Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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.

2.
Sci Rep ; 7(1): 13506, 2017 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-29044153

RESUMO

Paclitaxel (PTX) is one of the drugs of choice in the treatment of breast and lung cancer. However, its severe side effects, including mielosuppression, cardiotoxicity and neurotoxicity, frequently cause treatment to be discontinued. Solid lipid nanoparticles (NPs) of glyceril tripalmitate (tripalmitin) loaded with PTX (Tripalm-NPs-PTX) including modifications by the addition of hexa(ethylene glycol), ß-cyclodextrin and macelignan were developed. All NPs-PTX formulations displayed excellent hemocompatibility and significantly enhanced PTX antitumor activity in human breast (MCF7, MDAMB231, SKBR3 and T47D) and lung (A549, NCI-H520 and NCI-H460) cancer cells. Tripalm-NPs-PTX decreased PTX IC50 by as much as 40.5-fold in breast and 38.8-fold in lung cancer cells and Tripalm-NPs-PTX macelignan inhibited P-glycoprotein in resistant tumor cells. In addition, Tripalm-NPs-PTX significantly decreased the volume of breast and lung multicellular tumor spheroids that mimics in vivo tumor mass. Finally, Tripalm-NPs-PTX decreased the PTX IC50 of cancer stem cells (CSCs) derived from both lung and breast cancer cells (6.7- and 14.9-fold for MCF7 and A549 CSCs, respectively). These results offer a new PTX nanoformulation based on the use of tripalmitin which improves the antitumor activity of PTX and that may serve as an alternative PTX delivery system in breast and lung cancer treatment.


Assuntos
Antineoplásicos/administração & dosagem , Nanopartículas/química , Paclitaxel/administração & dosagem , Triglicerídeos/química , Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular , Feminino , Humanos , Lignanas/química , Neoplasias Pulmonares/patologia , Células MCF-7 , Células-Tronco Neoplásicas/efeitos dos fármacos , Paclitaxel/farmacologia , Polietilenoglicóis/química , Esferoides Celulares/efeitos dos fármacos , Células Tumorais Cultivadas , beta-Ciclodextrinas/química
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.
J Physiol ; 595(8): 2661-2679, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28105664

RESUMO

KEY POINTS: Voltage-gated sodium channels play a fundamental role in determining neuronal excitability. Specifically, voltage-gated sodium channel subtype NaV 1.7 is required for sensing acute and inflammatory somatic pain in mice and humans but its significance in pain originating from the viscera is unknown. Using comparative behavioural models evoking somatic and visceral pain pathways, we identify the requirement for NaV 1.7 in regulating somatic (noxious heat pain threshold) but not in visceral pain signalling. These results enable us to better understand the mechanisms underlying the transduction of noxious stimuli from the viscera, suggest that the investigation of pain pathways should be undertaken in a modality-specific manner and help to direct drug discovery efforts towards novel visceral analgesics. ABSTRACT: Voltage-gated sodium channel NaV 1.7 is required for acute and inflammatory pain in mice and humans but its significance for visceral pain is unknown. Here we examine the role of NaV 1.7 in visceral pain processing and the development of referred hyperalgesia using a conditional nociceptor-specific NaV 1.7 knockout mouse (NaV 1.7Nav1.8 ) and selective small-molecule NaV 1.7 antagonist PF-5198007. NaV 1.7Nav1.8 mice showed normal nociceptive behaviours in response to intracolonic application of either capsaicin or mustard oil, stimuli known to evoke sustained nociceptor activity and sensitization following tissue damage, respectively. Normal responses following induction of cystitis by cyclophosphamide were also observed in both NaV 1.7Nav1.8 and littermate controls. Loss, or blockade, of NaV 1.7 did not affect afferent responses to noxious mechanical and chemical stimuli in nerve-gut preparations in mouse, or following antagonism of NaV 1.7 in resected human appendix stimulated by noxious distending pressures. However, expression analysis of voltage-gated sodium channel α subunits revealed NaV 1.7 mRNA transcripts in nearly all retrogradely labelled colonic neurons, suggesting redundancy in function. By contrast, using comparative somatic behavioural models we identify that genetic deletion of NaV 1.7 (in NaV 1.8-expressing neurons) regulates noxious heat pain threshold and that this can be recapitulated by the selective NaV 1.7 antagonist PF-5198007. Our data demonstrate that NaV 1.7 (in NaV 1.8-expressing neurons) contributes to defined pain pathways in a modality-dependent manner, modulating somatic noxious heat pain, but is not required for visceral pain processing, and advocate that pharmacological block of NaV 1.7 alone in the viscera may be insufficient in targeting chronic visceral pain.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7/deficiência , Nociceptores/metabolismo , Dor Visceral/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Capsaicina/toxicidade , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Mostardeira/toxicidade , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Dor Nociceptiva/induzido quimicamente , Dor Nociceptiva/genética , Dor Nociceptiva/metabolismo , Nociceptores/efeitos dos fármacos , Óleos de Plantas/toxicidade , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia , Dor Visceral/induzido quimicamente , Dor Visceral/genética
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.
Eur J Pharmacol ; 711(1-3): 63-72, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23632394

RESUMO

While opioids are potent analgesics widely used in the management of pain, a number of well-known adverse effects limit their use. The sigma-1 receptor is a ligand-regulated molecular chaperone involved in pain processing, including modulation of opioid antinociception. However, data supporting the potential use of sigma-1 receptor ligands as suitable opioid adjuvants are based on studies that use non selective ligands. Also, safety issues derived from combination therapy are poorly addressed. In this study we used the new selective sigma-1 receptor antagonist S1RA (E-52862) to characterize the effect of selective sigma-1 receptor blockade on opioid-induced efficacy- and safety-related outcomes in mice. S1RA (40 mg/kg) had no effect in the tail-flick test but did enhance the antinociceptive potency of several opioids by a factor between 2 and 3.3. The potentiating effect of S1RA on morphine antinociception did not occur in sigma-1 receptor knockout mice, which supports the selective involvement of the sigma-1 receptor. Interestingly, S1RA co-administration restored morphine antinociception in tolerant mice and reverted the reward effects of morphine in the conditioned place preference paradigm. In addition, enhancement of antinociception was not accompanied by potentiation of other opioid-induced effects, such as the development of morphine analgesic tolerance, physical dependence, inhibition of gastrointestinal transit, or mydriasis. The use of sigma-1 receptor antagonists as opioid adjuvants could represent a promising pharmacological strategy to enhance opioid potency and, most importantly, to increase the safety margin of opioids. S1RA is currently in phase II clinical trials for the treatment of several pain conditions.


Assuntos
Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacologia , Receptores sigma/antagonistas & inibidores , Animais , Comportamento Animal/efeitos dos fármacos , Quimioterapia Adjuvante , Condicionamento Psicológico/efeitos dos fármacos , Sinergismo Farmacológico , Tolerância a Medicamentos , Trânsito Gastrointestinal/efeitos dos fármacos , Técnicas de Inativação de Genes , Intestinos/efeitos dos fármacos , Intestinos/fisiologia , Masculino , Camundongos , Morfina/efeitos adversos , Morfina/farmacologia , Midríase/induzido quimicamente , Naloxona/farmacologia , Receptores sigma/deficiência , Receptores sigma/genética , Recompensa , Comportamento Espacial/efeitos dos fármacos , Receptor Sigma-1
7.
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
8.
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
9.
Cent Nerv Syst Agents Med Chem ; 9(3): 172-83, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20021351

RESUMO

A large number of therapeutic roles have been proposed for sigma(1) receptors but the involvement of sigma(1) receptor in non-acute pain had not been well explored up to now. sigma(1) receptor knock-out mice became available offering us the possibility to study the role of sigma(1) receptor in nociception, particularly in models where central sensitization processes play a significant role. Given the attractive therapeutic potential, we have developed a chemical program aimed at the discovery of novel and selective sigma(1) ligands. Herein we discuss the rational basis of this approach and report preliminary pharmacological results of several chemical series and aspects of their structure-activity relationship on sigma(1) receptor. Functional data in pain models are presented mainly on one series that provide evidence to consider selective sigma(1) receptor antagonists an innovative and alternative approach for treating neuropathic pain.


Assuntos
Analgésicos Opioides/uso terapêutico , Dor/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Receptores sigma/antagonistas & inibidores , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Receptores sigma/química , Receptores sigma/efeitos dos fármacos , Receptores sigma/metabolismo , Receptor Sigma-1
10.
Pain ; 145(3): 294-303, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19505761

RESUMO

Sigma-1 receptor (sigma(1)R) is expressed in key CNS areas involved in nociceptive processing but only limited information is available about its functional role. In the present study we investigated the relevance of sigma(1)R in modulating nerve injury-evoked pain. For this purpose, wild-type mice and mice lacking the sigma(1)R gene were exposed to partial sciatic nerve ligation and neuropathic pain-related behaviors were investigated. To explore underlying mechanisms, spinal processing of repetitive nociceptive stimulation and expression of extracellular signal-regulated kinase (ERK) were also investigated. Sensitivity to noxious heat of homozygous sigma(1)R knockout mice did not differ from wild-type mice. Baseline values obtained in sigma(1)R knockout mice before nerve injury in the plantar, cold-plate and von Frey tests were also indistinguishable from those obtained in wild-type mice. However, cold and mechanical allodynia did not develop in sigma(1)R null mice exposed to partial sciatic nerve injury. Using isolated spinal cords we found that mice lacking sigma(1)R showed reduced wind-up responses respect to wild-type mice, as evidenced by a reduced number of action potentials induced by trains of C-fiber intensity stimuli. In addition, in contrast to wild-type mice, sigma(1)R knockout mice did not show increased phosphorylation of ERK in the spinal cord after sciatic nerve injury. Both wind-up and ERK activation have been related to mechanisms of spinal cord sensitization. Our findings identify sigma(1)R as a constituent of the mechanisms modulating activity-induced sensitization in pain pathways and point to sigma(1)R as a new potential target for drugs designed to alleviate neuropathic pain.


Assuntos
Hiperalgesia/etiologia , Limiar da Dor/fisiologia , Receptores sigma/fisiologia , Neuropatia Ciática/complicações , Neuropatia Ciática/genética , Medula Espinal/fisiopatologia , Análise de Variância , Animais , Biofísica , Modelos Animais de Doenças , Estimulação Elétrica/métodos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica/genética , Hiperalgesia/genética , Hiperalgesia/patologia , Masculino , Camundongos , Camundongos Knockout , Medição da Dor , Estimulação Física/efeitos adversos , Estimulação Física/métodos , Tempo de Reação/genética , Receptores sigma/deficiência , Neuropatia Ciática/patologia , Receptor Sigma-1
11.
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
12.
Pain ; 141(3): 239-247, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19118950

RESUMO

This work aimed to evaluate the potential role of the 5-HT(7) receptor in nociception secondary to a sensitizing stimulus in mice. For this purpose, the effects of relevant ligands (5-HT(7) receptor agonists: AS-19, MSD-5a, E-55888; 5-HT(7) receptor antagonists: SB-258719, SB-269970; 5-HT(1A) receptor agonist: F-13640; 5-HT(1A) receptor antagonist: WAY-100635) were assessed on capsaicin-induced mechanical hypersensitivity, a pain behavior involving hypersensitivity of dorsal horn neurons (central sensitization). For the 5-HT(7) receptor agonists used, binding profile and intrinsic efficacy to stimulate cAMP formation in HEK-293F cells expressing the human 5-HT(7) receptor were also evaluated. AS-19 and E-55888 were selective for 5-HT(7) receptors. E-55888 was a full agonist whereas AS-19 and MSD-5a behaved as partial agonists, with maximal effects corresponding to 77% and 61%, respectively, of the cAMP response evoked by the full agonist 5-HT. Our in vivo results revealed that systemic administration of 5-HT(7) receptor agonists exerted a clear-cut dose-dependent antinociceptive effect that was prevented by 5-HT(7) receptor antagonists, but not by the 5-HT(1A) receptor antagonist. The order of efficacy (E-55888>AS-19>MSD-5a) matched their in vitro efficacy as 5-HT(7) receptor agonists. Contrary to agonists, a dose-dependent promotion of mechanical hypersensitivity was observed after administration of 5-HT(7) receptor antagonists, substantiating the involvement of the 5-HT(7) receptor in the control of capsaicin-induced mechanical hypersensitivity. These findings suggest that serotonin exerts an inhibitory role in the control of nociception through activation of 5-HT(7) receptors, and point to a new potential therapeutic use of 5-HT(7) receptor agonists in the field of analgesia.


Assuntos
Capsaicina , Hiperalgesia/induzido quimicamente , Limiar da Dor/efeitos dos fármacos , Receptores de Serotonina/metabolismo , Animais , Linhagem Celular Transformada , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Humanos , Hiperalgesia/tratamento farmacológico , Masculino , Camundongos , Medição da Dor , Ligação Proteica/efeitos dos fármacos , Pirazóis/farmacologia , Tempo de Reação/efeitos dos fármacos , Receptores de Serotonina/efeitos dos fármacos , Serotoninérgicos/farmacologia
13.
Pain ; 137(3): 520-531, 2008 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-18037242

RESUMO

We evaluated the effect of low doses of systemically administered tetrodotoxin (TTX) on the development and expression of neuropathic pain induced by paclitaxel in mice. Treatment with paclitaxel (2mg/kg, i.p., once daily during 5 days) produced long-lasting (2-4 weeks) heat hyperalgesia (plantar test), mechanical allodynia (electronic Von Frey test) and cold allodynia (acetone drop method), with maximum effects observed on days 7, 10 and 10-14, respectively. Acute subcutaneous treatment with 1 or 3 microg/kg of TTX reduced the expression of mechanical allodynia, whereas higher doses (3 or 6 microg/kg) were required to reduce the expression of cold allodynia and heat hyperalgesia. In contrast, TTX (3 or 6 microg/kg, s.c.) did not affect the response to the same thermal and mechanical stimuli in control animals, which indicates that the antihyperalgesic and antiallodynic effects of TTX were not due to unspecific inhibition of the perception of these stimuli. Administration of TTX (6 microg/kg, s.c.) 30 min before each of the 5 doses of paclitaxel did not modify the development of heat hyperalgesia produced by the antineoplastic, but abolished the development of mechanical and cold allodynia. Coadministration of a lower dose of TTX (3 microg/kg) also prevented the development of mechanical allodynia. No signs of TTX-induced toxicity or motor incoordination were observed. These data suggest that low doses of TTX can be useful to prevent and treat paclitaxel-induced neuropathic pain, and that TTX-sensitive subtypes of sodium channels play a role in the pathogenesis of chemotherapy-induced neuropathic pain.


Assuntos
Hiperalgesia/induzido quimicamente , Hiperalgesia/prevenção & controle , Neuralgia/induzido quimicamente , Neuralgia/prevenção & controle , Paclitaxel/efeitos adversos , Medição da Dor/efeitos dos fármacos , Tetrodotoxina/administração & dosagem , Anestésicos Locais/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Relação Dose-Resposta a Droga , Feminino , Camundongos
14.
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
15.
Eur J Pharmacol ; 458(3): 291-7, 2003 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-12504785

RESUMO

The goal of this study was to investigate the effects of endomorphin-1 on Na(+),K(+)-ATPase activity in mouse brain synaptosome in vitro, and its antinociceptive interaction with the Na(+),K(+)-ATPase inhibitor ouabain. Endomorphin-1 (0.1 nM-10 microM) produced a concentration-dependent (EC(50): 43.19 nM, CI: 23.38-65.71 nM, E(max): 25.86%, CI: 24.53-27.20%), naloxone-reversible increase of the synaptosomal Na(+),K(+)-ATPase activity. The intrathecally (i.t.) administered endomorphin-1 (2-20 microg) produced a dose-dependent short-lasting increase in the tail-flick latency. Ouabain itself (1-1000 ng, i.t.) did not cause antinociception. Treatment with 10 ng ouabain significantly decreased the antinociceptive effect of 2 microg endomorphin-1, but none of the other combinations did significantly differ from the endomorhin-1-treated groups. These data indicate that endomorphin-1 increases the activity of Na(+),K(+)-ATPase in vitro but this effect may play a weak role in the antinociception induced by intrathecal endomorphin-1.


Assuntos
Analgésicos Opioides/farmacologia , Oligopeptídeos/farmacologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Sinaptossomos/efeitos dos fármacos , Analgésicos/farmacologia , Animais , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Injeções Espinhais , Masculino , Camundongos , Morfina/farmacologia , Ouabaína/farmacologia , Dor/prevenção & controle , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Sinaptossomos/enzimologia , Fatores de Tempo
16.
Brain Res ; 957(2): 311-9, 2002 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-12445973

RESUMO

Morphine through mu-opioid receptors and G(i/o) proteins modulates several cellular effector systems; however, the mechanisms involved in the regulation of Na(+),K(+)-ATPase are not well known. We evaluated the effect of two mu-opioid receptor agonists on ouabain-sensitive Na(+),K(+)-ATPase activity in mice forebrain synaptosomes, and examined the modulation of this effect by antagonists of opioid receptors and a blocker of G(i/o) proteins. Incubation of synaptosomes with morphine (10(-9) to 10(-4) M) or buprenorphine (10(-10) to 10(-5) M) concentration-dependently stimulated Na(+),K(+)-ATPase activity, morphine being less potent but more efficacious than buprenorphine. Morphine did not displace [3H]ouabain from its binding site (Na(+),K(+)-ATPase) to forebrain membranes, whereas ouabain did so in a concentration-dependent manner. Naloxone, an opioid antagonist (10(-6) M), added to the synaptosomal medium, antagonized the enhancement of Na(+),K(+)-ATPase activity induced by morphine, producing a parallel shift to the right of the morphine concentration-response curve. Treatment with beta-funaltrexamine, a mu antagonist (2.5 and 10 microg/mouse, i.c.v.) and naloxonazine, a mu1 antagonist (35 mg/kg, s.c.), 24 h before the synaptosomes were obtained, produced a dose-dependent reduction in the E(max) of the morphine-induced increase in Na(+),K(+)-ATPase activity in vitro, but did not significantly modify its EC(50). Pertussis toxin (G(i/o) protein blocker) treatment at a dose of 0.5 microg/mouse, administered i.c.v. 5 days before the synaptosomes were obtained, completely abolished the enhancement of Na(+),K(+)-ATPase activity induced by morphine. A lower dose (0.25 microg/mouse) decreased the E(max) of morphine by 50% but did not significantly affect its EC(50). These results suggest that morphine indirectly enhances Na(+),K(+)-ATPase activity in the brain by activating mu-opioid receptors and G(i/o) proteins.


Assuntos
Encéfalo/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Proteínas de Ligação ao GTP/efeitos dos fármacos , Morfina/farmacologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , ATPase Trocadora de Sódio-Potássio/efeitos dos fármacos , Animais , Encéfalo/enzimologia , Buprenorfina/farmacologia , Membrana Celular/enzimologia , Relação Dose-Resposta a Droga , Interações Medicamentosas/fisiologia , Inibidores Enzimáticos/farmacologia , Feminino , Proteínas de Ligação ao GTP/metabolismo , Camundongos , Antagonistas de Entorpecentes/farmacologia , Ouabaína/farmacologia , Toxina Pertussis/farmacologia , Terminações Pré-Sinápticas/enzimologia , Receptores Opioides mu/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...