Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pain ; 157(3): 552-559, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26492527

RESUMO

Mambalgins are 57-amino acid peptides isolated from snake venom that evoke naloxone-resistant analgesia after local (intraplantar) and central (intrathecal) injections through inhibition of particular subtypes of acid-sensing ion channels (ASICs). We now show that mambalgins also have an opioid-independent effect on both thermal and mechanical inflammatory pain after systemic intravenous (i.v.) administration and are effective against neuropathic pain. By combining the use of knockdown and knockout animals, we show the critical involvement of peripheral ASIC1b-containing channels, along with a contribution of ASIC1a-containing channels, in the i.v. effects of these peptides against inflammatory pain. The potent analgesic effect on neuropathic pain involves 2 different mechanisms depending on the route of administration, a naloxone-insensitive and ASIC1a-independent effect associated with i.v. injection and an ASIC1a-dependent and partially naloxone-sensitive effect associated with intrathecal injection. These data further support the role of peripheral and central ASIC1-containing channels in pain, demonstrate their participation in neuropathic pain, and highlight differences in the repertoire of channels involved in different pain conditions. They also strengthen the therapeutic potential of mambalgin peptides that are active in a broader range of experimental pain models and through i.v. systemic delivery.


Assuntos
Bloqueadores do Canal Iônico Sensível a Ácido/uso terapêutico , Analgésicos/uso terapêutico , Venenos Elapídicos/uso terapêutico , Neuralgia/tratamento farmacológico , Peptídeos/uso terapêutico , Animais , Feminino , Inflamação/tratamento farmacológico , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuralgia/patologia , Ratos , Resultado do Tratamento , Xenopus laevis
2.
Pain ; 155(12): 2534-2544, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25239074

RESUMO

Two-pore domain background K(+) channels (K2p or KCNK) produce hyperpolarizing currents that control cell membrane polarity and neuronal excitability throughout the nervous system. The TREK2 channel as well as the related TREK1 and TRAAK channels are mechanical-, thermal- and lipid-gated channels that share many regulatory properties. TREK2 is one of the major background channels expressed in rodent nociceptive neurons of the dorsal root ganglia that innervate the skin and deep body tissues, but its role in somatosensory perception and nociception has remained poorly understood. We now report that TREK2 is a regulatory channel that controls the perception of non aversive warm, between 40°C and 46°C, and moderate ambient cool temperatures, between 20°C and 25°C, in mice. TREK2 controls the firing activity of peripheral sensory C-fibers in response to changes in temperature. The role of TREK2 in thermosensation is different from that of TREK1 and TRAAK channels; rather, TREK2, TREK1, and TRAAK channels appear to have complementary roles in thermosensation. TREK2 is also involved in mechanical pain perception and in osmotic pain after sensitization by prostaglandin E2. TREK2 is involved in the cold allodynia that characterizes the neuropathy commonly associated with treatments with the anticancer drug oxaliplatin. These results suggest that positive modulation of the TREK2 channel may have beneficial analgesic effects in these neuropathic conditions.


Assuntos
Regulação da Expressão Gênica/genética , Percepção da Dor/fisiologia , Limiar da Dor/psicologia , Doenças do Sistema Nervoso Periférico/fisiopatologia , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Sensação Térmica/genética , Animais , Antineoplásicos/toxicidade , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperalgesia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fibras Nervosas Amielínicas/fisiologia , Compostos Organoplatínicos/toxicidade , Oxaliplatina , Medição da Dor , Percepção da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Limiar da Dor/fisiologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Estimulação Física , Canais de Potássio/genética , Canais de Potássio de Domínios Poros em Tandem/genética , Sensação Térmica/efeitos dos fármacos
3.
Neuron ; 84(1): 123-136, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25242222

RESUMO

One feature of neuropathic pain is a reduced GABAergic inhibitory function. Nociceptors have been suggested to play a key role in this process. However, the mechanisms behind nociceptor-mediated modulation of GABA signaling remain to be elucidated. Here we describe the identification of GINIP, a Gαi-interacting protein expressed in two distinct subsets of nonpeptidergic nociceptors. GINIP null mice develop a selective and prolonged mechanical hypersensitivity in models of inflammation and neuropathy. GINIP null mice show impaired responsiveness to GABAB, but not to delta or mu opioid receptor agonist-mediated analgesia specifically in the spared nerve injury (SNI) model. Consistently, GINIP-deficient dorsal root ganglia neurons had lower baclofen-evoked inhibition of high-voltage-activated calcium channels and a defective presynaptic inhibition of lamina IIi interneurons. These results further support the role of unmyelinated C fibers in injury-induced modulation of spinal GABAergic inhibition and identify GINIP as a key modulator of peripherally evoked GABAB-receptors signaling.


Assuntos
Analgesia/métodos , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Receptores de GABA-B/fisiologia , Sequência de Aminoácidos , Animais , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/fisiologia , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Ratos
4.
Int Urogynecol J ; 25(8): 1081-7, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24599179

RESUMO

INTRODUCTION AND HYPOTHESIS: Polypropylene (PP) mesh shrinkage represents a serious complication, as a significant cause of pain and recurrence of pelvic organ prolapse or ventral hernias, frequently requiring several surgical interventions. The retraction seems to be caused by the host, in response to the implantation, through the occurrence of periprosthetic adhesions and fibrosis. We hypothesized that avoiding the postoperative adhesions can prevent PP mesh shrinkage. METHODS: Sixty rats were randomly assigned to three groups. A standardized hernia defect was induced on the abdominal wall, which was repaired using an extraperitoneal PP mesh alone (group 1), with application of a hyaluronate carboxymethylcellulose-based bioresorbable membrane (Seprafilm, group 2), or an auto-cross-linked polysaccharide hyaluronan-based solution (Hyalobarrier gel, group 3). Eight weeks after the procedure, a repeat laparotomy was performed. After scoring the adhesion and measuring the mesh surface, a microscopic study of the prosthesis-host tissue interfaces was performed. RESULTS: Group 1 displayed a median shrinkage of 29% of the mesh. The Seprafilm group (p = 0.0238) and Hyalobarrier gel group (p = 0.0072) displayed a significantly smaller reduction of 19.12 and 17 %, respectively. Control group 1 displayed a significantly greater adhesion score (30.40) than the Seprafilm (11.67, p = 0.0028) and Hyalobarrier gel groups (11.19, p = 0.0013). The fibrosis was reduced in the Hyalobarrier gel group only. CONCLUSION: This experimental study revealed that Hyalobarrier gel and Seprafilm can prevent PP mesh shrinkage and postoperative adhesions. They might be integrated in a mesh size-saving strategy, which should preserve the quality and durability of the surgical repair and limit the postoperative pain.


Assuntos
Ácido Hialurônico/uso terapêutico , Polipropilenos , Complicações Pós-Operatórias/prevenção & controle , Telas Cirúrgicas , Aderências Teciduais/prevenção & controle , Animais , Materiais Biocompatíveis/uso terapêutico , Feminino , Fibrose , Géis , Hérnia Abdominal/cirurgia , Complicações Pós-Operatórias/patologia , Estudos Prospectivos , Falha de Prótese/etiologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Aderências Teciduais/complicações , Aderências Teciduais/patologia
5.
Pain ; 155(4): 764-772, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24447516

RESUMO

To exert its analgesic action, paracetamol requires complex metabolism to produce a brain-specific lipoamino acid compound, AM404, which targets central transient receptor potential vanilloid receptors (TRPV1). Lipoamino acids are also known to induce analgesia through T-type calcium-channel inhibition (Ca(v)3.2). In this study we show that the antinociceptive effect of paracetamol in mice is lost when supraspinal Ca(v)3.2 channels are inhibited. Therefore, we hypothesized a relationship between supraspinal Ca(v)3.2 and TRPV1, via AM404, which mediates the analgesic effect of paracetamol. AM404 is able to activate TRPV1 and weakly inhibits Ca(v)3.2. Interestingly, activation of TRPV1 induces a strong inhibition of Ca(v)3.2 current. Supporting this, intracerebroventricular administration of AM404 or capsaicin produces antinociception that is lost in Ca(v)3.2(-/-) mice. Our study, for the first time, (1) provides a molecular mechanism for the supraspinal antinociceptive effect of paracetamol; (2) identifies the relationship between TRPV1 and the Ca(v)3.2 channel; and (3) suggests supraspinal Ca(v)3.2 inhibition as a potential pharmacological strategy to alleviate pain.


Assuntos
Acetaminofen/farmacologia , Analgésicos não Narcóticos/farmacologia , Canais de Cálcio Tipo T/deficiência , Medula Espinal/efeitos dos fármacos , 4-Aminopiridina/farmacologia , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Animais , Artrite/induzido quimicamente , Artrite/tratamento farmacológico , Canais de Cálcio Tipo T/genética , Células Cultivadas , Vias de Administração de Medicamentos , Interações Medicamentosas , Gânglios Espinais/citologia , Humanos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Knockout , Morfina/farmacologia , Neurônios/efeitos dos fármacos , Medição da Dor/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Medula Espinal/citologia , Canais de Cátion TRPV/deficiência , Canais de Cátion TRPV/genética , Tetraetilamônio/farmacologia
6.
Nat Commun ; 4: 2941, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24346231

RESUMO

Morphine is the gold-standard pain reliever for severe acute or chronic pain but it also produces adverse side effects that can alter the quality of life of patients and, in some rare cases, jeopardize the vital prognosis. Morphine elicits both therapeutic and adverse effects primarily through the same µ opioid receptor subtype, which makes it difficult to separate the two types of effects. Here we show that beneficial and deleterious effects of morphine are mediated through different signalling pathways downstream from µ opioid receptor. We demonstrate that the TREK-1 K(+) channel is a crucial contributor of morphine-induced analgesia in mice, while it is not involved in morphine-induced constipation, respiratory depression and dependence-three main adverse effects of opioid analgesic therapy. These observations suggest that direct activation of the TREK-1 K(+) channel, acting downstream from the µ opioid receptor, might have strong analgesic effects without opioid-like adverse effects.


Assuntos
Analgesia/métodos , Morfina/efeitos adversos , Morfina/uso terapêutico , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/uso terapêutico , Animais , Células COS , Chlorocebus aethiops , Constipação Intestinal , Cruzamentos Genéticos , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Naloxona/química , Dor/tratamento farmacológico , Manejo da Dor , Receptores Opioides mu/metabolismo , Insuficiência Respiratória , Transdução de Sinais , Fatores de Tempo
7.
Cell Rep ; 5(2): 378-88, 2013 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-24139797

RESUMO

C-low-threshold mechanoreceptors (C-LTMRs) are unique among C-unmyelinated primary sensory neurons. These neurons convey two opposite aspects of touch sensation: a sensation of pleasantness, and a sensation of injury-induced mechanical pain. Here, we show that TAFA4 is a specific marker of C-LTMRs. Genetic labeling in combination with electrophysiological recordings show that TAFA4+ neurons have intrinsic properties of mechano-nociceptors. TAFA4-null mice exhibit enhanced mechanical and chemical hypersensitivity following inflammation and nerve injury as well as increased excitability of spinal cord lamina IIi neurons, which could be reversed by intrathecal or bath application of recombinant TAFA4 protein. In wild-type C57/Bl6 mice, intrathecal administration of TAFA4 strongly reversed carrageenan-induced mechanical hypersensitivity, suggesting a potent analgesic role of TAFA4 in pain relief. Our data provide insights into how C-LTMR-derived TAFA4 modulates neuronal excitability and controls the threshold of somatic sensation.


Assuntos
Citocinas/metabolismo , Nociceptores/metabolismo , Dor/fisiopatologia , Estresse Mecânico , Animais , Carragenina/toxicidade , Citocinas/genética , Citocinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/fisiologia , Dor/metabolismo , Limiar da Dor/efeitos dos fármacos , Técnicas de Patch-Clamp , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia
8.
Pain ; 154(2): 283-293, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23257507

RESUMO

T-type calcium channels encoded by the Ca(V)3.2 isoform are expressed in nociceptive primary afferent neurons where they contribute to hyperalgesia and thus are considered as a potential therapeutic target to treat pathological pain. Here we report that the small organic state-dependent T-type channel antagonist TTA-A2 efficiently inhibits recombinant and native Ca(V)3.2 currents. Although TTA-A2 is a pan Ca(V)3 blocker, it demonstrates a higher potency for Ca(V)3.2 compared to Ca(V)3.1. TTA-A2 selectivity for T-type currents was demonstrated in sensory neurons where it lowered cell excitability uniquely on neurons expressing T-type channels. In vivo pharmacology in Ca(V)3.2 knockout and wild type mice reveal that TTA-A2-mediated antinociception critically depends on Ca(V)3.2 expression. The pathophysiology of irritable bowel syndrome (IBS) was recently demonstrated to involve Ca(V)3.2 in a rat model of this disease. Oral administration of TTA-A2 produced a dose-dependent reduction of hypersensitivity in an IBS model, demonstrating its therapeutic potential for the treatment of pathological pain. Overall, our results suggest that the high potency of TTA-A2 in the depolarized state strengthen its analgesic efficacy and selectivity toward pathological pain syndromes. This characteristic would be beneficial for the development of analgesics targeting T-type channels, in particular for the treatment of pain associated with IBS.


Assuntos
Benzenoacetamidas/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/metabolismo , Hiperalgesia/tratamento farmacológico , Neurônios/efeitos dos fármacos , Piridinas/farmacologia , Animais , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio Tipo T/genética , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Hiperalgesia/genética , Hiperalgesia/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley
9.
Nature ; 490(7421): 552-5, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23034652

RESUMO

Polypeptide toxins have played a central part in understanding physiological and physiopathological functions of ion channels. In the field of pain, they led to important advances in basic research and even to clinical applications. Acid-sensing ion channels (ASICs) are generally considered principal players in the pain pathway, including in humans. A snake toxin activating peripheral ASICs in nociceptive neurons has been recently shown to evoke pain. Here we show that a new class of three-finger peptides from another snake, the black mamba, is able to abolish pain through inhibition of ASICs expressed either in central or peripheral neurons. These peptides, which we call mambalgins, are not toxic in mice but show a potent analgesic effect upon central and peripheral injection that can be as strong as morphine. This effect is, however, resistant to naloxone, and mambalgins cause much less tolerance than morphine and no respiratory distress. Pharmacological inhibition by mambalgins combined with the use of knockdown and knockout animals indicates that blockade of heteromeric channels made of ASIC1a and ASIC2a subunits in central neurons and of ASIC1b-containing channels in nociceptors is involved in the analgesic effect of mambalgins. These findings identify new potential therapeutic targets for pain and introduce natural peptides that block them to produce a potent analgesia.


Assuntos
Bloqueadores do Canal Iônico Sensível a Ácido/farmacologia , Canais Iônicos Sensíveis a Ácido/metabolismo , Analgésicos/farmacologia , Venenos Elapídicos/farmacologia , Dor/tratamento farmacológico , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Bloqueadores do Canal Iônico Sensível a Ácido/química , Bloqueadores do Canal Iônico Sensível a Ácido/uso terapêutico , Canais Iônicos Sensíveis a Ácido/classificação , Canais Iônicos Sensíveis a Ácido/genética , Analgésicos/efeitos adversos , Analgésicos/química , Analgésicos/uso terapêutico , Animais , Tolerância a Medicamentos , Venenos Elapídicos/administração & dosagem , Venenos Elapídicos/química , Venenos Elapídicos/uso terapêutico , Injeções Espinhais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Morfina/efeitos adversos , Morfina/farmacologia , Naloxona/farmacologia , Nociceptores/química , Nociceptores/metabolismo , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Dor/metabolismo , Peptídeos/administração & dosagem , Peptídeos/química , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/metabolismo , Ratos , Insuficiência Respiratória/induzido quimicamente , Xenopus laevis
10.
Proc Natl Acad Sci U S A ; 108(27): 11268-73, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21690417

RESUMO

The symptoms of irritable bowel syndrome (IBS) include significant abdominal pain and bloating. Current treatments are empirical and often poorly efficacious, and there is a need for the development of new and efficient analgesics aimed at IBS patients. T-type calcium channels have previously been validated as a potential target to treat certain neuropathic pain pathologies. Here we report that T-type calcium channels encoded by the Ca(V)3.2 isoform are expressed in colonic nociceptive primary afferent neurons and that they contribute to the exaggerated pain perception in a butyrate-mediated rodent model of IBS. Both the selective genetic inhibition of Ca(V)3.2 channels and pharmacological blockade with calcium channel antagonists attenuates IBS-like painful symptoms. Mechanistically, butyrate acts to promote the increased insertion of Ca(V)3.2 channels into primary sensory neuron membranes, likely via a posttranslational effect. The butyrate-mediated regulation can be recapitulated with recombinant Ca(V)3.2 channels expressed in HEK cells and may provide a convenient in vitro screening system for the identification of T-type channel blockers relevant to visceral pain. These results implicate T-type calcium channels in the pathophysiology of chronic visceral pain and suggest Ca(V)3.2 as a promising target for the development of efficient analgesics for the visceral discomfort and pain associated with IBS.


Assuntos
Canais de Cálcio Tipo T/fisiologia , Colo/inervação , Colo/fisiopatologia , Síndrome do Intestino Irritável/fisiopatologia , Animais , Sequência de Bases , Butiratos/toxicidade , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/deficiência , Canais de Cálcio Tipo T/genética , Modelos Animais de Doenças , Fenômenos Eletrofisiológicos , Técnicas de Silenciamento de Genes , Síndrome do Intestino Irritável/induzido quimicamente , Síndrome do Intestino Irritável/tratamento farmacológico , Masculino , Neuralgia/tratamento farmacológico , Neuralgia/fisiopatologia , Nociceptores/fisiologia , Percepção da Dor/fisiologia , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley
11.
J Neurosci ; 31(16): 6059-66, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21508231

RESUMO

Iatrogenic pain consecutive to a large number of surgical procedures has become a growing health concern. The etiology and pathophysiology of postoperative pain are still poorly understood, but hydrogen ions appear to be important in this process. We have investigated the role of peripheral acid-sensing ion channels (ASICs), which form depolarizing channels activated by extracellular protons, in a rat model of postoperative pain (i.e., hindpaw skin/muscle incision). We report high levels of ASIC-type currents (∼ 77%) in sensory neurons innervating the hindpaw muscles, with a prevalence of ASIC3-like currents. The ASIC3 protein is largely expressed in lumbar DRG neurons innervating the plantar muscle, and its mRNA and protein levels are increased by plantar incision 24 h after surgery. Pharmacological inhibition of ASIC3 channels with the specific toxin APETx2 or in vivo knockdown of ASIC3 subunit by small interfering RNA led to a significant reduction of postoperative spontaneous, thermal, and postural pain behaviors (spontaneous flinching, heat hyperalgesia, and weight bearing). ASIC3 appears to have an important role in deep tissue but also affects prolonged pain evoked by skin incision alone. The specific homomeric ASIC1a blocker PcTx1 has no effect on spontaneous flinching, when applied peripherally. Together, these data demonstrate a significant role for peripheral ASIC3-containing channels in postoperative pain.


Assuntos
Hiperalgesia/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Dor Pós-Operatória/metabolismo , Células Receptoras Sensoriais/fisiologia , Canais de Sódio/metabolismo , Canais Iônicos Sensíveis a Ácido , Animais , Eletrofisiologia , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiopatologia , Membro Posterior/inervação , Membro Posterior/metabolismo , Hiperalgesia/fisiopatologia , Masculino , Músculo Esquelético/inervação , Músculo Esquelético/metabolismo , Medição da Dor , Dor Pós-Operatória/fisiopatologia , RNA Interferente Pequeno , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
J Neurosci ; 29(42): 13106-14, 2009 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-19846698

RESUMO

Lipoamino acids are anandamide-related endogenous molecules that induce analgesia via unresolved mechanisms. Here, we provide evidence that the T-type/Cav3 calcium channels are important pharmacological targets underlying their physiological effects. Various lipoamino acids, including N-arachidonoyl glycine (NAGly), reversibly inhibited Cav3.1, Cav3.2, and Cav3.3 currents, with potent effects on Cav3.2 [EC(50) approximately 200 nm for N-arachidonoyl 3-OH-gamma-aminobutyric acid (NAGABA-OH)]. This inhibition involved a large shift in the Cav3.2 steady-state inactivation and persisted during fatty acid amide hydrolase (FAAH) inhibition as well as in cell-free outside-out patch. In contrast, lipoamino acids had weak effects on high-voltage-activated (HVA) Cav1.2 and Cav2.2 calcium currents, on Nav1.7 and Nav1.8 sodium currents, and on anandamide-sensitive TRPV1 and TASK1 currents. Accordingly, lipoamino acids strongly inhibited native Cav3.2 currents in sensory neurons with small effects on sodium and HVA calcium currents. In addition, we demonstrate here that lipoamino acids NAGly and NAGABA-OH produced a strong thermal analgesia and that these effects (but not those of morphine) were abolished in Cav3.2 knock-out mice. Collectively, our data revealed lipoamino acids as a family of endogenous T-type channel inhibitors, suggesting that these ligands can modulate multiple cell functions via this newly evidenced regulation.


Assuntos
Analgésicos/farmacologia , Ácidos Araquidônicos/farmacologia , Canais de Cálcio Tipo T/metabolismo , Glicina/análogos & derivados , Ácido gama-Aminobutírico/análogos & derivados , Animais , Comportamento Animal/fisiologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo T/classificação , Canais de Cálcio Tipo T/genética , Células Cultivadas , Modelos Animais de Doenças , Estimulação Elétrica/métodos , Gânglios Espinais/citologia , Glicina/farmacologia , Proteínas de Fluorescência Verde/genética , Humanos , Hiperalgesia/tratamento farmacológico , Hiperalgesia/genética , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfina/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.7 , Proteínas do Tecido Nervoso/genética , Neuroblastoma , Técnicas de Patch-Clamp/métodos , Canais de Potássio de Domínios Poros em Tandem/genética , Células Receptoras Sensoriais , Canais de Sódio/genética , Canais de Cátion TRPV/genética , Transfecção , Ácido gama-Aminobutírico/farmacologia
13.
EMBO J ; 28(9): 1308-18, 2009 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-19279663

RESUMO

The sensation of cold or heat depends on the activation of specific nerve endings in the skin. This involves heat- and cold-sensitive excitatory transient receptor potential (TRP) channels. However, we show here that the mechano-gated and highly temperature-sensitive potassium channels of the TREK/TRAAK family, which normally work as silencers of the excitatory channels, are also implicated. They are important for the definition of temperature thresholds and temperature ranges in which excitation of nociceptor takes place and for the intensity of excitation when it occurs. They are expressed with thermo-TRP channels in sensory neurons. TRAAK and TREK-1 channels control pain produced by mechanical stimulation and both heat and cold pain perception in mice. Expression of TRAAK alone or in association with TREK-1 controls heat responses of both capsaicin-sensitive and capsaicin-insensitive sensory neurons. Together TREK-1 and TRAAK channels are important regulators of nociceptor activation by cold, particularly in the nociceptor population that is not activated by menthol.


Assuntos
Temperatura Baixa , Temperatura Alta , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Canais de Potássio/fisiologia , Sensação Térmica/fisiologia , Animais , Células Cultivadas , Eletrofisiologia , Gânglios Espinais/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Dor , Canais de Potássio/genética , Canais de Potássio de Domínios Poros em Tandem/genética , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo , Sensação Térmica/genética
14.
EMBO J ; 27(22): 3047-55, 2008 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-18923424

RESUMO

Acid-sensing ion channels (ASICs) are cationic channels activated by extracellular acidosis that are expressed in both central and peripheral nervous systems. Although peripheral ASICs seem to be natural sensors of acidic pain (e.g., in inflammation, ischaemia, lesions or tumours), a direct demonstration is still lacking. We show that approximately 60% of rat cutaneous sensory neurons express ASIC3-like currents. Native as well as recombinant ASIC3 respond synergistically to three different inflammatory signals that are slight acidifications (approximately pH 7.0), hypertonicity and arachidonic acid (AA). Moderate pH, alone or in combination with hypertonicity and AA, increases nociceptors excitability and produces pain suppressed by the toxin APETx2, a specific blocker of ASIC3. Both APETx2 and the in vivo knockdown of ASIC3 with a specific siRNA also have potent analgesic effects against primary inflammation-induced hyperalgesia in rat. Peripheral ASIC3 channels are thus essential sensors of acidic pain and integrators of molecular signals produced during inflammation where they contribute to primary hyperalgesia.


Assuntos
Inflamação/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Dor/metabolismo , Canais de Sódio/metabolismo , Canais Iônicos Sensíveis a Ácido , Acidose/metabolismo , Potenciais de Ação/fisiologia , Animais , Ácido Araquidônico/farmacologia , Células Cultivadas , Venenos de Cnidários/metabolismo , Gânglios Espinais/citologia , Temperatura Alta/efeitos adversos , Humanos , Soluções Hipertônicas , Inflamação/metabolismo , Masculino , Proteínas do Tecido Nervoso/genética , Neurônios Aferentes/citologia , Neurônios Aferentes/metabolismo , Medição da Dor , Peptídeos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Pele/efeitos dos fármacos , Pele/inervação , Canais de Sódio/genética , Venenos de Aranha/metabolismo
15.
Pain ; 137(1): 112-124, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17900808

RESUMO

Glutamate plays a key role in modulation of nociceptive processing. This excitatory amino acid exerts its action through two distinct types of receptors, ionotropic and metabotropic glutamate receptors (mGluRs). Eight mGluRs have been identified and divided in three groups based on their sequence similarity, pharmacology and G-protein coupling. While the role of group I and II mGluRs is now well established, little is known about the part played by group III mGluRs in pain. In this work, we studied comparatively the involvement of spinal group III mGluR in modulation of acute, inflammatory and neuropathic pain. While intrathecal injection of ACPT-I, a selective group III mGluR agonist, failed to induce any change in vocalization thresholds of healthy animals submitted to mechanical or thermal stimuli, it dose-dependently inhibited the nociceptive behavior of rats submitted to the formalin test and the mechanical hyperalgesia associated with different animal models of inflammatory (carrageenan-treated and monoarthritic rats) or neuropathic pain (mononeuropathic and vincristine-treated rats). Similar effects were also observed following intrathecal injection of PHCCC, a positive allosteric modulator of mGlu4. Antihyperalgesia induced by ACPT-I was blocked either by LY341495, a nonselective antagonist of mGluR, by MAP4, a selective group III antagonist. This study provide new evidences supporting the role of spinal group III mGluRs in the modulation of pain perception in different pathological pain states of various etiologies but not in normal conditions. It more particularly highlights the specific involvement of mGlu4 in this process and may be a useful therapeutic approach to chronic pain treatment.


Assuntos
Modelos Animais de Doenças , Hiperalgesia/prevenção & controle , Neuralgia/prevenção & controle , Receptores de Glutamato Metabotrópico/fisiologia , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Agonistas de Aminoácidos Excitatórios/farmacologia , Agonistas de Aminoácidos Excitatórios/uso terapêutico , Humanos , Hiperalgesia/fisiopatologia , Inflamação/fisiopatologia , Inflamação/prevenção & controle , Masculino , Neuralgia/fisiopatologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/agonistas
16.
Nat Neurosci ; 10(8): 943-5, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17632507

RESUMO

Psalmotoxin 1, a peptide extracted from the South American tarantula Psalmopoeus cambridgei, has very potent analgesic properties against thermal, mechanical, chemical, inflammatory and neuropathic pain in rodents. It exerts its action by blocking acid-sensing ion channel 1a, and this blockade results in an activation of the endogenous enkephalin pathway. The analgesic properties of the peptide are suppressed by antagonists of the mu and delta-opioid receptors and are lost in Penk1-/- mice.


Assuntos
Analgésicos/uso terapêutico , Encefalinas/fisiologia , Proteínas de Membrana/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Dor/tratamento farmacológico , Canais de Sódio/fisiologia , Venenos de Aranha/uso terapêutico , Canais Iônicos Sensíveis a Ácido , Animais , Área Sob a Curva , Comportamento Animal , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Encefalinas/deficiência , Proteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , Morfina/administração & dosagem , Naloxona/administração & dosagem , Naltrexona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Proteínas do Tecido Nervoso/deficiência , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Medição da Dor/métodos , Peptídeos , Precursores de Proteínas/deficiência , Tempo de Reação/efeitos dos fármacos , Canais de Sódio/deficiência , Medula Espinal/patologia , Fatores de Tempo
17.
EMBO J ; 25(11): 2368-76, 2006 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-16675954

RESUMO

The TREK-1 channel is a temperature-sensitive, osmosensitive and mechano-gated K+ channel with a regulation by Gs and Gq coupled receptors. This paper demonstrates that TREK-1 qualifies as one of the molecular sensors involved in pain perception. TREK-1 is highly expressed in small sensory neurons, is present in both peptidergic and nonpeptidergic neurons and is extensively colocalized with TRPV1, the capsaicin-activated nonselective ion channel. Mice with a disrupted TREK-1 gene are more sensitive to painful heat sensations near the threshold between anoxious warmth and painful heat. This phenotype is associated with the primary sensory neuron, as polymodal C-fibers were found to be more sensitive to heat in single fiber experiments. Knockout animals are more sensitive to low threshold mechanical stimuli and display an increased thermal and mechanical hyperalgesia in conditions of inflammation. They display a largely decreased pain response induced by osmotic changes particularly in prostaglandin E2-sensitized animals. TREK-1 appears as an important ion channel for polymodal pain perception and as an attractive target for the development of new analgesics.


Assuntos
Nociceptores/metabolismo , Dor/metabolismo , Percepção/fisiologia , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Gânglios Espinais/citologia , Hibridização In Situ , Camundongos , Camundongos Knockout , Fibras Nervosas Amielínicas/metabolismo , Neurônios Aferentes/citologia , Neurônios Aferentes/metabolismo , Medição da Dor , Técnicas de Patch-Clamp , Canais de Potássio de Domínios Poros em Tandem/genética , RNA Mensageiro/metabolismo
18.
Pain ; 114(3): 482-490, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15777873

RESUMO

The regulation of nociceptive processing by 5-HT at the spinal level is intricate since the neurotransmitter has been implicated in both pro and antinociception. The aim of our study was to investigate, according to the nature of the noxious stimulus, how the blockade of spinal 5-HT(1A) receptors could influence the antinociceptive actions of exogenous 5-HT as well as two analgesics involving endogenous 5-HT, paracetamol and venlafaxine. Rats were submitted either to the formalin test (tonic pain) or the paw pressure test (acute pain). WAY-100635 (40 microg/rat, i.t.), a selective 5-HT(1A) receptor antagonist, had no intrinsic action in either test. However, in the formalin test, it blocked the antinociceptive action of 5-HT (50 microg/rat, i.t.) and paracetamol (300 mg/kg, i.v.) in both phases of biting/licking behaviour and that of venlafaxine (2.5 mg/kg, s.c.) in the late phase only. In the paw pressure test, the combination of sub-effective doses of 5-HT (0.01 microg/rat, i.t.), paracetamol (50 mg/kg, i.v.) or venlafaxine (20 mg/kg, s.c.) with WAY-100635 led to a significant antinociceptive effect, which seems to depend on the reinforcement of the activity of inhibitory GABAergic interneurones. In conclusion, both direct stimulation of the spinal 5-HT(1A) receptors by 5-HT, and indirect stimulation using paracetamol or venlafaxine can differently influence pain transmission. We propose that the nature of the applied nociceptive stimulus would be responsible for the dual effect of the 5-HT(1A) receptors rather than the hyperalgesic state or the supraspinal integration of the pain message.


Assuntos
Acetaminofen/farmacologia , Analgésicos não Narcóticos/farmacologia , Piperazinas/farmacologia , Piridinas/farmacologia , Receptor 5-HT1A de Serotonina/fisiologia , Antagonistas da Serotonina/farmacologia , Medula Espinal/efeitos dos fármacos , Animais , Cicloexanóis/farmacologia , Interações Medicamentosas , Masculino , Nociceptores/efeitos dos fármacos , Nociceptores/fisiologia , Medição da Dor , Pressão , Ratos , Ratos Sprague-Dawley , Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Medula Espinal/fisiologia , Cloridrato de Venlafaxina
19.
EMBO J ; 24(2): 315-24, 2005 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-15616581

RESUMO

Analgesic therapies are still limited and sometimes poorly effective, therefore finding new targets for the development of innovative drugs is urgently needed. In order to validate the potential utility of blocking T-type calcium channels to reduce nociception, we explored the effects of intrathecally administered oligodeoxynucleotide antisenses, specific to the recently identified T-type calcium channel family (CaV3.1, CaV3.2, and CaV3.3), on reactions to noxious stimuli in healthy and mononeuropathic rats. Our results demonstrate that the antisense targeting CaV3.2 induced a knockdown of the CaV3.2 mRNA and protein expression as well as a large reduction of 'CaV3.2-like' T-type currents in nociceptive dorsal root ganglion neurons. Concomitantly, the antisense treatment resulted in major antinociceptive, anti-hyperalgesic, and anti-allodynic effects, suggesting that CaV3.2 plays a major pronociceptive role in acute and chronic pain states. Taken together, the results provide direct evidence linking CaV3.2 T-type channels to pain perception and suggest that CaV3.2 may offer a specific molecular target for the treatment of pain.


Assuntos
Canais de Cálcio Tipo T/genética , Inativação Gênica , Neurônios Aferentes/metabolismo , Animais , Sequência de Bases , Western Blotting , Primers do DNA , Dor/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Brain Res ; 1008(2): 288-92, 2004 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-15145768

RESUMO

In mononeuropathic rats submitted to a C-fiber reflex responses paradigm, repeated administration (five successive injections every half-life) of 10 mg/kg, s.c. of venlafaxine, but not of 2.5 mg/kg, s.c., a mixed monoamine reuptake inhibitor with preferential inhibitory activity in 5-HT reuptake, induced a progressive reduction of spinal wind-up. Repeated co-administration of the selective 5-HT1A receptor antagonist WAY 100,635 i.c.v. (50 microg/injection) significantly increased the effect of venlafaxine s.c., indicating that venlafaxine-induced inhibition of spinal wind-up in mononeuropathic rats is potentiated by blockade of central 5-HT1A receptors.


Assuntos
Cicloexanóis/farmacologia , Doenças do Sistema Nervoso Periférico/patologia , Receptor 5-HT1A de Serotonina/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Medula Espinal/citologia , Animais , Masculino , Fibras Nervosas Amielínicas/efeitos dos fármacos , Piperazinas/farmacologia , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Reflexo/efeitos dos fármacos , Antagonistas da Serotonina/farmacologia , Medula Espinal/efeitos dos fármacos , Cloridrato de Venlafaxina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...