Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Med ; 46(2): 782-794, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32468069

RESUMO

Nav1.7 is closely associated with neuropathic pain. Hydrogen sulfide (H2S) has recently been reported to be involved in numerous biological functions, and it has been shown that H2S can enhance the sodium current density, and inhibiting the endogenous production of H2S mediated by cystathionine ß­synthetase (CBS) using O­(carboxymethyl)hydroxylamine hemihydrochloride (AOAA) can significantly reduce the expression of Nav1.7 and thus the sodium current density in rat dorsal root ganglion (DRG) neurons. In the present study, it was shown that the fluorescence intensity of H2S was increased in a spared nerve injury (SNI) model and AOAA inhibited this increase. Nav1.7 is expressed in DRG neurons, and the expression of CBS and Nav1.7 were increased in DRG neurons 7, 14 and 21 days post­operation. AOAA inhibited the increase in the expression of CBS, phosphorylated (p)­MEK1/2, p­ERK1/2 and Nav1.7 induced by SNI, and U0126 (a MEK blocker) was able to inhibit the increase in p­MEK1/2, p­ERK1/2 and Nav1.7 expression. However, PF­04856264 did not inhibit the increase in CBS, p­MEK1/2, p­ERK1/2 or Nav1.7 expression induced by SNI surgery. The current density of Nav1.7 was significantly increased in the SNI model and administration of AOAA and U0126 both significantly decreased the density. In addition, AOAA, U0126 and PF­04856264 inhibited the decrease in rheobase, and the increase in action potential induced by SNI in DRG neurons. There was no significant difference in thermal withdrawal latency among each group. However, the time the animals spent with their paw lifted increased significantly following SNI, and the time the animals spent with their paw lifted decreased significantly following the administration of AOAA, U0126 and PF­04856264. In conclusion, these data show that Nav1.7 expression in DRG neurons is upregulated by CBS­derived endogenous H2S in an SNI model, contributing to the maintenance of neuropathic pain.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Neuralgia/metabolismo , Animais , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Regulação para Cima/genética , Regulação para Cima/fisiologia
2.
Neuropharmacology ; 103: 174-82, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26188144

RESUMO

Prolactin (PRL) is a polypeptide hormone produced and released from the pituitary and extrapituitary tissues. It regulates activity of nociceptors and causes hyperalgesia in pain conditions, but little is known the molecular mechanism. We report here that PRL can exert a potentiating effect on the functional activity of acid-sensing ion channels (ASICs), key sensors for extracellular protons. First, PRL dose-dependently increased the amplitude of ASIC currents with an EC50 of (5.89 ± 0.28) × 10(-8) M. PRL potentiation of ASIC currents was also pH dependent. Second, PRL potentiation of ASIC currents was blocked by Δ1-9-G129R-hPRL, a PRL receptor antagonist, and removed by intracellular dialysis of either protein kinase C inhibitor GF109203X, protein interacting with C-kinase 1(PICK1) inhibitor FSC-231, or PI3K inhibitor AS605240. Third, PRL altered acidosis-evoked membrane excitability of DRG neurons and caused a significant increase in the amplitude of the depolarization and the number of spikes induced by acid stimuli. Four, PRL exacerbated nociceptive responses to injection of acetic acid in female rats. Finally, PRL displayed a stronger effect on ASIC mediated-currents and nociceptive behavior in intact female rats than OVX female and male rats and thus modulation of PRL may be gender-dependent. These results suggest that PRL up-regulates the activity of ASICs and enhances ASIC mediated nociceptive responses in female rats, which reveal a novel peripheral mechanism underlying PRL involvement in hyperalgesia.


Assuntos
Canais Iônicos Sensíveis a Ácido/fisiologia , Gânglios Espinais/fisiologia , Prolactina/fisiologia , Células Receptoras Sensoriais/fisiologia , Ácido Acético/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Feminino , Gânglios Espinais/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Prolactina/análogos & derivados , Prolactina/farmacologia , Prótons , Ratos , Ratos Sprague-Dawley , Receptores da Prolactina/antagonistas & inibidores , Receptores da Prolactina/fisiologia , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
3.
Eur J Pharmacol ; 767: 24-9, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26435025

RESUMO

Prokineticin 2 (PK2), a new chemokine, causes mechanical hypersensitivity in the rat hind paw, but little is known about the molecular mechanism. Here, we have found that ionotropic P2X receptor is essential to mechanical allodynia induced by PK2. First, intraplantar injection of high dose (3 or 10 pmol) of PK2 significantly increased paw withdrawal response frequency (%) to innocuous mechanical stimuli (mechanical allodynia). And the mechanical allodynia induced by PK2 was prevented by co-administration of TNP-ATP, a selective P2X receptor antagonist. Second, although low dose (0.3 or 1 pmol) of PK2 itself did not produce an allodynic response, it significantly facilitated the mechanical allodynia evoked by intraplantar injection of α,ß-methylene ATP (α,ß-meATP). Third, PK2 concentration-dependently potentiated α,ß-meATP-activated currents in rat dorsal root ganglion (DRG) neurons. Finally, PK2 receptors and intracellular signal transduction were involved in PK2 potentiation of α,ß-meATP-induced mechanical allodynia and α,ß-meATP-activated currents, since the potentiation were blocked by PK2 receptor antagonist PKRA and selective PKC inhibitor GF 109203X. These results suggested that PK2 facilitated mechanical allodynia induced by α,ß-meATP through a mechanism involved in sensitization of cutaneous P2X receptors expressed by nociceptive nerve endings.


Assuntos
Trifosfato de Adenosina/análogos & derivados , Hormônios Gastrointestinais/farmacologia , Hiperalgesia/induzido quimicamente , Neuropeptídeos/farmacologia , Trifosfato de Adenosina/efeitos adversos , Trifosfato de Adenosina/farmacologia , Animais , Sinergismo Farmacológico , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/fisiologia , Hormônios Gastrointestinais/antagonistas & inibidores , Hiperalgesia/fisiopatologia , Indóis/farmacologia , Masculino , Maleimidas/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neuropeptídeos/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , Antagonistas do Receptor Purinérgico P2X/farmacologia , Ratos , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de Peptídeos/antagonistas & inibidores , Receptores Purinérgicos P2X3/efeitos dos fármacos , Receptores Purinérgicos P2X3/fisiologia
4.
Endocrinology ; 156(12): 4660-71, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26441237

RESUMO

Sex differences have been reported in a number of pain conditions. Women are more sensitive to most types of painful stimuli than men, and estrogen plays a key role in the sex differences in pain perception. However, it is unclear whether there is a sex difference in acidosis-evoked pain. We report here that both male and female rats exhibit nociceptive behaviors in response to acetic acid, with females being more sensitive than males. Local application of exogenous 17ß-estradiol (E2) exacerbated acidosis-evoked nociceptive response in male rats. E2 and estrogen receptor (ER)-α agonist 1,3,5-Tris(4-hydroxyphenyl)-4-propyl-1H-pyrazole, but not ERß agonist 2,3-bis(4-hydroxyphenyl)-propionitrile, replacement also reversed attenuation of the acetic acid-induced nociceptive response in ovariectomized females. Moreover, E2 can exert a rapid potentiating effect on the functional activity of acid-sensing ion channels (ASICs), which mediated the acidosis-induced events. E2 dose dependently increased the amplitude of ASIC currents with a 42.8 ± 1.6 nM of EC50. E2 shifted the concentration-response curve for proton upward with a 50.1% ± 6.2% increase of the maximal current response to proton. E2 potentiated ASIC currents via an ERα and ERK1/2 signaling pathway. E2 also altered acidosis-evoked membrane excitability of dorsal root ganglia neurons and caused a significant increase in the amplitude of the depolarization and the number of spikes induced by acidic stimuli. E2 potentiation of the functional activity of ASICs revealed a peripheral mechanism underlying this sex difference in acetic acid-induced nociception.


Assuntos
Ácido Acético/farmacologia , Canais Iônicos Sensíveis a Ácido/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Estradiol/farmacologia , Estrogênios/farmacologia , Nociceptividade/efeitos dos fármacos , Nociceptores/efeitos dos fármacos , Percepção da Dor/efeitos dos fármacos , Células Receptoras Sensoriais/efeitos dos fármacos , Canais Iônicos Sensíveis a Ácido/metabolismo , Acidose , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nitrilas/farmacologia , Nociceptores/metabolismo , Técnicas de Patch-Clamp , Fenóis/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/metabolismo , Fatores Sexuais
5.
Neurosci Lett ; 593: 61-5, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25782631

RESUMO

Arginine vasopressin (AVP) plays a regulatory role in nociception. Intrathecal administration of AVP displays an antinociceptive effect. However, little is understood about the mechanism underlying spinal AVP analgesia. Here, we have found that spinal AVP dose dependently reduced the second, but not first, phase of formalin-induced spontaneous nociception in mice. The AVP analgesia was completely blocked by intrathecal injected SR 49059, a vasopressin-1A (V1A) receptor antagonist. However, spinal AVP failed to exert its antinociceptive effect on the second phase formalin-induced spontaneous nociception in V1A receptor knock-out (V1A-/-) mice. The AVP analgesia was also reversed by bicuculline, a GABAA receptor antagonist. Moreover, AVP potentiated GABA-activated currents in dorsal root ganglion neurons from wild-type littermates, but not from V1A-/- mice. Our results may reveal a novel spinal mechanism of AVP analgesia by enhancing the GABAA receptor function in the spinal cord through V1A receptors.


Assuntos
Arginina Vasopressina/metabolismo , Nociceptividade/efeitos dos fármacos , Dor Nociceptiva/psicologia , Receptores de GABA-A/metabolismo , Receptores de Vasopressinas/metabolismo , Medula Espinal/metabolismo , Analgésicos/farmacologia , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos/farmacologia , Arginina Vasopressina/farmacologia , Antagonistas de Receptores de GABA-A/farmacologia , Gânglios Espinais/fisiopatologia , Injeções Espinhais , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/fisiologia , Dor Nociceptiva/metabolismo , Dor Nociceptiva/fisiopatologia , Medição da Dor , Receptores de Vasopressinas/genética , Medula Espinal/efeitos dos fármacos
6.
J Neurosci Res ; 93(2): 333-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25395088

RESUMO

Levo-tetrahydropalmatine (l-THP), a main bioactive Chinese herbal constituent from the genera Stephania and Corydalis, has been in use in clinical practice for years in China as a traditional analgesic agent. However, the mechanism underlying the analgesic action of l-THP is poorly understood. This study shows that l-THP can exert an inhibitory effect on the functional activity of native acid-sensing ion channels (ASICs), which are believed to mediate pain caused by extracellular acidification. l-THP dose dependently decreased the amplitude of proton-gated currents mediated by ASICs in rat dorsal root ganglion (DRG) neurons. l-THP shifted the proton concentration-response curve downward, with a decrease of 40.93% ± 8.45% in the maximum current response to protons, with no significant change in the pH0.5 value. Moreover, l-THP can alter the membrane excitability of rat DRG neurons to acid stimuli. It significantly decreased the number of action potentials and the amplitude of the depolarization induced by an extracellular pH drop. Finally, peripherally administered l-THP inhibited the nociceptive response to intraplantar injection of acetic acid in rats. These results indicate that l-THP can inhibit the functional activity of ASICs in dissociated primary sensory neurons and relieve acidosis-evoked pain in vivo, which for the first time provides a novel peripheral mechanism underlying the analgesic action of l-THP.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Alcaloides de Berberina/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Gânglios Espinais/citologia , Neurônios/efeitos dos fármacos , Bloqueadores do Canal Iônico Sensível a Ácido/farmacologia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Concentração de Íons de Hidrogênio , Masculino , Potenciais da Membrana/efeitos dos fármacos , Dor/induzido quimicamente , Dor/prevenção & controle , Medição da Dor/efeitos dos fármacos , Técnicas de Patch-Clamp , Prótons/efeitos adversos , Ratos , Ratos Sprague-Dawley
7.
Neurosci Lett ; 567: 35-9, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24680850

RESUMO

Chlorogenic acid (CGA) is one of the most abundant polyphenol compounds in the human diet. Recently, it is demonstrated to have potent antinociceptive effect. However, little is understood about the mechanism underlying CGA analgesia. Here, we have found that CGA can exert an inhibitory effect on the functional activity of native acid-sensing ion channels (ASICs) in rat dorsal root ganglion (DRG) neurons. First, CGA decreased the peak amplitude of proton-gated currents mediated by ASICs in a concentration-dependent manner. Second, CGA shifted the proton concentration-response curve downward, with a decrease of 41.76 ± 8.65% in the maximum current response to protons but with no significant change in the pH0.5 value. Third, CGA altered acidosis-evoked membrane excitability of rat DRG neurons and caused a significant decrease in the amplitude of the depolarization and the number of action potentials induced by acid stimuli. Finally, peripheral administered CGA attenuated nociceptive response to intraplantar injection of acetic acid in rats. ASICs are distributed in peripheral sensory neurons and participate in nociception. Our findings CGA inhibition of native ASICs indicated that CGA may exert analgesic action by modulating ASICs in the primary afferent neurons, which revealed a novel cellular and molecular mechanism underlying CGA analgesia.


Assuntos
Bloqueadores do Canal Iônico Sensível a Ácido/farmacologia , Canais Iônicos Sensíveis a Ácido/fisiologia , Analgésicos/farmacologia , Ácido Clorogênico/farmacologia , Gânglios Espinais/efeitos dos fármacos , Células Receptoras Sensoriais/efeitos dos fármacos , Ácido Acético , Potenciais de Ação , Animais , Membrana Celular/fisiologia , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Ativação do Canal Iônico , Masculino , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Dor/induzido quimicamente , Dor/fisiopatologia , Técnicas de Patch-Clamp , Ratos Sprague-Dawley , Células Receptoras Sensoriais/fisiologia
8.
Br J Pharmacol ; 171(12): 3065-76, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24641084

RESUMO

BACKGROUND AND PURPOSE: A growing number of studies have demonstrated that oxytocin (OT) plays an analgesic role in modulation of nociception and pain. Most work to date has focused on the central mechanisms of OT analgesia, but little is known about whether peripheral mechanisms are also involved. Acid-sensing ion channels (ASICs) are distributed in peripheral sensory neurons and participate in nociception. Here, we investigated the effects of OT on the activity of ASICs in dorsal root ganglion (DRG) neurons. EXPERIMENTAL APPROACH: Electrophysiological experiments were performed on neurons from rat DRG. Nociceptive behaviour was induced by acetic acid in rats and mice lacking vasopressin, V1A receptors. KEY RESULTS: OT inhibited the functional activity of native ASICs. Firstly, OT dose-dependently decreased the amplitude of ASIC currents in DRG neurons. Secondly, OT inhibition of ASIC currents was mimicked by arginine vasopressin (AVP) and completely blocked by the V1A receptor antagonist SR49059, but not by the OT receptor antagonist L-368899. Thirdly, OT altered acidosis-evoked membrane excitability of DRG neurons and significantly decreased the amplitude of the depolarization and number of action potentials induced by acid stimuli. Finally, peripherally administered OT or AVP inhibited nociceptive responses to intraplantar injection of acetic acid in rats. Both OT and AVP also induced an analgesic effect on acidosis-evoked pain in wild-type mice, but not in V1A receptor knockout mice. CONCLUSIONS AND IMPLICATIONS: These results reveal a novel peripheral mechanism for the analgesic effect of OT involving the modulation of native ASICs in primary sensory neurons mediated by V1A receptors.


Assuntos
Bloqueadores do Canal Iônico Sensível a Ácido/farmacologia , Canais Iônicos Sensíveis a Ácido/efeitos dos fármacos , Analgésicos/farmacologia , Gânglios Espinais/efeitos dos fármacos , Ocitocina/farmacologia , Receptores de Vasopressinas/efeitos dos fármacos , Células Receptoras Sensoriais/efeitos dos fármacos , Ácido Acético , Canais Iônicos Sensíveis a Ácido/metabolismo , Potenciais de Ação , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Gânglios Espinais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nociceptividade/efeitos dos fármacos , Dor Nociceptiva/induzido quimicamente , Dor Nociceptiva/prevenção & controle , Dor Nociceptiva/psicologia , Ratos Sprague-Dawley , Receptores de Vasopressinas/genética , Receptores de Vasopressinas/metabolismo , Células Receptoras Sensoriais/metabolismo
9.
Eur J Pharmacol ; 731: 50-7, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24642360

RESUMO

Acid-sensing ion channels (ASICs), a family of proton-gated cation channels, are believed to mediate pain caused by extracellular acidification. Gastrodin is a main bioactive constituent of the traditional herbal Gastrodia elata Blume, which has been widely used in Oriental countries for centuries. As an analgesic, gastrodin has been used clinically to treat pain such as migraine and headache. However, the mechanisms underlying analgesic action of gastrodin are still poorly understood. Here, we have found that gastrodin inhibited the activity of native ASICs in rat dorsal root ganglion (DRG) neurons. Gastrodin dose-dependently inhibited proton-gated currents mediated by ASICs. Gastrodin shifted the proton concentration-response curve downwards, with a decrease of 36.92 ± 6.23% in the maximum current response but with no significant change in the pH0.5 value. Moreover, gastrodin altered acid-evoked membrane excitability of rat DRG neurons and caused a significant decrease in the amplitude of the depolarization and the number of action potentials induced by acid stimuli. Finally, peripheral applied gastrodin relieved pain evoked by intraplantar injection of acetic acid in rats. Our results indicate that gastrodin can inhibit the activity of ASICs in the primary sensory neurons, which provided a novel mechanism underlying analgesic action of gastrodin.


Assuntos
Bloqueadores do Canal Iônico Sensível a Ácido/farmacologia , Canais Iônicos Sensíveis a Ácido/metabolismo , Álcoois Benzílicos/farmacologia , Gânglios Espinais/citologia , Glucosídeos/farmacologia , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/efeitos dos fármacos , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Formaldeído/farmacologia , Masculino , Nociceptividade/efeitos dos fármacos , Prótons , Ratos , Ratos Sprague-Dawley
10.
Brain Res ; 1554: 12-20, 2014 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-24491633

RESUMO

Extracellular acidosis is a common feature in pain-generating pathological conditions. Acid-sensing ion channels (ASICs), pH sensors, are distributed in peripheral sensory neurons and participate in nociception. Morphine exerts potent analgesic effects through the activation of opioid receptors for various pain conditions. A cross-talk between ASICs and opioid receptors in peripheral sensory neurons has not been shown so far. Here, we have found that morphine inhibits the activity of native ASICs in rat dorsal root ganglion (DRG) neurons. Morphine dose-dependently inhibited proton-gated currents mediated by ASICs in the presence of the TRPV1 inhibitor capsazepine. Morphine shifted the proton concentration-response curve downwards, with a decrease of 51.4±3.8% in the maximum current response but with no significant change in the pH0.5 value. Another µ-opioid receptor agonist DAMGO induced a similar decrease in ASIC currents compared with morphine. The morphine inhibition of ASIC currents was blocked by naloxone, a specific opioid receptor antagonist. Pretreatment of forskolin, an adenylyl cyclase activator, or the addition of cAMP reversed the inhibitory effect of morphine. Moreover, morphine altered acid-evoked excitability of rat DRG neurons and decreased the number of action potentials induced by acid stimuli. Finally, peripheral applied morphine relieved pain evoked by intraplantar of acetic acid in rats. Our results indicate that morphine can inhibit the activity of ASICs via µ-opioid receptor and cAMP dependent signal pathway. These observations demonstrate a cross-talk between ASICs and opioid receptors in peripheral sensory neurons, which was a novel analgesic mechanism of morphine.


Assuntos
Bloqueadores do Canal Iônico Sensível a Ácido/farmacologia , Canais Iônicos Sensíveis a Ácido/metabolismo , Gânglios Espinais/efeitos dos fármacos , Morfina/farmacologia , Neurônios/efeitos dos fármacos , Ácido Acético , Bloqueadores do Canal Iônico Sensível a Ácido/administração & dosagem , Potenciais de Ação/efeitos dos fármacos , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacologia , Animais , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Gânglios Espinais/fisiologia , Técnicas In Vitro , Masculino , Morfina/administração & dosagem , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Neurônios/fisiologia , Nociceptividade/efeitos dos fármacos , Dor/tratamento farmacológico , Dor/fisiopatologia , Prótons , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...