Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 314(2): C233-C241, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29141923

RESUMO

Pituitary adenylate cyclase activating polypeptide (PACAP, Adcyap1) activation of PAC1 receptors ( Adcyap1r1) significantly increases excitability of guinea pig cardiac neurons. This modulation of excitability is mediated in part by plasma membrane G protein-dependent activation of adenylyl cyclase and downstream signaling cascades. However, additional mechanisms responsible for the enhanced excitability are activated following internalization of the PAC1 receptor and endosomal signaling. Src family kinases play critical roles mediating endocytosis of many trophic factor and G protein-coupled receptors. The present study investigated whether Src family kinases also support the PACAP-induced PAC1 receptor internalization, phosphorylation of ERK, and enhanced neuronal excitability. Using human embryonic kidney cells stably expressing a green fluorescent protein-tagged PAC1 receptor, treatment with the Src family kinase inhibitor PP2 (10 µM) markedly reduced the PACAP-induced PAC1 receptor internalization, and in parallel, both PP2 and Src inhibitor 1 (Src-1, 2 µM) reduced ERK activation determined by Western blot analysis. In contrast, Src family kinase inhibitors did not eliminate a PACAP-induced rise in global calcium generated by inositol (1,4,5)-trisphosphate-induced release of calcium from endoplasmic reticulum stores. From confocal analysis of phosphorylated ERK immunostaining, PP2 treatment significantly attenuated PACAP activation of ERK in neurons within cardiac ganglia whole mount preparations. Intracellular recordings demonstrated that PP2 also significantly blunted a PACAP-induced increase in cardiac neuron excitability. These studies demonstrate Src-related kinase activity in PAC1 receptor internalization, activation of MEK/ERK signaling, and regulation of neuronal excitability. The present results provide further support for the importance of PAC1 receptor endosomal signaling as a key mechanism regulating cellular function.


Assuntos
Endocitose/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Coração/inervação , Neurônios/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/agonistas , Quinases da Família src/antagonistas & inibidores , Animais , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Ativação Enzimática , Feminino , Cobaias , Humanos , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neurônios/enzimologia , Fosforilação , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Quinases da Família src/metabolismo
2.
Am J Physiol Cell Physiol ; 313(2): C219-C227, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28592413

RESUMO

Forskolin, a selective activator of adenylyl cyclase (AC), commonly is used to establish actions of G protein-coupled receptors (GPCRs) that are initiated primarily through activation of AC/cAMP signaling pathways. In the present study, forskolin was used to evaluate the potential role of AC/cAMP, which is a major signaling mechanism for the pituitary adenylate cyclase-activating polypeptide (PACAP)-selective PAC1 receptor, in the regulation of guinea pig cardiac neuronal excitability. Forskolin (5-10 µM) increases excitability in ~60% of the cardiac neurons. The forskolin-mediated increase in excitability was considered related to cAMP regulation of a cyclic nucleotide gated channel or via protein kinase A (PKA)/ERK signaling, mechanisms that have been linked to PAC1 receptor activation. However, unlike PACAP mechanisms, forskolin enhancement of excitability was not significantly reduced by treatment with cesium to block currents through hyperpolarization-activated nonselective cation channels (Ih) or by treatment with PD98059 to block MEK/ERK signaling. In contrast, treatment with the clathrin inhibitor Pitstop2 or the dynamin inhibitor dynasore eliminated the forskolin-induced increase in excitability; treatments with the inactive Pitstop analog or PP2 treatment to inhibit Src-mediated endocytosis mechanisms were ineffective. The PKA inhibitor KT5702 significantly suppressed the forskolin-induced change in excitability; further, KT5702 and Pitstop2 reduced the forskolin-stimulated MEK/ERK activation in cardiac neurons. Collectively, the present results suggest that forskolin activation of AC/cAMP/PKA signaling leads to the recruitment of clathrin/dynamin-dependent endosomal transduction cascades, including MEK/ERK signaling, and that endosomal signaling is the critical mechanism underlying the forskolin-induced increase in cardiac neuron excitability.


Assuntos
Adenilil Ciclases/metabolismo , Colforsina/administração & dosagem , Coração/efeitos dos fármacos , Miocárdio/metabolismo , Neurônios/efeitos dos fármacos , Animais , Carbazóis/administração & dosagem , Clatrina/efeitos dos fármacos , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Flavonoides/administração & dosagem , Cobaias , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Miocárdio/patologia , Neurônios/metabolismo , Neurônios/patologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Pirróis/administração & dosagem , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo
3.
Am J Physiol Cell Physiol ; 311(4): C643-C651, 2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27488668

RESUMO

Pituitary adenylate cyclase (PAC)-activating polypeptide (PACAP) peptides (Adcyap1) signaling at the selective PAC1 receptor (Adcyap1r1) participate in multiple homeostatic and stress-related responses, yet the cellular mechanisms underlying PACAP actions remain to be completely elucidated. PACAP/PAC1 receptor signaling increases excitability of neurons within the guinea pig cardiac ganglia, and as these neurons are readily accessible, this neuronal system is particularly amenable to study of PACAP modulation of ionic conductances. The present study investigated how PACAP activation of MEK/ERK signaling contributed to the peptide-induced increase in cardiac neuron excitability. Treatment with the MEK inhibitor PD 98059 blocked PACAP-stimulated phosphorylated ERK and, in parallel, suppressed the increase in cardiac neuron excitability. However, PD 98059 did not blunt the ability of PACAP to enhance two inward ionic currents, one flowing through hyperpolarization-activated nonselective cationic channels (Ih) and another flowing through low-voltage-activated calcium channels (IT), which support the peptide-induced increase in excitability. Thus a PACAP- and MEK/ERK-sensitive, voltage-dependent conductance(s), in addition to Ih and IT, modulates neuronal excitability. Despite prior work implicating PACAP downregulation of the KV4.2 potassium channel in modulation of excitability in other cells, treatment with the KV4.2 current blocker 4-aminopyridine did not replicate the PACAP-induced increase in excitability in cardiac neurons. However, cardiac neurons express the ERK target, the NaV1.7 sodium channel, and treatment with the selective NaV1.7 channel inhibitor PF-04856264 decreased the PACAP modulation of excitability. From these results, PACAP/PAC1 activation of MEK/ERK signaling may phosphorylate the NaV1.7 channel, enhancing sodium currents near the threshold, an action contributing to repetitive firing of the cardiac neurons exposed to PACAP.


Assuntos
Potenciais de Ação/fisiologia , Coração/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Transdução de Sinais/fisiologia , Animais , Canais de Cálcio/metabolismo , Feminino , Cobaias , Masculino , Miocárdio/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Canais de Potássio Shal/metabolismo
4.
Am J Physiol Cell Physiol ; 308(11): C857-66, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25810261

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a potent intercellular signaling molecule involved in multiple homeostatic functions. PACAP/PAC1 receptor signaling increases excitability of neurons within the guinea pig cardiac ganglia, making them a unique system to establish mechanisms underlying PACAP modulation of neuronal function. Calcium influx is required for the PACAP-increased cardiac neuron excitability, although the pathway is unknown. This study tested whether PACAP enhancement of calcium influx through either T-type or R-type channels contributed to the modulation of excitability. Real-time quantitative polymerase chain reaction analyses indicated transcripts for Cav3.1, Cav3.2, and Cav3.3 T-type isoforms and R-type Cav2.3 in cardiac neurons. These neurons often exhibit a hyperpolarization-induced rebound depolarization that remains when cesium is present to block hyperpolarization-activated nonselective cationic currents (Ih). The T-type calcium channel inhibitors, nickel (Ni(2+)) or mibefradil, suppressed the rebound depolarization, and treatment with both drugs hyperpolarized cardiac neurons by 2-4 mV. Together, these results are consistent with the presence of functional T-type channels, potentially along with R-type channels, in these cardiac neurons. Fifty micromolar Ni(2+), a concentration that suppresses currents in both T-type and R-type channels, blunted the PACAP-initiated increase in excitability. Ni(2+) also blunted PACAP enhancement of the hyperpolarization-induced rebound depolarization and reversed the PACAP-mediated increase in excitability, after being initiated, in a subset of cells. Lastly, low voltage-activated currents, measured under perforated patch whole cell recording conditions and potentially flowing through T-type or R-type channels, were enhanced by PACAP. Together, our results suggest that a PACAP-enhanced, Ni(2+)-sensitive current contributes to PACAP-induced modulation of neuronal excitability.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Níquel/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Animais , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo R/genética , Canais de Cálcio Tipo R/metabolismo , Canais de Cálcio Tipo T/genética , Canais de Cálcio Tipo T/metabolismo , Feminino , Expressão Gênica , Cobaias , Masculino , Mibefradil/farmacologia , Microeletrodos , Miocárdio/citologia , Miocárdio/metabolismo , Neurônios/citologia , Técnicas de Patch-Clamp , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Análise de Célula Única
5.
Am J Physiol Renal Physiol ; 305(10): F1504-12, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24049141

RESUMO

Quantitative real-time PCR was used to test whether cavernous nerve injury leads to a decrease in major pelvic ganglia (MPG) neuronal nicotinic ACh receptor (nAChR) subunit and postsynaptic density (PSD)-93 transcript levels. Subunits α3, ß4, and α7, commonly expressed in the MPG, were selected for analysis. After 72 h in explant culture, MPG transcript levels for α3, ß4, α7, and PSD-93 were significantly depressed. Three days after cavernous nerve axotomy or crush in vivo, transcript levels for α3, ß4, and PSD-93, but not for α7, were significantly depressed. Three days after dissection of the cavernous nerve free of underlying tissue and application of a 5-mm lateral stretch (manipulation), transcript levels for α3 and PSD-93 were also significantly decreased. Seven days after all three surgical procedures, α3 transcript levels remained depressed, but PSD-93 transcript levels were still decreased only after axotomy or nerve crush. At 30 days postsurgery, transcript levels for the nAChR subunits and PSD-93 had recovered. ACh-induced currents were significantly smaller in MPG neurons dissociated from 3-day explant cultured ganglia than from those recorded in neurons dissociated from acutely isolated ganglia; this observation provides direct evidence showing that a decrease in nAChR function was coincident with a decrease in nAChR subunit transcript levels. We conclude that a downregulation of nAChR subunit and PSD-93 expression after cavernous nerve injury, or even manipulation, could interrupt synaptic transmission within the MPG and thus contribute to the loss of neural control of urogenital organs after pelvic surgeries.


Assuntos
Gânglios Autônomos/metabolismo , Guanilato Quinases/metabolismo , Plexo Hipogástrico/metabolismo , Proteínas de Membrana/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo , RNA Mensageiro/metabolismo , Receptores Nicotínicos/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Acetilcolina/metabolismo , Animais , Modelos Animais de Doenças , Regulação para Baixo , Galanina/genética , Galanina/metabolismo , Guanilato Quinases/genética , Plexo Hipogástrico/lesões , Masculino , Potenciais da Membrana , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Traumatismos dos Nervos Periféricos/genética , Receptores Nicotínicos/genética , Transmissão Sináptica , Fatores de Tempo , Técnicas de Cultura de Tecidos , Receptor Nicotínico de Acetilcolina alfa7/genética
6.
J Neurosci ; 33(10): 4614-22, 2013 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-23467377

RESUMO

After G-protein-coupled receptor activation and signaling at the plasma membrane, the receptor complex is often rapidly internalized via endocytic vesicles for trafficking into various intracellular compartments and pathways. The formation of signaling endosomes is recognized as a mechanism that produces sustained intracellular signals that may be distinct from those generated at the cell surface for cellular responses including growth, differentiation, and survival. Pituitary adenylate cyclase activating polypeptide (PACAP; Adcyap1) is a potent neurotransmitter/neurotrophic peptide and mediates its diverse cellular functions in part through internalization of its cognate G-protein-coupled PAC1 receptor (PAC1R; Adcyap1r1). In the present study, we examined whether PAC1R endocytosis participates in the regulation of neuronal excitability. Although PACAP increased excitability in 90% of guinea pig cardiac neurons, pretreatment with Pitstop 2 or dynasore to inhibit clathrin and dynamin I/II, respectively, suppressed the PACAP effect. Subsequent addition of inhibitor after the PACAP-induced increase in excitability developed gradually attenuated excitability with no changes in action potential properties. Likewise, the PACAP-induced increase in excitability was markedly decreased at ambient temperature. Receptor trafficking studies with GFP-PAC1 cell lines demonstrated the efficacy of Pitstop 2, dynasore, and low temperatures at suppressing PAC1R endocytosis. In contrast, brefeldin A pretreatments to disrupt Golgi vesicle trafficking did not blunt the PACAP effect, and PACAP/PAC1R signaling still increased neuronal cAMP production even with endocytic blockade. Our results demonstrate that PACAP/PAC1R complex endocytosis is a key step for the PACAP modulation of cardiac neuron excitability.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Miocárdio/citologia , Neurônios/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Animais Recém-Nascidos , Compostos de Bário/farmacologia , Biofísica , Brefeldina A/farmacologia , Células Cultivadas , Cloretos/farmacologia , AMP Cíclico/metabolismo , Esquema de Medicação , Estimulação Elétrica , Endossomos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Cobaias , Humanos , Hidrazonas/farmacologia , Técnicas In Vitro , Masculino , Neurônios/fisiologia , Técnicas de Patch-Clamp , Ratos , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Transdução de Sinais/fisiologia , Sulfonamidas/farmacologia , Gânglio Cervical Superior/citologia , Temperatura , Tiazolidinas/farmacologia , Transfecção
7.
J Mol Neurosci ; 48(3): 721-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22528456

RESUMO

Calcium influx is required for the pituitary adenylyl cyclase activating polypeptide (PACAP)-induced increase in guinea pig cardiac neuron excitability, noted as a change from a phasic to multiple action potential firing pattern. Intracellular recordings indicated that pretreatment with the nonselective cationic channel inhibitors, 2-aminoethoxydiphenylborate (2-APB), 1-[ß-[3-(4-methoxyphenyl)propoxy]-4-methoxyphenethyl]-1H-imidazole HCl (SKF 96365), and flufenamic acid (FFA) reduced the 20-nM PACAP-induced excitability increase. Additional experiments tested whether 2-APB, FFA, and SKF 96365 could suppress the increase in excitability by PACAP once it had developed. The increased action potential firing remained following application of 2-APB but was diminished by FFA. SKF 96365 transiently depressed the PACAP-induced excitability increase. A decrease and recovery of action potential amplitude paralleled the excitability shift. Since semiquantitative PCR indicated that cardiac neurons express TRPC subunit transcripts, we hypothesize that PACAP activates calcium-permeable, nonselective cationic channels, which possibly are members of the TRPC family. Our results are consistent with calcium influx being required for the initiation of the PACAP-induced increase in excitability, but suggest that it may not be required to sustain the peptide effect. The present results also demonstrate that nonselective cationic channel inhibitors could have other actions, which might contribute to the inhibition of the PACAP-induced excitability increase.


Assuntos
Compostos de Boro/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Ácido Flufenâmico/farmacologia , Sistema de Condução Cardíaco/efeitos dos fármacos , Imidazóis/farmacologia , Canais Iônicos/efeitos dos fármacos , Moduladores de Transporte de Membrana/farmacologia , Neurônios/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/antagonistas & inibidores , Canais de Cátion TRPC/antagonistas & inibidores , Potenciais de Ação/efeitos dos fármacos , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/fisiologia , Feminino , Cobaias , Sistema de Condução Cardíaco/citologia , Transporte de Íons/efeitos dos fármacos , Microdissecção e Captura a Laser , Masculino , Neurônios/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Reação em Cadeia da Polimerase/métodos , Análise de Célula Única , Canais de Cátion TRPC/biossíntese , Canais de Cátion TRPC/genética
8.
Am J Physiol Cell Physiol ; 299(4): C836-43, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20668213

RESUMO

Prior studies indicated that a Ca(2+)-dependent release of ATP can be initiated from the soma of sympathetic neurons dissociated from guinea pig stellate ganglia. Previous studies also indicated that Ca(2+)-induced Ca(2+) release (CICR) can modulate membrane excitability in these same neurons. As Ca(2+) release from internal stores is thought to support somatodendritic transmitter release in other neurons, the present study investigated whether CICR is essential for somatic ATP release from dissociated sympathetic neurons. Caffeine increased intracellular Ca(2+) and activated two inward currents: a slow inward current (SIC) in 85% of cells, and multiple faster inward currents [asynchronous transient inward currents (ASTICs)] in 40% of cells voltage-clamped to negative potentials. Caffeine evoked both currents when cells were bathed in a Ca(2+)-deficient solution, indicating that both were initiated by Ca(2+) release from ryanodine-sensitive stores in the endoplasmic reticulum. Sodium influx contributed to generation of both SICs and ASTICs, but only ASTICs were inhibited by the presence of the P2X receptor blocker PPADs. Thus ASTICs, but not SICs, resulted from an ATP activation of P2X receptors. Ionomycin induced ASTICs in a Ca(2+)-containing solution, but not when it was applied in a Ca(2+)-deficient solution, demonstrating the key requirement for external Ca(2+) in initiating ASTICs by ionomycin. Pretreatment with drugs to deplete the internal stores of Ca(2+) did not block the ability of ionomycin or long depolarizing voltage steps to initiate ASTICs. Although a caffeine-induced release of Ca(2+) from internal stores can elicit both SICs and ASTICs in dissociated sympathetic neurons, CICR is not required for the somatic release of ATP.


Assuntos
Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Neurônios/fisiologia , Gânglio Estrelado/citologia , Animais , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Cobaias , Ionomicina/farmacologia , Ionóforos/farmacologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Rianodina/farmacologia , Sódio/metabolismo , Gânglio Estrelado/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
9.
Ann N Y Acad Sci ; 1070: 317-21, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16888185

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) effects on intracellular calcium ([Ca2+]i) and excitability have been studied in adult guinea pig intracardiac neurons. PACAP increased excitability, but did not elicit Ca2+ release from intracellular stores. Exposure to a Ca2+-deficient solution did not deplete [Ca2+]i stores but did eliminate the PACAP-induced increase in excitability. We postulate that Ca2+ influx is required for the PACAP-induced increase in excitability.


Assuntos
Cálcio/metabolismo , Coração/efeitos dos fármacos , Canais Iônicos/metabolismo , Miocárdio/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Animais , Citosol/efeitos dos fármacos , Citosol/metabolismo , Cobaias , Ativação do Canal Iônico
10.
J Neurophysiol ; 95(4): 2134-42, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16371456

RESUMO

Mechanisms modulating the pituitary adenylate cyclase activating polypeptide (PACAP)-induced increase in excitability have been studied using dissociated guinea pig intrinsic cardiac neurons and intact ganglion preparations. Measurements of intracellular calcium (Ca2+) with the fluorescent Ca2+ indicator dye fluo-3 indicated that neither PACAP nor vasoactive intestinal polypeptide (VIP) at either 100 nM or 1 microM produced a discernible elevation of intracellular Ca2+ in dissociated intracardiac neurons. For neurons in ganglion whole mount preparations kept in control bath solution, local application of PACAP significantly increased excitability, as indicated by the number of action potentials generated by long depolarizing current pulses. However, in a Ca2+ -deficient solution in which external Ca2+ was replaced by Mg2+ or when cells were bathed in control solution containing 200 microM Cd2+, PACAP did not enhance action potential firing. In contrast, in a Ca2+ -deficient solution with Ca2+ replaced by strontium (Sr2+), PACAP increased excitability. PACAP increased excitability in cells treated with a combination of 20 microM ryanodine and 10 mM caffeine to interrupt release of Ca2+ from internal stores. Experiments using fluo-3 showed that ryanodine/caffeine pretreatment eliminated subsequent caffeine-induced Ca2+ release from intracellular stores, whereas exposure to the Ca2+ -deficient solution did not. In dissociated intracardiac neurons voltage clamped with the perforated patch recording technique, 100 nM PACAP decreased the voltage-dependent barium current (IBa). These results show that, in the guinea pig intracardiac neurons, the PACAP-induced increase in excitability apparently requires Ca2+ influx through Cd2+ -sensitive calcium permeable channels other than voltage-dependent Ca2+ channels, but not Ca2+ release from internal stores.


Assuntos
Cálcio/metabolismo , Coração/inervação , Neurônios/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Bário/metabolismo , Cádmio/farmacologia , Cafeína/farmacologia , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/fisiologia , Eletrofisiologia , Feminino , Cobaias , Magnésio/metabolismo , Masculino , Neurônios/química , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Rianodina/farmacologia , Estrôncio/metabolismo , Peptídeo Intestinal Vasoativo/farmacologia
11.
Regul Pept ; 123(1-3): 123-33, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15518902

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) peptides, which are co-localized with acetylcholine in preganglionic parasympathetic fibers innervating guinea pig intracardiac ganglia, depolarize and increase excitability of intracardiac neurons. Perforated patch whole cell recordings were used to test whether PACAP27-enhanced activation of Ih contributed to the increase in excitability. In current clamp, 100 nM PACAP27 increased rectification during 500-ms hyperpolarizations and increased the number of anodal break action potentials (APs). PACAP27 also increased the number of APs produced by 500-ms depolarizing currents. In voltage clamp, the effects of 100 nM PACAP27 were determined during hyperpolarizing steps from -50 mV to voltages between -60 and -120 mV. PACAP27 increased the amplitude and rate of activation of Ih. PACAP27 shifted the voltage dependence of activation of Ih by 6.6 mV. The effect of PACAP27 was eliminated by pretreatment with the Ih inhibitor ZD7288 (100 microM). The adenylyl cyclase activator forskolin (10 microM) produced a similar shift in the voltage dependence of Ih activation. We conclude that PACAP27 enhances Ih by shifting the voltage dependence of activation and propose that this effect is mediated primarily by PAC1 receptor activation of adenylyl cyclase and generation of cAMP. Furthermore, we propose that the peptide-enhanced Ih contributes to the PACAP27-induced increase in membrane excitability.


Assuntos
Coração/inervação , Fatores de Crescimento Neural/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/farmacologia , Neurotransmissores/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Colforsina/farmacologia , Condutividade Elétrica , Feminino , Cobaias , Coração/efeitos dos fármacos , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Miocárdio/citologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Pirimidinas/farmacologia
12.
J Physiol ; 555(Pt 3): 627-35, 2004 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-14724192

RESUMO

Experiments were done using guinea-pig sympathetic neurones dissociated from the stellate ganglia to establish whether calcium-induced calcium release (CICR) modulated action potential (AP) generation in mammalian neurones. Using measurements of intracellular calcium ([Ca(2+)](i)) with the Ca(2+)-sensitive dye fluo-3, we demonstrated that 10 mM caffeine activated ryanodine receptors and caused a rise in [Ca(2+)](i) in both Ca(2+)-containing and Ca(2+)-deficient solutions. We also demonstrated that combined treatment with caffeine and 1 microm thapsigargin or caffeine and 20 microm ryanodine blocked subsequent caffeine-induced elevations of [Ca(2+)](i). Treatment with thapsigargin, ryanodine or 200 microM Cd(2+) to disrupt CICR decreased the latency to AP generation during 400 ms depolarizing current ramps using the perforated patch whole cell patch clamp in current clamp mode. Treatment with 500 microM tetraethylammonium also decreased the latency to AP generation during depolarizing current ramps in control cells, but not in cells pretreated with thapsigargin to deplete internal Ca(2+) stores. In summary, we propose that an outward current, carried at least in part through BK channels, is activated by CICR at membrane voltages approaching the threshold for AP initiation and that this current opposed depolarizing current ramps applied to guinea-pig sympathetic stellate neurones.


Assuntos
Cálcio/metabolismo , Gânglios Simpáticos/fisiologia , Neurônios/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Cádmio/farmacologia , Cafeína/farmacologia , Gânglios Simpáticos/citologia , Gânglios Simpáticos/efeitos dos fármacos , Gânglios Simpáticos/metabolismo , Cobaias , Membranas Intracelulares/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Concentração Osmolar , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Cálcio-Ativados/fisiologia , Tempo de Reação/efeitos dos fármacos , Rianodina/farmacologia , Tetraetilamônio/farmacologia , Tapsigargina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...