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










Base de datos
Intervalo de año de publicación
1.
Pain ; 157(3): 552-559, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26492527

RESUMEN

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.


Asunto(s)
Bloqueadores del Canal Iónico Sensible al Ácido/uso terapéutico , Analgésicos/uso terapéutico , Venenos Elapídicos/uso terapéutico , Neuralgia/tratamiento farmacológico , Péptidos/uso terapéutico , Animales , Femenino , Inflamación/tratamiento farmacológico , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuralgia/patología , Ratas , Resultado del Tratamiento , Xenopus laevis
2.
Pain ; 155(12): 2534-2544, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25239074

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica/genética , Percepción del Dolor/fisiología , Umbral del Dolor/psicología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Sensación Térmica/genética , Animales , Antineoplásicos/toxicidad , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Hiperalgesia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibras Nerviosas Amielínicas/fisiología , Compuestos Organoplatinos/toxicidad , Oxaliplatino , Dimensión del Dolor , Percepción del Dolor/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Estimulación Física , Canales de Potasio/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Sensación Térmica/efectos de los fármacos
3.
Neuron ; 84(1): 123-136, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25242222

RESUMEN

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.


Asunto(s)
Analgesia/métodos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Receptores de GABA-B/fisiología , Secuencia de Aminoácidos , Animales , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Ratas
4.
Int Urogynecol J ; 25(8): 1081-7, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24599179

RESUMEN

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.


Asunto(s)
Ácido Hialurónico/uso terapéutico , Polipropilenos , Complicaciones Posoperatorias/prevención & control , Mallas Quirúrgicas , Adherencias Tisulares/prevención & control , Animales , Materiales Biocompatibles/uso terapéutico , Femenino , Fibrosis , Geles , Hernia Abdominal/cirugía , Complicaciones Posoperatorias/patología , Estudios Prospectivos , Falla de Prótesis/etiología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Adherencias Tisulares/complicaciones , Adherencias Tisulares/patología
5.
Pain ; 155(4): 764-772, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24447516

RESUMEN

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.


Asunto(s)
Acetaminofén/farmacología , Analgésicos no Narcóticos/farmacología , Canales de Calcio Tipo T/deficiencia , Médula Espinal/efectos de los fármacos , 4-Aminopiridina/farmacología , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Animales , Artritis/inducido químicamente , Artritis/tratamiento farmacológico , Canales de Calcio Tipo T/genética , Células Cultivadas , Vías de Administración de Medicamentos , Interacciones Farmacológicas , Ganglios Espinales/citología , Humanos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Noqueados , Morfina/farmacología , Neuronas/efectos de los fármacos , Dimensión del Dolor/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Médula Espinal/citología , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética , Tetraetilamonio/farmacología
6.
Nat Commun ; 4: 2941, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24346231

RESUMEN

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.


Asunto(s)
Analgesia/métodos , Morfina/efectos adversos , Morfina/uso terapéutico , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/uso terapéutico , Animales , Células COS , Chlorocebus aethiops , Estreñimiento , Cruzamientos Genéticos , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Naloxona/química , Dolor/tratamiento farmacológico , Manejo del Dolor , Receptores Opioides mu/metabolismo , Insuficiencia Respiratoria , Transducción de Señal , Factores de Tiempo
7.
Cell Rep ; 5(2): 378-88, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-24139797

RESUMEN

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.


Asunto(s)
Citocinas/metabolismo , Nociceptores/metabolismo , Dolor/fisiopatología , Estrés Mecánico , Animales , Carragenina/toxicidad , Citocinas/genética , Citocinas/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Dolor/metabolismo , Umbral del Dolor/efectos de los fármacos , Técnicas de Placa-Clamp , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
8.
Pain ; 154(2): 283-293, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23257507

RESUMEN

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.


Asunto(s)
Bencenoacetamidas/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/metabolismo , Hiperalgesia/tratamiento farmacológico , Neuronas/efectos de los fármacos , Piridinas/farmacología , Animales , Bloqueadores de los Canales de Calcio/uso terapéutico , Canales de Calcio Tipo T/genética , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Hiperalgesia/genética , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
9.
Nature ; 490(7421): 552-5, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-23034652

RESUMEN

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.


Asunto(s)
Bloqueadores del Canal Iónico Sensible al Ácido/farmacología , Canales Iónicos Sensibles al Ácido/metabolismo , Analgésicos/farmacología , Venenos Elapídicos/farmacología , Dolor/tratamiento farmacológico , Péptidos/farmacología , Péptidos/uso terapéutico , Bloqueadores del Canal Iónico Sensible al Ácido/química , Bloqueadores del Canal Iónico Sensible al Ácido/uso terapéutico , Canales Iónicos Sensibles al Ácido/clasificación , Canales Iónicos Sensibles al Ácido/genética , Analgésicos/efectos adversos , Analgésicos/química , Analgésicos/uso terapéutico , Animales , Tolerancia a Medicamentos , Venenos Elapídicos/administración & dosificación , Venenos Elapídicos/química , Venenos Elapídicos/uso terapéutico , Inyecciones Espinales , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Morfina/efectos adversos , Morfina/farmacología , Naloxona/farmacología , Nociceptores/química , Nociceptores/metabolismo , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Dolor/metabolismo , Péptidos/administración & dosificación , Péptidos/química , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Ratas , Insuficiencia Respiratoria/inducido químicamente , Xenopus laevis
10.
Proc Natl Acad Sci U S A ; 108(27): 11268-73, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21690417

RESUMEN

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.


Asunto(s)
Canales de Calcio Tipo T/fisiología , Colon/inervación , Colon/fisiopatología , Síndrome del Colon Irritable/fisiopatología , Animales , Secuencia de Bases , Butiratos/toxicidad , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo T/deficiencia , Canales de Calcio Tipo T/genética , Modelos Animales de Enfermedad , Fenómenos Electrofisiológicos , Técnicas de Silenciamiento del Gen , Síndrome del Colon Irritable/inducido químicamente , Síndrome del Colon Irritable/tratamiento farmacológico , Masculino , Neuralgia/tratamiento farmacológico , Neuralgia/fisiopatología , Nociceptores/fisiología , Percepción del Dolor/fisiología , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley
11.
J Neurosci ; 31(16): 6059-66, 2011 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-21508231

RESUMEN

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.


Asunto(s)
Hiperalgesia/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Dolor Postoperatorio/metabolismo , Células Receptoras Sensoriales/fisiología , Canales de Sodio/metabolismo , Canales Iónicos Sensibles al Ácido , Animales , Electrofisiología , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiopatología , Miembro Posterior/inervación , Miembro Posterior/metabolismo , Hiperalgesia/fisiopatología , Masculino , Músculo Esquelético/inervación , Músculo Esquelético/metabolismo , Dimensión del Dolor , Dolor Postoperatorio/fisiopatología , ARN Interferente Pequeño , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
J Neurosci ; 29(42): 13106-14, 2009 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-19846698

RESUMEN

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.


Asunto(s)
Analgésicos/farmacología , Ácidos Araquidónicos/farmacología , Canales de Calcio Tipo T/metabolismo , Glicina/análogos & derivados , Ácido gamma-Aminobutírico/análogos & derivados , Animales , Conducta Animal/fisiología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo T/clasificación , Canales de Calcio Tipo T/genética , Células Cultivadas , Modelos Animales de Enfermedad , Estimulación Eléctrica/métodos , Ganglios Espinales/citología , Glicina/farmacología , Proteínas Fluorescentes Verdes/genética , Humanos , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/genética , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/farmacología , Canal de Sodio Activado por Voltaje NAV1.7 , Proteínas del Tejido Nervioso/genética , Neuroblastoma , Técnicas de Placa-Clamp/métodos , Canales de Potasio de Dominio Poro en Tándem/genética , Células Receptoras Sensoriales , Canales de Sodio/genética , Canales Catiónicos TRPV/genética , Transfección , Ácido gamma-Aminobutírico/farmacología
13.
EMBO J ; 28(9): 1308-18, 2009 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-19279663

RESUMEN

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.


Asunto(s)
Frío , Calor , Canales de Potasio de Dominio Poro en Tándem/fisiología , Canales de Potasio/fisiología , Sensación Térmica/fisiología , Animales , Células Cultivadas , Electrofisiología , Ganglios Espinales/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Dolor , Canales de Potasio/genética , Canales de Potasio de Dominio Poro en Tándem/genética , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/metabolismo , Sensación Térmica/genética
14.
EMBO J ; 27(22): 3047-55, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-18923424

RESUMEN

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.


Asunto(s)
Inflamación/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Dolor/metabolismo , Canales de Sodio/metabolismo , Canales Iónicos Sensibles al Ácido , Acidosis/metabolismo , Potenciales de Acción/fisiología , Animales , Ácido Araquidónico/farmacología , Células Cultivadas , Venenos de Cnidarios/metabolismo , Ganglios Espinales/citología , Calor/efectos adversos , Humanos , Soluciones Hipertónicas , Inflamación/metabolismo , Masculino , Proteínas del Tejido Nervioso/genética , Neuronas Aferentes/citología , Neuronas Aferentes/metabolismo , Dimensión del Dolor , Péptidos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Piel/efectos de los fármacos , Piel/inervación , Canales de Sodio/genética , Venenos de Araña/metabolismo
15.
Pain ; 137(1): 112-124, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17900808

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Hiperalgesia/prevención & control , Neuralgia/prevención & control , Receptores de Glutamato Metabotrópico/fisiología , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Agonistas de Aminoácidos Excitadores/farmacología , Agonistas de Aminoácidos Excitadores/uso terapéutico , Humanos , Hiperalgesia/fisiopatología , Inflamación/fisiopatología , Inflamación/prevención & control , Masculino , Neuralgia/fisiopatología , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/agonistas
16.
Nat Neurosci ; 10(8): 943-5, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17632507

RESUMEN

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.


Asunto(s)
Analgésicos/uso terapéutico , Encefalinas/fisiología , Proteínas de la Membrana/fisiología , Proteínas del Tejido Nervioso/fisiología , Dolor/tratamiento farmacológico , Canales de Sodio/fisiología , Venenos de Araña/uso terapéutico , Canales Iónicos Sensibles al Ácido , Animales , Área Bajo la Curva , Conducta Animal , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Encefalinas/deficiencia , Proteínas de la Membrana/deficiencia , Ratones , Ratones Noqueados , Morfina/administración & dosificación , Naloxona/administración & dosificación , Naltrexona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Proteínas del Tejido Nervioso/deficiencia , Neuronas/efectos de los fármacos , Neuronas/fisiología , Dimensión del Dolor/métodos , Péptidos , Precursores de Proteínas/deficiencia , Tiempo de Reacción/efectos de los fármacos , Canales de Sodio/deficiencia , Médula Espinal/patología , Factores de Tiempo
17.
EMBO J ; 25(11): 2368-76, 2006 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-16675954

RESUMEN

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.


Asunto(s)
Nociceptores/metabolismo , Dolor/metabolismo , Percepción/fisiología , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Animales , Ganglios Espinales/citología , Hibridación in Situ , Ratones , Ratones Noqueados , Fibras Nerviosas Amielínicas/metabolismo , Neuronas Aferentes/citología , Neuronas Aferentes/metabolismo , Dimensión del Dolor , Técnicas de Placa-Clamp , Canales de Potasio de Dominio Poro en Tándem/genética , ARN Mensajero/metabolismo
18.
Pain ; 114(3): 482-490, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15777873

RESUMEN

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.


Asunto(s)
Acetaminofén/farmacología , Analgésicos no Narcóticos/farmacología , Piperazinas/farmacología , Piridinas/farmacología , Receptor de Serotonina 5-HT1A/fisiología , Antagonistas de la Serotonina/farmacología , Médula Espinal/efectos de los fármacos , Animales , Ciclohexanoles/farmacología , Interacciones Farmacológicas , Masculino , Nociceptores/efectos de los fármacos , Nociceptores/fisiología , Dimensión del Dolor , Presión , Ratas , Ratas Sprague-Dawley , Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Médula Espinal/fisiología , Clorhidrato de Venlafaxina
19.
EMBO J ; 24(2): 315-24, 2005 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-15616581

RESUMEN

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.


Asunto(s)
Canales de Calcio Tipo T/genética , Silenciador del Gen , Neuronas Aferentes/metabolismo , Animales , Secuencia de Bases , Western Blotting , Cartilla de ADN , Dolor/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Brain Res ; 1008(2): 288-92, 2004 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-15145768

RESUMEN

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.


Asunto(s)
Ciclohexanoles/farmacología , Enfermedades del Sistema Nervioso Periférico/patología , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Médula Espinal/citología , Animales , Masculino , Fibras Nerviosas Amielínicas/efectos de los fármacos , Piperazinas/farmacología , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Reflejo/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , Médula Espinal/efectos de los fármacos , Clorhidrato de Venlafaxina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...