Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
J Biol Chem ; 299(3): 102953, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36731795

RESUMEN

Acid-sensing ion channels (ASICs) play an important role in pain associated with tissue acidification. Peripheral inhibitory group II metabotropic glutamate receptors (mGluRs) have analgesic effects in a variety of pain conditions. Whether there is a link between ASICs and mGluRs in pain processes is still unclear. Herein, we show that the group II mGluR agonist LY354740 inhibited acid-evoked ASIC currents and action potentials in rat dorsal root ganglia neurons. LY354740 reduced the maximum current response to protons, but it did not change the sensitivity of ASICs to protons. LY354740 inhibited ASIC currents by activating group II mGluRs. We found that the inhibitory effect of LY354740 was blocked by intracellular application of the Gi/o protein inhibitor pertussis toxin and the cAMP analogue 8-Br-cAMP and mimicked by the protein kinase A (PKA) inhibitor H-89. LY354740 also inhibited ASIC3 currents in CHO cells coexpressing mGluR2 and ASIC3 but not in cells expressing ASIC3 alone. In addition, intraplantar injection of LY354740 dose-dependently alleviated acid-induced nociceptive behavior in rats through local group II mGluRs. Together, these results suggested that activation of peripheral group II mGluRs inhibited the functional activity of ASICs through a mechanism that depended on Gi/o proteins and the intracellular cAMP/PKA signaling pathway in rat dorsal root ganglia neurons. We propose that peripheral group II mGluRs are an important therapeutic target for ASIC-mediated pain.


Asunto(s)
Canales Iónicos Sensibles al Ácido , Ganglios Espinales , Receptores de Glutamato Metabotrópico , Células Receptoras Sensoriales , Animales , Cricetinae , Ratas , Canales Iónicos Sensibles al Ácido/metabolismo , Cricetulus , Ganglios Espinales/metabolismo , Dolor , Protones , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/metabolismo , Células Receptoras Sensoriales/metabolismo , Potenciales de Acción , Células CHO
2.
J Neurochem ; 2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-37987505

RESUMEN

Resolvin D2 (RvD2), an endogenous lipid mediator derived from docosahexaenoic acid, has been demonstrated to have analgesic effects. However, little is known about the mechanism underlying RvD2 in pain relief. Herein, we demonstrate that RvD2 targeted the P2X3 receptor as an analgesic. The electrophysiological activity of P2X3 receptors was suppressed by RvD2 in rat dorsal root ganglia (DRG) neurons. RvD2 pre-application dose-dependently decreased α,ß-methylene-ATP (α,ß-meATP)-induced inward currents. RvD2 remarkably decreased the maximum response to α,ß-meATP, without influencing the affinity of P2X3 receptors. RvD2 also voltage-independently suppressed ATP currents. An antagonist of the G protein receptor 18 (GPR18), O-1918, prevented the RvD2-induced suppression of ATP currents. Additionally, intracellular dialysis of the Gαi/o -protein antagonist pertussis toxin (PTX), the PKA antagonist H89, or the cAMP analog 8-Br-cAMP also blocked the RvD2-induced suppression. Furthermore, α,ß-meATP-triggered depolarization of membrane potential along with the action potential bursts in DRG neurons were inhibited by RvD2. Lastly, RvD2 attenuated spontaneous nociceptive behaviors as well as mechanical allodynia produced by α,ß-meATP in rats via the activation of the peripheral GPR18. These findings indicated that RvD2 inhibited P2X3 receptors in rat primary sensory neurons through GPR18, PTX-sensitive Gαi/o -proteins, and intracellular cAMP/PKA signaling, revealing a novel mechanism that underlies its analgesic effects by targeting P2X3 receptors.

3.
J Neurochem ; 163(4): 327-337, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35986707

RESUMEN

Lysophosphatidic acid (LPA) is a phospholipid which has been implicated in pain. Acid-sensing ion channels (ASICs) are important players in pain associated with tissue acidification. However, it is still unclear whether there is a link between LPA signaling and ASICs in pain processes. Herein, we show that a functional interaction between them in rat dorsal root ganglia (DRG) neurons. Pre-application of LPA enhanced ASIC-mediated and acid-evoked inward currents in a concentration-dependent manner. LPA shifted the concentration-response curve for protons upwards, with an increase of 41.79 ± 4.71% in the maximal current response of ASICs to protons in the presence of LPA. Potentiation of ASIC currents by LPA was blocked by the LPA1 receptor antagonist Ki16198, but not by the LPA2 receptor antagonist H2L5185303. The LPA-induced potentiation was also prevented by intracellular application of either G protein inhibitor or protein kinase C (PKC) inhibitor, but not by Rho inhibitor. LPA also enhanced ASIC3 currents in CHO cells co-expressing ASIC3 and LPA1 receptors, but not in cells expressing ASIC3 alone. Moreover, LPA increased the amplitude of the depolarization and the number of spikes induced by acid stimuli. Finally, LPA exacerbated acid-induced nociceptive behaviors in rats. These results suggested that LPA enhanced ASIC-mediated electrophysiological activity and nociception via a LPA1 receptor and its downstream PKC rather than Rho signaling pathway, which provided a novel peripheral mechanism underlying the sensitization of pain.


Asunto(s)
Ganglios Espinales , Protones , Ratas , Animales , Cricetinae , Cricetulus , Ratas Sprague-Dawley , Canales Iónicos Sensibles al Ácido/metabolismo , Neuronas/metabolismo , Dolor/metabolismo
4.
J Neurosci Res ; 100(9): 1755-1764, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35592934

RESUMEN

Resveratrol can relieve pain under various pain conditions. One of the mechanisms of resveratrol analgesia is the regulation of ion channels. Acid-sensing ion channels (ASICs) are expressed predominantly in nociceptive sensory neurons to detect changes in extracellular pH. ASICs are important players in pain associated with tissue acidification. However, it is still unclear whether ASICs are resveratrol targets. Electrophysiological recordings showed that resveratrol decreased acid-induced and ASIC-mediated currents in male rat dorsal root ganglion (DRG) neurons in a concentration-dependent manner. Resveratrol downwardly shifted the concentration-response curve for protons, suggesting that it inhibited ASICs not by changing the pH0.5 , but by suppressing the proton-induced maximum response. It also suppressed acid-triggered action potentials in the rat DRG neurons. Finally, intraplantar pretreatment with resveratrol relieved acid-induced nociceptive responses in male rats in a dose-dependent manner. These results indicated that resveratrol inhibited ASIC-mediated electrophysiological activity and nociception, suggesting a novel peripheral mechanism underlying its analgesic effect.


Asunto(s)
Canales Iónicos Sensibles al Ácido , Ganglios Espinales , Animales , Ganglios Espinales/fisiología , Masculino , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Protones , Ratas , Ratas Sprague-Dawley , Resveratrol , Células Receptoras Sensoriales
5.
J Neuroinflammation ; 18(1): 92, 2021 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-33853615

RESUMEN

BACKGROUND: Tumor necrosis factor-α (TNF-α) is a pro-inflammatory cytokine involved in pain processing and hypersensitivity. It regulates not only the expression of a variety of inflammatory mediators but also the functional activity of some ion channels. Acid-sensing ion channels (ASICs), as key sensors for extracellular protons, are expressed in nociceptive sensory neurons and contribute to pain signaling caused by tissue acidosis. It is still unclear whether TNF-α has an effect on functional activity of ASICs. Herein, we reported that a brief exposure of TNF-α acutely sensitized ASICs in rat dorsal root ganglion (DRG) neurons. METHODS: Electrophysiological experiments on rat DRG neurons were performed in vitro and acetic acid induced nociceptive behavior quantified in vitro. RESULTS: A brief (5min) application of TNF-α rapidly enhanced ASIC-mediated currents in rat DRG neurons. TNF-α (0.1-10 ng/ml) dose-dependently increased the proton-evoked ASIC currents with an EC50 value of 0.12 ± 0.01 nM. TNF-α shifted the concentration-response curve of proton upwards with a maximal current response increase of 42.34 ± 7.89%. In current-clamp recording, an acute application of TNF-α also significantly increased acid-evoked firing in rat DRG neurons. The rapid enhancement of ASIC-mediated electrophysiological activity by TNF-α was prevented by p38 mitogen-activated protein kinase (MAPK) inhibitor SB202190, but not by non-selective cyclooxygenase inhibitor indomethacin, suggesting that p38 MAPK is necessary for this enhancement. Behaviorally, TNF-α exacerbated acid-induced nociceptive behaviors in rats via activation of local p38 MAPK pathway. CONCLUSIONS: These results suggest that TNF-α rapidly enhanced ASIC-mediated functional activity via a p38 MAPK pathway, which revealed a novel peripheral mechanism underlying TNF-α involvement in rapid hyperalgesia by sensitizing ASICs in primary sensory neurons.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Ganglios Espinales/citología , Neuronas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Ácido Acético/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Masculino , Neuronas/metabolismo , Nociceptores/metabolismo , Nociceptores/fisiología , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
6.
Acta Pharmacol Sin ; 41(8): 1049-1057, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32107467

RESUMEN

Endothelin-1 (ET-1), an endogenous vasoactive peptide, has been found to play an important role in peripheral pain signaling. Acid-sensing ion channels (ASICs) are key sensors for extracellular protons and contribute to pain caused by tissue acidosis. It remains unclear whether an interaction exists between ET-1 and ASICs in primary sensory neurons. In this study, we reported that ET-1 enhanced the activity of ASICs in rat dorsal root ganglia (DRG) neurons. In whole-cell voltage-clamp recording, ASIC currents were evoked by brief local application of pH 6.0 external solution in the presence of TRPV1 channel blocker AMG9810. Pre-application with ET-1 (1-100 nM) dose-dependently increased the proton-evoked ASIC currents with an EC50 value of 7.42 ± 0.21 nM. Pre-application with ET-1 (30 nM) shifted the concentration-response curve of proton upwards with a maximal current response increase of 61.11% ± 4.33%. We showed that ET-1 enhanced ASIC currents through endothelin-A receptor (ETAR), but not endothelin-B receptor (ETBR) in both DRG neurons and CHO cells co-expressing ASIC3 and ETAR. ET-1 enhancement was inhibited by blockade of G-protein or protein kinase C signaling. In current-clamp recording, pre-application with ET-1 (30 nM) significantly increased acid-evoked firing in rat DRG neurons. Finally, we showed that pharmacological blockade of ASICs by amiloride or APETx2 significantly alleviated ET-1-induced flinching and mechanical hyperalgesia in rats. These results suggest that ET-1 sensitizes ASICs in primary sensory neurons via ETAR and PKC signaling pathway, which may contribute to peripheral ET-1-induced nociceptive behavior in rats.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Endotelina-1/farmacología , Células Receptoras Sensoriales/efectos de los fármacos , Agonistas de los Canales de Sodio/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Células CHO , Cricetulus , Ganglios Espinales/citología , Hiperalgesia/inducido químicamente , Masculino , Ratas Sprague-Dawley , Receptor de Endotelina A/metabolismo , Transducción de Señal/efectos de los fármacos
7.
J Neurosci Res ; 97(10): 1298-1305, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31240740

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) is an important member of multifunctional growth factor superfamily. It has been implicated in pain signaling, but little is known about the underlying mechanisms. Herein, we report that TGF-ß1 can exert a sustained enhancing effect on the functional activity of acid-sensing ion channels (ASICs) in rat dorsal root ganglia (DRG) neurons. Pre-application of TGF-ß1 increased the amplitude of proton-gated currents in a dose-dependent manner. Enhancement of ASIC currents lasted for more than 30 min although TGF-ß1 was treated once only. This sustained enhancement by TGF-ß1 could be blocked by extracellular treatment of selective TGF-ß receptor I antagonist SD-208, and abolished by blockade of intracellular several non-Smad-signaling pathways. TGF-ß1 also sustainedly enhanced proton-evoked spikes in rat DRG neurons. Moreover, peripheral pre-treatment with TGF-ß1 dose-dependently exacerbated nociceptive behaviors evoked by intraplantar injection of acetic acid through TGF-ß receptor I in rats. These results suggested that TGF-ß1 potentiated ASIC-mediated electrophysiological activity and nociceptive behaviors, which revealed a novel mechanism underlying TGF-ß1 implicated in peripheral pain signaling by sensitizing ASICs.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Nocicepción/fisiología , Dolor Nociceptivo/fisiopatología , Células Receptoras Sensoriales/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Ganglios Espinales/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
8.
J Neuroinflammation ; 14(1): 150, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28754162

RESUMEN

BACKGROUND: Tissue acidosis and inflammatory mediators play critical roles in pain. Pro-inflammatory agents trypsin and tryptase cleave and activate proteinase-activated receptor 2 (PAR2) expressed on sensory nerves, which is involved in peripheral mechanisms of inflammation and pain. Extracellular acidosis activates acid-sensing ion channel 3 (ASIC3) to trigger pain sensation. Here, we show that a functional interaction of PAR2 and ASIC3 could contribute to acidosis-induced nociception. METHODS: Electrophysiological experiments were performed on both rat DRG neurons and Chinese hamster ovary (CHO) cells expressing ASIC3 and PAR2. Nociceptive behavior was induced by acetic acid in rats. RESULTS: PAR2-AP, PAR2-activating peptide, concentration-dependently increased the ASIC3 currents in CHO cells transfected with ASIC3 and PAR2. The proton concentration-response relationship was not changed, but that the maximal response increased 58.7 ± 3.8% after pretreatment of PAR2-AP. PAR2 mediated the potentiation of ASIC3 currents via an intracellular cascade. PAR2-AP potentiation of ASIC3 currents disappeared after inhibition of intracellular G protein, PLC, PKC, or PKA signaling. Moreover, PAR2 activation increased proton-evoked currents and spikes mediated by ASIC3 in rat dorsal root ganglion neurons. Finally, peripheral administration of PAR2-AP dose-dependently exacerbated acidosis-induced nocifensive behaviors in rats. CONCLUSIONS: These results indicated that PAR2 signaling sensitized ASIC3, which may contribute to acidosis-induced nociception. These represent a novel peripheral mechanism underlying PAR2 involvement in hyperalgesia by sensitizing ASIC3 in primary sensory neurons.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Acidosis/complicaciones , Nocicepción/fisiología , Dolor/inducido químicamente , Receptor PAR-2/metabolismo , Transducción de Señal/fisiología , Canales Iónicos Sensibles al Ácido/genética , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Animales , Células CHO , Células Cultivadas , Cricetulus , Modelos Animales de Enfermedad , Ganglios Espinales/citología , Concentración de Iones de Hidrógeno , Masculino , Neuronas/efectos de los fármacos , Nocicepción/efectos de los fármacos , Oligopéptidos/farmacología , Técnicas de Placa-Clamp , Ratas , Receptor PAR-2/genética , Transducción de Señal/efectos de los fármacos
9.
Pharmacol Res ; 107: 19-26, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26946972

RESUMEN

Glutamate activates peripheral group I metabotropic glutamate receptors (mGluRs) and contributes to inflammatory pain. However, it is still not clear the mechanisms are involved in group I mGluR-mediated peripheral sensitization. Herein, we report that group I mGluRs signaling sensitizes acid-sensing ion channels (ASICs) in dorsal root ganglion (DRG) neurons and contributes to acidosis-evoked pain. DHPG, a selective group I mGluR agonist, can potentiate the functional activity of ASICs, which mediated the proton-induced events. DHPG concentration-dependently increased proton-gated currents in DRG neurons. It shifted the proton concentration-response curve upwards, with a 47.3±7.0% increase of the maximal current response to proton. Group I mGluRs, especially mGluR5, mediated the potentiation of DHPG via an intracellular cascade. DHPG potentiation of proton-gated currents disappeared after inhibition of intracellular Gq/11 proteins, PLCß, PKC or PICK1 signaling. Moreover, DHPG enhanced proton-evoked membrane excitability of rat DRG neurons and increased the amplitude of the depolarization and the number of spikes induced by acid stimuli. Finally, peripherally administration of DHPG dose-dependently exacerbated nociceptive responses to intraplantar injection of acetic acid in rats. Potentiation of ASIC activity by group I mGluR signaling in rat DRG neurons revealed a novel peripheral mechanism underlying group I mGluRs involvement in hyperalgesia.


Asunto(s)
Canales Iónicos Sensibles al Ácido/fisiología , Ganglios Espinales/fisiología , Neuronas/fisiología , Dolor/fisiopatología , Receptores de Glutamato Metabotrópico/fisiología , Ácido Acético , Acidosis/complicaciones , Acidosis/fisiopatología , Animales , Capsaicina/análogos & derivados , Capsaicina/farmacología , Ganglios Espinales/efectos de los fármacos , Masculino , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Neuronas/efectos de los fármacos , Dolor/inducido químicamente , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/agonistas , Bloqueadores de los Canales de Sodio/farmacología , Canales Catiónicos TRPV/antagonistas & inhibidores , Tetrodotoxina/farmacología
10.
Purinergic Signal ; 12(1): 69-78, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26538146

RESUMEN

Peripheral purinergic signaling plays an important role in nociception. Increasing evidence suggests that metabotropic P2Y receptors are also involved, but little is known about the underlying mechanism. Herein, we report that selective P2Y receptor agonist uridine 5'-triphosphate (UTP) can exert an enhancing effect on the functional activity of acid-sensing ion channels (ASICs), key sensors for extracellular protons, in rat dorsal root ganglia (DRG) neurons. First, UTP dose-dependently increased the amplitude of ASIC currents. UTP also shifted the concentration-response curve for proton upwards, with a 56.6 ± 6.4% increase of the maximal current response to proton. Second, UTP potentiation of proton-gated currents can be mimicked by adenosine 5'-triphosphate (ATP), but not by P2Y1 receptor agonist ADP. Potentiation of UTP was blocked by P2Y receptor antagonist suramin and by inhibition of intracellular G protein, phospholipase C (PLC), protein kinase C (PKC), or protein interacting with C-kinase 1 (PICK1) signaling. Third, UTP 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. Finally, UTP dose-dependently exacerbated nociceptive responses to injection of acetic acid in rats. These results suggest that UTP enhanced ASIC-mediated currents and nociceptive responses, which reveal a novel peripheral mechanism underlying UTP-sensitive P2Y2 receptor involvement in hyperalgesia by sensitizing ASICs in primary sensory neurons.


Asunto(s)
Canales Iónicos Sensibles al Ácido/efectos de los fármacos , Agonistas del Receptor Purinérgico P2Y/farmacología , Células Receptoras Sensoriales/efectos de los fármacos , Uridina Trifosfato/farmacología , Canales Iónicos Sensibles al Ácido/metabolismo , Acidosis/fisiopatología , Adenosina Difosfato/farmacología , Adenosina Trifosfato/farmacología , Animales , Relación Dosis-Respuesta a Droga , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Dolor/psicología , Dimensión del Dolor/efectos de los fármacos , Protones , Antagonistas del Receptor Purinérgico P2Y/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2Y1/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Suramina/farmacología , Uridina Trifosfato/antagonistas & inhibidores
11.
J Neurosci Res ; 93(2): 333-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25395088

RESUMEN

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.


Asunto(s)
Canales Iónicos Sensibles al Ácido/metabolismo , Alcaloides de Berberina/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Ganglios Espinales/citología , Neuronas/efectos de los fármacos , Bloqueadores del Canal Iónico Sensible al Ácido/farmacología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Esquema de Medicación , Concentración de Iones de Hidrógeno , Masculino , Potenciales de la Membrana/efectos de los fármacos , Dolor/inducido químicamente , Dolor/prevención & control , Dimensión del Dolor/efectos de los fármacos , Técnicas de Placa-Clamp , Protones/efectos adversos , Ratas , Ratas Sprague-Dawley
12.
J Circadian Rhythms ; 13: 2, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27103928

RESUMEN

The mammalian circadian clock is composed of single-cell oscillators. Neurochemical and electrical signaling among these oscillators is important for the normal expression of circadian rhythms. Prokineticin 2 (PK2), encoding a cysteine-rich secreted protein, has been shown to be a critical signaling molecule for the regulation of circadian rhythms. PK2 expression in the suprachiasmatic nucleus (SCN) is highly rhythmic, peaking during the day and being essentially absent during the night. Mice with disrupted PK2 gene or its receptor PKR2 display greatly reduced rhythmicity of broad circadian parameters such as locomotor activity, body temperature and sleep/wake patterns. PK2 has been shown to increase the firing rate of SCN neurons, with unknown molecular mechanisms. Here we report that TRPV2, an ion channel belonging to the family of TRP, is co-expressed with PKR2 in the SCN neurons. Further, TRPV2 protein, but not TRPV2 mRNA, was shown to oscillate in the SCN in a PK2-dependent manner. Functional studies revealed that TRPV2 enhanced signaling of PKR2 in calcium mobilization or ion current conductance, likely via the increased trafficking of TRPV2 to the cell surface. Taken together, these results indicate that TRPV2 is likely part of the downstream signaling of PK2 in the regulation of the circadian rhythms.

13.
Sheng Li Xue Bao ; 66(6): 647-57, 2014 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-25516513

RESUMEN

A growing number of studies have shown that arginine vasopressin (AVP) plays an analgesia role in the modulation of nociception. Previous studies have focused on the central mechanisms of AVP analgesia. The aim of the present study was to find out whether peripheral mechanisms are also involved. The effect of AVP on GABA-activated currents (IGABA) and GABAA receptor function in freshly isolated dorsal root ganglion (DRG) neurons of rats were studied using whole cell patch clamp technique. The result showed that, IGABA were potentiated by pre-treatment with AVP (1 × 10⁻¹°-1 × 10⁻5 mol/L) in a concentration-dependent manner. Meanwhile, the GABA concentration-response curve was shifted upwards, with an increase of (49.1 ± 4.0)% in the maximal current response but with no significant change in the EC50 values. These results indicate that the enhancing effect is non-competitive. In addition, the effects of AVP on IGABA might be voltage-independent. This potentiation of IGABA induced by AVP was almost completely blocked by the V1a receptor antagonist SR49059 (3 × 10⁻6 mol/L). Also it could be removed by intracellular dialysis of either GDP-ß-S (5 × 10⁻4mol/L), a non-hydrolyzable GDP analog, or GF109203X (2 × 10⁻6 mol/L), a selective protein kinase C (PKC) inhibitor, with the re-patch clamp. These results suggest that AVP up-regulates the function of the GABAA receptor via G protein-coupled receptors and PKC-dependent signal pathways in rat DRG neurons, and this potentiation may underlie the analgesia induced by AVP.


Asunto(s)
Arginina Vasopresina/farmacología , Ganglios Espinales/citología , Neuronas/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacología , Animales , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Indoles , Maleimidas , Potenciales de la Membrana , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/metabolismo , Transducción de Señal , Tionucleótidos/farmacología
14.
Front Synaptic Neurosci ; 15: 1191383, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37216004

RESUMEN

Introduction: Cancer patients treated with paclitaxel often develop chemotherapy-induced peripheral neuropathy, which has not been effectively treated with drugs. The anti-diabetic drug metformin is effective in the treatment of neuropathic pain. The aim of this study was to elucidate effect of metformin on paclitaxel-induced neuropathic pain and spinal synaptic transmission. Methods: Electrophysiological experiments on rat spinal slices were performed in vitro and mechanical allodynia quantified in vitro. Results: The present data demonstrated that intraperitoneal injection of paclitaxel produced mechanical allodynia and potentiated spinal synaptic transmission. Intrathecal injection of metformin significantly reversed the established mechanical allodynia induced by paclitaxel in rats. Either spinal or systemic administration of metformin significantly inhibited the increased frequency of spontaneous excitatory postsynaptic currents (sEPSCs) in spinal dorsal horn neurons from paclitaxel-treated rats. We found that 1 h incubation of metformin also reduced the frequency rather than the amplitude of sEPSCs in the spinal slices from paclitaxel-treated rats. Discussion: These results suggested that metformin was able to depress the potentiated spinal synaptic transmission, which may contribute to alleviating the paclitaxel-induced neuropathic pain.

15.
Neuropharmacology ; 241: 109739, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37820935

RESUMEN

Cholecystokinin (CCK) is a peptide that has been implicated in pain modulation. Acid sensitive ion channels (ASICs) also play an important role in pain associated with tissue acidification. However, it is still unclear whether there is an interaction between CCK signaling and ASICs during pain process. Herein, we report that a functional link between them in rat dorsal root ganglion (DRG) neurons. Pretreatment with CCK-8 concentration-dependently increased acid-evoked ASIC currents. CCK-8 increased the maximum response of ASICs to acid, but did not changed their acid sensitivity. Enhancement of ASIC currents by CCK-8 was mediated by the stimulation of CCK2 receptor (CCK2R), rather than CCK1R. The enhancement of ASIC currents by CCK-8 was prevented by application of either G-protein inhibitor GDP-ß-S or protein kinase C (PKC) inhibitor GF109203×, but not by protein kinase A (PKA) inhibitor H-89 or JNK inhibitor SP600125. Moreover, CCK-8 increased the number of action potentials triggered by acid stimuli by activating CCK2R. Finally, CCK-8 dose-dependently exacerbated acid-induced nociceptive behavior in rats through local CCK2R. Together, these results indicated that CCK-8/CCK2R activation enhanced ASIC-mediated electrophysiological activity in DRG neurons and nociception in rats. The enhancement effect depended on G-proteins and intracellular PKC signaling rather than PKA and JNK signaling pathway. These findings provided that CCK-8/CCK2R is an important therapeutic target for ASIC-mediated pain.


Asunto(s)
Canales Iónicos Sensibles al Ácido , Sincalida , Ratas , Animales , Ratas Sprague-Dawley , Sincalida/farmacología , Sincalida/metabolismo , Canales Iónicos Sensibles al Ácido/metabolismo , Células Receptoras Sensoriales , Dolor/metabolismo , Ganglios Espinales/metabolismo
16.
Neuropharmacology ; 227: 109443, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36709909

RESUMEN

P2X3 receptors and group II metabotropic glutamate receptors (mGluRs) have been found to be expressed in primary sensory neurons. P2X3 receptors participate in a variety of pain processes, while the activation of mGluRs has an analgesic effect. However, it's still unclear whether there is a link between them in pain. Herein, we reported that the group II mGluR activation inhibited the electrophysiological activity of P2X3 receptors in rat dorsal root ganglia (DRG) neurons. Group II mGluR agonist LY354740 concentration-dependently decreased P2X3 receptor-mediated and α,ß-methylene-ATP (α,ß-meATP)-evoked inward currents in DRG neurons. LY354740 significantly suppressed the maximum response of P2X3 receptor to α,ß-meATP, but did not change their affinity. Inhibition of ATP currents by LY354740 was blocked by the group II mGluR antagonist LY341495, also prevented by the intracellular dialysis of either the Gi/o protein inhibitor pertussis toxin, the cAMP analog 8-Br-cAMP, or the protein kinase A (PKA) inhibitor H-89. Moreover, LY354740 decreased α,ß-meATP-induced membrane potential depolarization and action potential bursts in DRG neurons. Finally, intraplantar injection of LY354740 also relieved α,ß-meATP-induced spontaneous nociceptive behaviors and mechanical allodynia in rats by activating peripheral group Ⅱ mGluRs. These results indicated that peripheral group II mGluR activation inhibited the functional activity of P2X3 receptors via a Gi/o protein and cAMP/PKA signaling pathway in rat DRG neurons, which revealed a novel mechanism underlying analgesic effects of peripheral group II mGluRs. This article is part of the Special Issue on "Purinergic Signaling: 50 years".


Asunto(s)
Receptores de Glutamato Metabotrópico , Ratas , Animales , Receptores de Glutamato Metabotrópico/metabolismo , Ganglios Espinales/metabolismo , Receptores Purinérgicos P2X3/metabolismo , Dolor/metabolismo , Neuronas , Adenosina Trifosfato/metabolismo , Analgésicos/farmacología
17.
J Neuroinflammation ; 9: 108, 2012 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-22642848

RESUMEN

BACKGROUND: Prokineticin 2 (PK2) is a secreted protein and causes potent hyperalgesia in vivo, and is therefore considered to be a new pronociceptive mediator. However, the molecular targets responsible for the pronociceptive effects of PK2 are still poorly understood. Here, we have found that PK2 potentiates the activity of acid-sensing ion channels in the primary sensory neurons. METHODS: In the present study, experiments were performed on neurons freshly isolated from rat dorsal root ganglion by using whole-cell patch clamp and voltage-clamp recording techniques. RESULTS: PK2 dose-dependently enhanced proton-gated currents with an EC50 of 0.22 ± 0.06 nM. PK2 shifted the proton concentration-response curve upwards, with a 1.81 ± 0.11 fold increase of the maximal current response. PK2 enhancing effect on proton-gated currents was completely blocked by PK2 receptor antagonist. The potentiation was also abolished by intracellular dialysis of GF109203X, a protein kinase C inhibitor, or FSC-231, a protein interacting with C-kinase 1 inhibitor. Moreover, PK2 enhanced the acid-evoked membrane excitability of rat dorsal root ganglion neurons and caused a significant increase in the amplitude of the depolarization and the number of spikes induced by acid stimuli. Finally, PK2 exacerbated nociceptive responses to the injection of acetic acid in rats. CONCLUSION: These results suggest that PK2 increases the activity of acid-sensing ion channels via the PK2 receptor and protein kinase C-dependent signal pathways in rat primary sensory neurons. Our findings support that PK2 is a proalgesic factor and its signaling likely contributes to acidosis-evoked pain by sensitizing acid-sensing ion channels.


Asunto(s)
Canales Iónicos Sensibles al Ácido/fisiología , Ganglios Espinales/metabolismo , Hormonas Gastrointestinales/fisiología , Neuronas/química , Neuropéptidos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Receptores de Péptidos/fisiología , Canales Iónicos Sensibles al Ácido/metabolismo , Animales , Sinergismo Farmacológico , Ganglios Espinales/enzimología , Ganglios Espinales/fisiología , Hormonas Gastrointestinales/química , Masculino , Neuronas/enzimología , Neuronas/metabolismo , Neuropéptidos/química , Proteína Quinasa C/química , Proteína Quinasa C/fisiología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química , Receptores de Péptidos/agonistas , Receptores de Péptidos/química , Transducción de Señal/fisiología
18.
Front Pharmacol ; 13: 928647, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35795546

RESUMEN

Lysophosphatidic acid (LPA), a lipid metabolite, plays a role in both neuropathic and inflammatory pain through LPA1 receptors. P2X3 receptor has also been shown to participate in these pathological processes. However, it is still unclear whether there is a link between LPA signaling and P2X3 receptors in pain. Herein, we show that a functional interaction between them in rat dorsal root ganglia (DRG) neurons. Pretreatment of LPA concentration-dependently enhanced α,ß-methylene-ATP (α,ß-meATP)-induced inward currents mediated by P2X3 receptors. LPA significantly increased the maximal current response of α,ß-meATP, showing an upward shift of the concentration-response curve for α,ß-meATP. The LPA enhancement was independent on the clamping-voltage. Enhancement of P2X3 receptor-mediated currents by LPA was prevented by the LPA1 receptor antagonist Ki16198, but not by the LPA2 receptor antagonist H2L5185303. The LPA-induced potentiation was also attenuated by intracellular dialysis of either G-protein inhibitor or protein kinase C (PKC) inhibitor, but not by Rho inhibitor. Moreover, LPA significantly changed the membrane potential depolarization and action potential burst induced by α,ß-meATP in DRG neurons. Finally, LPA exacerbated α,ß-meATP- induced nociceptive behaviors in rats. These results suggested that LPA potentiated the functional activity of P2X3 receptors in rat primary sensory neurons through activation of the LPA1 receptor and its downstream PKC rather than Rho signaling pathway, indicating a novel peripheral mechanism underlying the sensitization of pain.

19.
CNS Neurosci Ther ; 28(2): 289-297, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34862748

RESUMEN

AIMS: The α2 -adrenergic receptor (α2 -AR) agonists have been shown to be effective in the treatment of various pain. For example, dexmedetomidine (DEX), a selective α2A -AR agonist, can be used for peripheral analgesia. However, it is not yet fully elucidated for the precise molecular mechanisms. P2X3 receptor is a major receptor processing nociceptive information in primary sensory neurons. Herein, we show that a functional interaction of α2A -ARs and P2X3 receptors in dorsal root ganglia (DRG) neurons could contribute to peripheral analgesia of DEX. METHODS: Electrophysiological recordings were carried out on rat DRG neurons, and nociceptive behavior was quantified in rats. RESULTS: The activation of α2A -ARs by DEX suppressed P2X3 receptor-mediated and α,ß-methylene-ATP (α,ß-meATP)-evoked inward currents in a concentration-dependent and voltage-independent manner. Pre-application of DEX shifted the α,ß-meATP concentration-response curve downwards, with a decrease of 50.43 ± 4.75% in the maximal current response of P2X3 receptors to α,ß-meATP in the presence of DEX. Suppression of α,ß-meATP-evoked currents by DEX was blocked by the α2A -AR antagonist BRL44408 and prevented by intracellular application of the Gi/o protein inhibitor pertussis toxin, the adenylate cyclase activator forskolin, and the cAMP analog 8-Br-cAMP. DEX also suppressed α,ß-meATP-evoked action potentials through α2A -ARs in rat DRG neurons. Finally, the activation of peripheral α2A -ARs by DEX had an analgesic effect on the α,ß-meATP-induced nociception. CONCLUSIONS: These results suggested that activation of α2A -ARs by DEX suppressed P2X3 receptor-mediated electrophysiological and behavioral activity via a Gi/o proteins and cAMP signaling pathway, which was a novel potential mechanism underlying analgesia of peripheral α2A -AR agonists.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Fenómenos Electrofisiológicos/efectos de los fármacos , Ganglios Espinales/efectos de los fármacos , Nocicepción/efectos de los fármacos , Receptores Adrenérgicos alfa 2/efectos de los fármacos , Receptores Purinérgicos P2X3/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Dexmedetomidina/farmacología , Masculino , Ratas , Ratas Sprague-Dawley
20.
Neuropharmacology ; 205: 108924, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34919904

RESUMEN

Peripheral A1 adenosine receptor signaling has been shown to have analgesic effects in a variety of pain conditions. However, it is not yet fully elucidated for the precise molecular mechanisms. Acid sensing ion channels (ASICs) are expressed predominantly in nociceptive sensory neurons responding to protons. Given that both A1 adenosine receptors and ASICs are present in dorsal root ganglia (DRG) neurons, we therefore investigated whether there was a cross-talk between the two types of receptors. Herein, electrophysiological recordings showed that the A1 adenosine receptor agonist N6-cyclopentyladenosine (CPA) suppressed acid-induced currents and action potentials, which were mediated by ASICs, in rat DRG neurons. CPA inhibited the maximum response to protons, as shown a downward shift of concentration-response curve for protons. The CPA-induced suppression of ASIC currents was blocked by the A1 adenosine receptor antagonist KW-3902 and also prevented by intracellular application of the Gi/o-protein inhibitor pertussis toxin, the adenylate cyclase activator forskolin, and the cAMP analog 8-Br-cAMP. Finally, intraplantar pretreatment of CPA dose-dependently relieved acid-induced nociceptive responses in rats through peripheral A1 adenosine receptors. These results suggested that CPA suppressed ASICs via A1 adenosine receptors and intracellular Gi/o-proteins and cAMP signaling cascades in rat DRG neurons, which was a novel potential mechanism underlying analgesia of peripheral A1 adenosine receptors.


Asunto(s)
Canales Iónicos Sensibles al Ácido/efectos de los fármacos , Agonistas del Receptor de Adenosina A1/farmacología , Antagonistas del Receptor de Adenosina A1/farmacología , Analgesia , Fenómenos Electrofisiológicos/efectos de los fármacos , Ganglios Espinales/efectos de los fármacos , Nocicepción/efectos de los fármacos , Nociceptores/efectos de los fármacos , Receptor de Adenosina A1/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA