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










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(1)2020 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-33379368

RESUMO

Transient receptor potential ankyrin member 1 (TRPA1) belongs to the family of thermo TRP cation channels that detect harmful temperatures, acids and numerous chemical pollutants. TRPA1 is expressed in nervous tissue, where it participates in the genesis of nociceptive signals in response to noxious stimuli and mediates mechanical hyperalgesia and allodynia associated with different neuropathies. The glutamate N-methyl-d-aspartate receptor (NMDAR), which plays a relevant role in allodynia to mechanical stimuli, is connected via histidine triad nucleotide-binding protein 1 (HINT1) and type 1 sigma receptor (σ1R) to mu-opioid receptors (MORs), which mediate the most potent pain relief. Notably, neuropathic pain causes a reduction in MOR antinociceptive efficacy, which can be reversed by blocking spinal NMDARs and TRPA1 channels. Thus, we studied whether TRPA1 channels form complexes with MORs and NMDARs that may be implicated in the aforementioned nociceptive signals. Our data suggest that TRPA1 channels functionally associate with MORs, delta opioid receptors and NMDARs in the dorsal root ganglia, the spinal cord and brain areas. These associations were altered in response to pharmacological interventions and the induction of inflammatory and also neuropathic pain. The MOR-TRPA1 and NMDAR-TRPA1 associations do not require HINT1 or σ1R but appear to be mediated by calcium-activated calmodulin. Thus, TRPA1 channels may associate with NMDARs to promote ascending acute and chronic pain signals and to control MOR antinociception.


Assuntos
Calmodulina/metabolismo , Ácido Glutâmico/metabolismo , Tecido Nervoso/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides/metabolismo , Canal de Cátion TRPA1/metabolismo , Animais , Constrição Patológica , Formaldeído , Inflamação/complicações , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Neuralgia/complicações , Neuralgia/metabolismo , Neuralgia/patologia , Especificidade de Órgãos , Ligação Proteica , Subunidades Proteicas/metabolismo
2.
Front Pharmacol ; 10: 613, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31263413

RESUMO

Sigma-1 (σ1) receptor antagonists are promising tools for neuropathic pain treatment, but it is unknown whether σ1 receptor inhibition ameliorates the neuropathic signs induced by nerve transection, in which the pathophysiological mechanisms and response to drug treatment differ from other neuropathic pain models. In addition, σ1 antagonism ameliorates inflammatory pain through modulation of the endogenous opioid system, but it is unknown whether this occurs during neuropathic pain. We investigated the effect of σ1 inhibition on the painful hypersensitivity associated with the spared nerve injury (SNI) model in mice. Wild-type (WT) mice developed prominent cold (acetone test), mechanical (von Frey test), and heat hypersensitivity (Hargreaves test) after SNI. σ1 receptor knockout (ခσ1-KO) mice did not develop cold allodynia and showed significantly less mechanical allodynia, although they developed heat hyperalgesia after SNI. The systemic acute administration of the selective σ1 receptor antagonist S1RA attenuated all three types of SNI-induced hypersensitivity in WT mice. These ameliorative effects of S1RA were reversed by the administration of the σ1 agonist PRE-084, and were absent in σ1-KO mice, indicating the selectivity of S1RA-induced effects. The opioid antagonist naloxone and its peripherally restricted analog naloxone methiodide prevented S1RA-induced effects in mechanical and heat hypersensitivity, but not in cold allodynia, indicating that opioid-dependent and -independent mechanisms are involved in the effects of this σ1 antagonist. The repeated administration of S1RA twice a day during 10 days reduced SNI-induced cold, mechanical, and heat hypersensitivity without inducing analgesic tolerance during treatment. These effects were observed up to 12 h after the last administration, when S1RA was undetectable in plasma or brain, indicating long-lasting pharmacodynamic effects. These data suggest that σ1 antagonism may have therapeutic value for the treatment of neuropathic pain induced by the transection of peripheral nerves.

3.
Adv Exp Med Biol ; 964: 109-132, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28315268

RESUMO

The sigma-1 receptor is a unique ligand-operated chaperone present in key areas for pain control, in both the peripheral and central nervous system. Sigma-1 receptors interact with a variety of protein targets to modify their function. These targets include several G-protein-coupled receptors such as the µ-opioid receptor, and ion channels such as the N-methyl-D-aspartate receptor (NMDAR). Sigma-1 antagonists modify the chaperoning activity of sigma-1 receptor by increasing opioid signaling and decreasing NMDAR responses, consequently enhancing opioid antinociception and decreasing the sensory hypersensitivity that characterizes pathological pain conditions. However, the participation in pain relief of other protein partners of sigma-1 receptors in addition to opioid receptors and NMDARs cannot be ruled out. The enhanced opioid antinociception by sigma-1 antagonism is not accompanied by an increase in opioid side effects , including tolerance, dependence or constipation, so the use of sigma-1 antagonists may increase the therapeutic index of opioids. Furthermore, sigma-1 antagonists (in the absence of opioids) have been shown to exert antinociceptive effects in preclinical models of neuropathic pain induced by nerve trauma or chemical injury (the antineoplastic paclitaxel), and more recently in inflammatory and ischemic pain. Although most studies attributed the analgesic properties of sigma-1 antagonists to their central actions, it is now known that peripheral sigma-1 receptors also participate in their effects. Overwhelming preclinical evidence of the role of sigma-1 receptors in pain has led to the development of the first selective sigma-1 antagonist with an intended indication for pain treatment, which is currently in Phase II clinical trials.


Assuntos
Analgésicos/farmacologia , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Receptores sigma/antagonistas & inibidores , Receptores sigma/metabolismo , Animais , Tolerância a Medicamentos/fisiologia , Humanos , Receptores Opioides mu/metabolismo , Receptor Sigma-1
4.
Inflamm Res ; 64(6): 377-81, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25902777

RESUMO

INTRODUCTION: The sigma-1 receptor (Sig-1R) is a unique ligand-regulated molecular chaperone that interacts with several protein targets such as G protein-coupled receptors and ion channels to modulate their activity. Sig-1R is located in areas of the central and peripheral nervous system that are key to pain control. Previous preclinical studies have suggested a potential therapeutic use of Sig-1R antagonists for the management of neuropathic pain. DISCUSSION: Recent studies using pharmacological and genetic tools have explored the role of Sig-1R in inflammatory pain conditions. Mice lacking the Sig-1R have shown different patterns of phenotypic responses to inflammatory injury. Systemic or peripheral administration of several Sig-1R antagonists, including the selective Sig-1R antagonist S1RA, inhibited both mechanical and thermal hypersensitivity in several preclinical models of inflammatory pain. These recent studies are summarized in the present commentary. CONCLUSION: Central and peripheral pharmacological blockade of Sig-1R could be an effective option to treat inflammatory pain.


Assuntos
Inflamação/fisiopatologia , Dor/fisiopatologia , Receptores sigma , Animais , Humanos , Inflamação/complicações , Camundongos , Dor/etiologia , Receptores sigma/antagonistas & inibidores , Receptores sigma/genética , Receptores sigma/fisiologia , Receptor Sigma-1
5.
J Pharmacol Exp Ther ; 348(1): 32-45, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24155346

RESUMO

We evaluated the effects of σ1-receptor inhibition on µ-opioid-induced mechanical antinociception and constipation. σ1-Knockout mice exhibited marked mechanical antinociception in response to several µ-opioid analgesics (fentanyl, oxycodone, morphine, buprenorphine, and tramadol) at systemic (subcutaneous) doses that were inactive in wild-type mice and even unmasked the antinociceptive effects of the peripheral µ-opioid agonist loperamide. Likewise, systemic (subcutaneous) or local (intraplantar) treatment of wild-type mice with the selective σ1 antagonists BD-1063 [1-[2-(3,4-dichlorophenyl)ethyl]-4-methylpiperazine dihydrochloride] or S1RA [4-[2-[[5-methyl-1-(2-naphthalenyl)1H-pyrazol-3-yl]oxy]ethyl] morpholine hydrochloride] potentiated µ-opioid antinociception; these effects were fully reversed by the σ1 agonist PRE-084 [2-(4-morpholinethyl)1-phenylcyclohexanecarboxylate) hydrochloride], showing the selectivity of the pharmacological approach. The µ-opioid antinociception potentiated by σ1 inhibition (by σ1-receptor knockout or σ1-pharmacological antagonism) was more sensitive to the peripherally restricted opioid antagonist naloxone methiodide than opioid antinociception under normal conditions, indicating a key role for peripheral opioid receptors in the enhanced antinociception. Direct interaction between the opioid drugs and σ1 receptor cannot account for our results, since the former lacked affinity for σ1 receptors (labeled with [(3)H](+)-pentazocine). A peripheral role for σ1 receptors was also supported by their higher density (Western blot results) in peripheral nervous tissue (dorsal root ganglia) than in several central areas involved in opioid antinociception (dorsal spinal cord, basolateral amygdala, periaqueductal gray, and rostroventral medulla). In contrast to its effects on nociception, σ1-receptor inhibition did not alter fentanyl- or loperamide-induced constipation, a peripherally mediated nonanalgesic opioid effect. Therefore, σ1-receptor inhibition may be used as a systemic or local adjuvant to enhance peripheral µ-opioid analgesia without affecting opioid-induced constipation.


Assuntos
Analgésicos Opioides/farmacologia , Medição da Dor/métodos , Receptores Opioides mu/fisiologia , Receptores sigma/fisiologia , Analgésicos Opioides/antagonistas & inibidores , Animais , Constipação Intestinal/induzido quimicamente , Constipação Intestinal/genética , Constipação Intestinal/metabolismo , Feminino , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiologia , Camundongos , Camundongos Knockout , Receptores Opioides mu/metabolismo , Receptores sigma/deficiência , Receptores sigma/genética , Receptor Sigma-1
6.
Neuropharmacology ; 70: 348-58, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23524304

RESUMO

We studied the modulation of morphine-induced mechanical antinociception and side effects by σ1 receptor inhibition. Both wild-type (WT) and σ1 receptor knockout (σ1-KO) mice showed similar responses to paw pressure (100-600 g). The systemic (subcutaneous) or local (intraplantar) administration of σ1 antagonists (BD-1063, BD-1047, NE-100 and S1RA) was devoid of antinociceptive effects in WT mice. However, σ1-KO mice exhibited an enhanced mechanical antinociception in response to systemic morphine (1-16 mg/kg). Similarly, systemic treatment of WT mice with σ1 antagonists markedly potentiated morphine-induced antinociception, and its effects were reversed by the selective σ1 agonist PRE-084. Although the local administration of morphine (50-200 µg) was devoid of antinociceptive effects in WT mice, it induced dose-dependent antinociception in σ1-KO mice. This effect was limited to the injected paw. Enhancement of peripheral morphine antinociception was replicated in WT mice locally co-administered with σ1 antagonists and the opioid. None of the σ1 antagonists tested enhanced morphine-antinociception in σ1-KO mice, confirming a σ1-mediated action. Morphine-induced side-effects (hyperlocomotion and inhibition of gastrointestinal transit) were unaltered in σ1-KO mice. These results cannot be explained by a direct interaction of σ1 ligands with µ-opioid receptors or adaptive changes of µ-receptors in σ1-KO mice, given that [(3)H]DAMGO binding in forebrain, spinal cord, and hind-paw skin membranes was unaltered in mutant mice, and none of the σ1 drugs tested bound to µ-opioid receptors. These results show that σ1 receptor inhibition potentiates morphine-induced mechanical analgesia but not its acute side effects, and that this enhanced analgesia can be induced at peripheral level.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Receptores sigma/antagonistas & inibidores , Analgésicos Opioides/uso terapêutico , Animais , Sinergismo Farmacológico , Ala(2)-MePhe(4)-Gly(5)-Encefalina , Motilidade Gastrointestinal/efeitos dos fármacos , Membro Posterior/metabolismo , Hiperalgesia/tratamento farmacológico , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Prosencéfalo/metabolismo , Receptores sigma/genética , Receptores sigma/metabolismo , Medula Espinal/metabolismo , Trítio , Receptor Sigma-1
7.
J Pain ; 13(11): 1107-21, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23063344

RESUMO

UNLABELLED: Sigma-1 (σ(1)) receptors play a role in different types of pain and in central sensitization mechanisms; however, it is unknown whether they are involved in chemotherapy-induced neuropathic pain. We compared the ability of paclitaxel to induce cold (acetone test) and mechanical (electronic Von Frey test) allodynia in wild-type (WT) and σ(1) receptor knockout (σ(1)-KO) mice. We also tested the effect on paclitaxel-induced painful neuropathy of BD-1063 (16-64 mg/kg, subcutaneously) and S1RA (32-128 mg/kg, subcutaneously), 2 selective σ(1) receptor antagonists that bind to the σ(1) receptor with high affinity and competitively. The responses to cold and mechanical stimuli were similar in WT and σ(1)-KO mice not treated with paclitaxel; however, treatment with paclitaxel (2 mg/kg, intraperitoneally, once per day during 5 consecutive days) produced cold and mechanical allodynia and an increase in spinal cord diphosphorylated extracellular signal-regulated kinase (pERK) in WT but not in σ(1)-KO mice. The administration of BD-1063 or S1RA 30 minutes before each paclitaxel dose prevented the development of cold and mechanical allodynia in WT mice. Moreover, the acute administration of both σ(1) receptor antagonists dose dependently reversed both types of paclitaxel-induced allodynia after they had fully developed. These results suggest that σ(1) receptors play a key role in paclitaxel-induced painful neuropathy. PERSPECTIVE: Antagonists of the σ(1) receptor may have therapeutic value for the treatment and/or prevention of paclitaxel-induced neuropathic pain. This possibility is especially interesting in the context of chemotherapy-induced neuropathy, where the onset of nerve damage is predictable and preventive treatment could be administered.


Assuntos
Antineoplásicos Fitogênicos , Neuralgia/induzido quimicamente , Paclitaxel , Receptores sigma/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Western Blotting , Encéfalo/metabolismo , Temperatura Baixa , Feminino , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Sistema de Sinalização das MAP Quinases/fisiologia , Membranas/efeitos dos fármacos , Membranas/metabolismo , Camundongos , Camundongos Knockout , Morfolinas/farmacologia , Entorpecentes/metabolismo , Medição da Dor/efeitos dos fármacos , Pentazocina/metabolismo , Estimulação Física , Piperazinas/farmacologia , Equilíbrio Postural/efeitos dos fármacos , Pirazóis/farmacologia , Receptores sigma/agonistas , Receptores sigma/genética , Receptor Sigma-1
8.
Mar Drugs ; 10(2): 281-305, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22412801

RESUMO

Tetrodotoxin (TTX) is a potent neurotoxin that blocks voltage-gated sodium channels (VGSCs). VGSCs play a critical role in neuronal function under both physiological and pathological conditions. TTX has been extensively used to functionally characterize VGSCs, which can be classified as TTX-sensitive or TTX-resistant channels according to their sensitivity to this toxin. Alterations in the expression and/or function of some specific TTX-sensitive VGSCs have been implicated in a number of chronic pain conditions. The administration of TTX at doses below those that interfere with the generation and conduction of action potentials in normal (non-injured) nerves has been used in humans and experimental animals under different pain conditions. These data indicate a role for TTX as a potential therapeutic agent for pain. This review focuses on the preclinical and clinical evidence supporting a potential analgesic role for TTX. In addition, the contribution of specific TTX-sensitive VGSCs to pain is reviewed.


Assuntos
Analgésicos não Narcóticos/uso terapêutico , Neurotoxinas/uso terapêutico , Bloqueadores dos Canais de Sódio/uso terapêutico , Canais de Sódio/metabolismo , Tetrodotoxina/uso terapêutico , Dor Aguda/tratamento farmacológico , Dor Aguda/imunologia , Dor Aguda/metabolismo , Animais , Humanos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuralgia/tratamento farmacológico , Neuralgia/imunologia , Neuralgia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Canais de Sódio/química , Canais de Sódio/genética
9.
Pain ; 143(3): 252-261, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19375855

RESUMO

We evaluated the role of sigma(1) receptors on capsaicin-induced mechanical hypersensitivity and on nociceptive pain induced by punctate mechanical stimuli, using wild-type and sigma(1) receptor knockout (sigma(1)-KO) mice and selective sigma(1) receptor-acting drugs. Mutation in sigma(1)-KO mice was confirmed by PCR analysis of genomic DNA and, at the protein level, by [(3)H](+)-pentazocine binding assays. Both wild-type and sigma(1)-KO mice not treated with capsaicin showed similar responses to different intensities of mechanical stimuli (0.05-8 g force), ranging from innocuous to noxious, applied to the hind paw. This indicates that sigma(1) gene inactivation does not modify the perception of punctate mechanical stimuli. The intraplantar (i.pl.) administration of capsaicin induced dose-dependent mechanical allodynia in wild-type mice (markedly reducing both the threshold force necessary to induce paw withdrawal and the latency to paw withdrawal induced by a given force). In contrast, capsaicin was completely unable to induce mechanical hypersensitivity in sigma(1)-KO mice. The high-affinity and selective sigma(1) antagonists BD-1063, BD-1047 and NE-100, administered subcutaneously (s.c.), dose-dependently inhibited mechanical allodynia induced by capsaicin (1 microg,i.pl.), yielding ED(50) (mg/kg) values of 15.80+/-0.93, 29.31+/-1.65 and 40.74+/-7.20, respectively. The effects of the sigma(1) antagonists were reversed by the sigma(1) agonist PRE-084 (32 mg/kg, s.c.). None of the drugs tested modified the responses induced by a painful mechanical punctate stimulus (4 g force) in nonsensitized animals. These results suggest that sigma(1) receptors are essential for capsaicin-induced mechanical hypersensitivity, but are not involved in mechanical nociceptive pain.


Assuntos
Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Nociceptores/metabolismo , Limiar da Dor/fisiologia , Receptores sigma/genética , Medula Espinal/metabolismo , Animais , Anisóis/farmacologia , Capsaicina/farmacologia , DNA/análise , Etilenodiaminas/farmacologia , Feminino , Hiperalgesia/induzido quimicamente , Camundongos , Camundongos Knockout , Morfolinas/farmacologia , Nociceptores/efeitos dos fármacos , Medição da Dor/métodos , Limiar da Dor/efeitos dos fármacos , Estimulação Física , Piperazinas/farmacologia , Reação em Cadeia da Polimerase , Propilaminas/farmacologia , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Receptores sigma/agonistas , Receptores sigma/antagonistas & inibidores , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Fármacos do Sistema Sensorial/farmacologia , Medula Espinal/efeitos dos fármacos , Receptor Sigma-1
10.
Eur J Pharmacol ; 500(1-3): 203-19, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15464034

RESUMO

Four families of potassium channels with different structures, functional characteristics and pharmacological sensitivity, are distinguished in neurons: voltage-gated (K(v)), calcium-activated (K(Ca)), inward rectifier (K(ir)) and two-pore (K(2P)) K(+) channels. During the last 15 years, numerous studies have demonstrated that the opening of some of these K(+) channels plays an important role in the antinociception induced by agonists of many G-protein-coupled receptors (alpha(2)-adrenoceptors, opioid, GABA(B), muscarinic M(2), adenosine A(1), serotonin 5-HT(1A) and cannabinoid receptors), as well as by other antinociceptive drugs (nonsteroidal antiinflammatory drugs [NSAIDs], tricyclic antidepressants, etc.) and natural products. Several specific types of K(+) channels are involved in antinociception. The most widely studied are the ATP-sensitive K(+) channels (K(ATP)), members of the K(ir) family, which participate in the antinociception induced by many drugs that activate them in both the central and the peripheral nervous system. The opening of G-protein-regulated inwardly rectifying K(+) channels (GIRK or K(ir)3), K(v)1.1 and two types of K(Ca) channels, the small- and large-conductance calcium-activated K(+) channels (SK and BK channels, respectively), also play a role in the antinociceptive effect of different drugs and natural products. Recently, drugs that open K(+) channels by direct activation (such as openers of neuronal K(v)7 and K(ATP) channels) have been shown to produce antinociception in models of acute and chronic pain, which suggests that other neuronal K(+) channels (e.g. K(v)1.4 channels) may represent an interesting target for the development of new K(+) channel openers with antinociceptive effects.


Assuntos
Dor/metabolismo , Canais de Potássio/fisiologia , Analgésicos não Narcóticos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Ensaios Clínicos como Assunto , Humanos , Ativação do Canal Iônico , Dor/tratamento farmacológico , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/agonistas , Receptores Acoplados a Proteínas G/agonistas , Receptores Opioides/agonistas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...