016 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-27140924

RESUMO

G-protein-coupled receptor modulation of voltage-gated ion channels is a common means of fine-tuning the response of channels to changes in membrane potential. Such modulation impacts physiological processes such as synaptic transmission, and hence therapeutic strategies often directly or indirectly target these pathways. As an exemplar of channel modulation, we examine strategies for investigating G-protein modulation of CaV2.2 or N-type voltage-gated Ca(2+) channels. We focus on biochemical and genetic tools for defining the molecular mechanisms underlying the various forms of CaV2.2 channel modulation initiated following ligand binding to G-protein-coupled receptors.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Técnicas Citológicas/métodos , Proteínas de Ligação ao GTP/metabolismo , Biologia Molecular/métodos , Receptores Acoplados a Proteínas G/metabolismo , Animais , Canais de Cálcio Tipo N/genética , Proteínas de Ligação ao GTP/genética , Humanos , Receptores Acoplados a Proteínas G/genética
18.
Sci Rep ; 6: 25137, 2016 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-27118437

RESUMO

Rem2 is a member of the RGK subfamily of RAS small GTPases. Rem2 inhibits high voltage activated calcium channels, is involved in synaptogenesis, and regulates dendritic morphology. Rem2 is the primary RGK protein expressed in the nervous system, but to date, the precise expression patterns of this protein are unknown. In this study, we characterized Rem2 expression in the mouse nervous system. In the CNS, Rem2 mRNA was detected in all regions examined, but was enriched in the striatum. An antibody specific for Rem2 was validated using a Rem2 knockout mouse model and used to show abundant expression in striatonigral and striatopallidal medium spiny neurons but not in several interneuron populations. In the PNS, Rem2 was abundant in a subpopulation of neurons in the trigeminal and dorsal root ganglia, but was absent in sympathetic neurons of superior cervical ganglia. Under basal conditions, Rem2 was subject to post-translational phosphorylation, likely at multiple residues. Further, Rem2 mRNA and protein expression peaked at postnatal week two, which corresponds to the period of robust neuronal maturation in rodents. This study will be useful for elucidating the functions of Rem2 in basal ganglia physiology.


Assuntos
Gânglios da Base/metabolismo , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Camundongos , Sistema Nervoso/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Gânglio Trigeminal/metabolismo
19.
J Neurosci ; 24(21): 5044-53, 2004 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-15163697

RESUMO

Human sensory neuron-specific G-protein-coupled receptors (SNSRs) are expressed solely in small diameter primary sensory neurons. This restricted expression pattern is of considerable therapeutic interest because small nociceptors transmit chronic pain messages. The neuronal function of human SNSRs is difficult to assess because rodent orthologs have yet to be clearly defined, and individual isoforms are found only in a small subset of primary sensory neurons. To circumvent this problem, we expressed human SNSR4 (hSNSR4; also known as Hs.mrgX1) in rat superior cervical ganglion (SCG), dorsal root ganglion (DRG), and hippocampal neurons using nuclear injection or recombinant adenoviruses and examined modulation of ion channels and neurotransmission using whole-cell patch-clamp techniques. BAM8-22 (a 15 amino acid C-terminal fragment of bovine adrenal medulla peptide 22), a peptide agonist derived from proenkephalin, inhibited high (but not low) voltage-activated Ca2+ current in both DRG and SCG neurons expressing hSNSR4, whereas no response was detected in control neurons. The Ca2+ current inhibition was concentration dependent and partially sensitive to Pertussis toxin (PTX) treatment. Additionally, the peptide was highly effective in modulating current arising from M-type K+ channels in SCG neurons expressing hSNSR4. In hippocampal neurons expressing hSNSR4, BAM8-22 induced presynaptic inhibition of transmission that was abolished after PTX treatment. Our data indicate that hSNSR4, when heterologously expressed in rat neurons, can be activated by an opioid-related peptide, couples to G(q/11)-proteins as well as PTX-sensitive G(i/o)-proteins, and modulates neuronal Ca2+ channels, K+ channels, and synaptic transmission.


Assuntos
Canais de Cálcio/metabolismo , Gânglios Espinais/metabolismo , Hipocampo/metabolismo , Neurônios Aferentes/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Gânglio Cervical Superior/metabolismo , Transmissão Sináptica/fisiologia , Animais , Células Cultivadas , Humanos , Técnicas de Patch-Clamp , Ratos , Ratos Wistar
20.
Sci Rep ; 5: 12880, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26260360

RESUMO

FFAR3 (GPR41) is a G-protein coupled receptor for which short-chain fatty acids serve as endogenous ligands. The receptor is found on gut enteroendocrine L-cells, pancreatic ß-cells, and sympathetic neurons, and is implicated in obesity, diabetes, allergic airway disease, and altered immune function. In primates, FFAR3 is segmentally duplicated resulting in GPR42, a gene currently classified as a suspected pseudogene. In this study, we sequenced FFAR3 and GPR42 open reading frames from 56 individuals and found an unexpectedly high frequency of polymorphisms contributing to several complex haplotypes. We also identified a frequent (18.8%) structural variation that results in GPR42 copy number polymorphism. Finally, sequencing revealed that 50.6% of GPR42 haplotypes differed from FFAR3 by only a single non-synonymous substitution and that the GPR42 reference sequence matched only 4.4% of the alleles. Sequencing of cDNA from human sympathetic ganglia and colon revealed processed transcripts matching the GPR42 genotype. Expression of several GPR42 haplotypes in rat sympathetic neurons revealed diverse pharmacological phenotypes that differed in potency and efficacy. Our data suggest that GPR42 be reclassified as a functioning gene and that recognition of sequence and copy number polymorphism of the FFAR3/GPR42 complex be considered during genetic and pharmacological investigation of these receptors.


Assuntos
Variações do Número de Cópias de DNA/genética , Pseudogenes , Receptores Acoplados a Proteínas G/genética , Animais , Gânglios Simpáticos/efeitos dos fármacos , Gânglios Simpáticos/metabolismo , Genótipo , Haplótipos/genética , Humanos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA