Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 180(8): 1148-1167, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36478100

RESUMO

BACKGROUND AND PURPOSE: Peripheral sensitization contributes to pathological pain. While prostaglandin E2 (PGE2) and nerve growth factor (NGF) sensitize peptidergic C-nociceptors (TRPV1+), glial cell line-derived neurotrophic factor (GDNF) sensitizes non-peptidergic C-neurons (IB4+). The sigma-1 receptor (sigma-1R) is a Ca2+ -sensing chaperone known to modulate opoid analgesia. This receptor binds both to TRPV1 and the µ opioid receptor, although the functional repercussions of these physical interactions in peripheral sensitization are unknown. EXPERIMENTAL APPROACH: We tested the effects of sigma-1 antagonism on PGE2-, NGF-, and GDNF-induced mechanical and heat hyperalgesia in mice. We used immunohistochemistry to determine the presence of endomorphin-2, an endogenous µ receptor agonist, on dorsal root ganglion (DRG) neurons. Recombinant proteins were used to study the interactions between sigma-1R, µ- receptor, and TRPV1. We used calcium imaging to study the effects of sigma-1 antagonism on PGE2-induced sensitization of TRPV1+ nociceptors. KEY RESULTS: Sigma1 antagonists reversed PGE2- and NGF-induced hyperalgesia but not GDNF-induced hyperalgesia. Endomorphin-2 was detected on TRPV1+ but not on IB4+ neurons. Peripheral opioid receptor antagonism by naloxone methiodide or administration of an anti-endomorphin-2 antibody to a sensitized paw reversed the antihyperalgesia induced by sigma-1 antagonists. Sigma-1 antagonism transfers sigma-1R from TRPV1 to µ receptors, suggesting that sigma-1R participate in TRPV1-µ receptor crosstalk. Moreover, sigma-1 antagonism reversed, in a naloxone-sensitive manner, PGE2-induced sensitization of DRG neurons to the calcium flux elicited by capsaicin, the prototypic TRPV1 agonist. CONCLUSION AND IMPLICATIONS: Sigma-1 antagonism harnesses endogenous opioids produced by TRPV1+ neurons to reduce hyperalgesia by increasing µ receptor activity.


Assuntos
Analgesia , Nociceptores , Camundongos , Animais , Nociceptores/metabolismo , Hiperalgesia/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacologia , Fator de Crescimento Neural/metabolismo , Cálcio/metabolismo , Dinoprostona/metabolismo , Dor/metabolismo , Peptídeos Opioides/metabolismo , Canais de Cátion TRPV/metabolismo , Gânglios Espinais/metabolismo , Receptor Sigma-1
2.
Neurosci Biobehav Rev ; 113: 238-261, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32147529

RESUMO

Pain measures traditionally used in rodents record mere reflexes evoked by sensory stimuli; the results thus may not fully reflect the human pain phenotype. Alterations in physical and emotional functioning, pain-depressed behaviors and facial pain expressions were recently proposed as additional pain outcomes to provide a more accurate measure of clinical pain in rodents, and hence to potentially enhance analgesic drug development. We aimed to review how preclinical pain assessment has evolved since the development of the tail flick test in 1941, with a particular focus on a critical analysis of some nonstandard pain outcomes, and a consideration of how sex differences may affect the performance of these pain surrogates. We tracked original research articles in Medline for the following periods: 1973-1977, 1983-1987, 1993-1997, 2003-2007, and 2014-2018. We identified 606 research articles about alternative surrogate pain measures, 473 of which were published between 2014 and 2018. This indicates that preclinical pain assessment is moving toward the use of these measures, which may soon become standard procedures in preclinical pain laboratories.


Assuntos
Analgésicos , Dor , Analgésicos/uso terapêutico , Animais , Modelos Animais de Doenças , Feminino , Masculino , Dor/tratamento farmacológico , Medição da Dor , Caracteres Sexuais
3.
Front Pharmacol ; 11: 613068, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33551815

RESUMO

Both TRPA1 and purinergic P2X receptors have been proposed as potential targets for the treatment of visceral pain. We found that the intracolonic administration of a low dose mustard oil (0.5%), a well-known TRPA1 agonist, produced nociceptive responses and abdominal wall referred mechanical hyperalgesia, without inducing apparent tissue damage. Both nociceptive responses and referred hyperalgesia were abolished by the ablation of TRPV1-expressing neurons (and the consequent ablation of TRPA1+ nociceptors) by resiniferatoxin (RTX) treatment, and by the TRPA1 antagonist AP18. However, a higher dose of mustard oil (2.5%) damaged the colonic epithelium and induced pERK activation in the spinal cord, and these processes were clearly independent of TRPV1-expressing neurons ablated by RTX. This higher dose of mustard oil induced nociceptive responses and referred mechanical hyperalgesia which were insensitive or only slightly sensitive to resiniferatoxin or AP18, but were markedly reduced by the P2X antagonist TNP-ATP, which is known to inhibit nociceptive actions induced by ATP released from injured tissues. In conclusion, whereas a low dose of intracolonic mustard oil induces visceral pain in a manner fully dependent on TRPA1 actions, when a high dose of this chemical irritant is used, visceral pain becomes mostly independent of TRPA1 activation but clearly enhanced by ATP purportedly released by the damaged colonic epithelium. Therefore, TRPA1 inhibition is not sufficient to substantially decrease visceral pain during tissue injury, whereas purinergic antagonism appears to be a more effective strategy.

4.
Front Pharmacol ; 10: 136, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30853912

RESUMO

Sigma-1 receptor antagonism increases the effects of morphine on nociceptive pain, even in morphine-tolerant animals. However, it is unknown whether these receptors are able to modulate morphine antinociception and tolerance during inflammatory pain. Here we used a mouse model to test the modulation of morphine effects by the selective sigma-1 antagonist S1RA (MR309), by determining its effect on inflammatory tactile allodynia (von Frey filaments) and on grip strength deficits induced by joint inflammation (a measure of pain-induced functional disability), and compared the results with those for nociceptive heat pain recorded with the unilateral hot plate (55°C) test. The subcutaneous (s.c.) administration of morphine induced antinociceptive effects to heat stimuli, and restored mechanical threshold and grip strength in mice with periarticular inflammation induced by Complete Freund's Adjuvant. S1RA (80 mg/kg, s.c.) administered alone did not induce any effect on nociceptive heat pain or inflammatory allodynia, but was able to partially reverse grip strength deficits. The association of S1RA with morphine, at doses inducing little or no analgesic-like effects when administered alone, resulted in a marked antinociceptive effect to heat stimuli and complete reversion of inflammatory tactile allodynia. However, S1RA administration did not increase the effect of morphine on grip strength deficits induced by joint inflammation. When S1RA (80 mg/kg, s.c.) was administered to morphine-tolerant animals, it rescued the analgesic-like effects of this opioid in all three pain measures. However, when S1RA was repeatedly given during the induction of morphine tolerance (and not on the day of behavioral evaluation) it failed to affect tolerance to the effects of morphine on nociceptive heat pain or inflammatory allodynia, but completely preserved the effects of this opioid on grip strength deficits. These effects of S1RA on morphine tolerance cannot be explained by pharmacokinetic interactions, given that the administration of S1RA did not modify concentrations of morphine or morphine-3-glucuronide (a major morphine metabolite) in morphine-tolerant animals in plasma or brain tissue. We conclude that sigma-1 receptors play a pivotal role in the control of morphine analgesia and tolerance in nociceptive and inflammatory pain, although in a manner dependent on the type of painful stimulus explored.

5.
Neuropharmacology ; 135: 328-342, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29580951

RESUMO

Morphine induces peripherally µ-opioid-mediated antinociception to heat but not to mechanical stimulation. Peripheral sigma-1 receptors tonically inhibit µ-opioid antinociception to mechanical stimuli, but it is unknown whether they modulate µ-opioid heat antinociception. We hypothesized that sigma-1 receptors might play a role in the modality-specific peripheral antinociceptive effects of morphine and other clinically relevant µ-opioid agonists. Mechanical nociception was assessed in mice with the paw pressure test (450 g), and heat nociception with the unilateral hot plate (55 °C) test. Local peripheral (intraplantar) administration of morphine, buprenorphine or oxycodone did not induce antinociception to mechanical stimulation but had dose-dependent antinociceptive effects on heat stimuli. Local sigma-1 antagonism unmasked peripheral antinociception by µ-opioid agonists to mechanical stimuli, but did not modify their effects on heat stimulation. TRPV1+ and IB4+ cells are segregated populations of small neurons in the dorsal root ganglia (DRG) and the density of sigma-1 receptors was higher in IB4+ cells than in the rest of small nociceptive neurons. The in vivo ablation of TRPV1-expressing neurons with resiniferatoxin did not alter IB4+ neurons in the DRG, mechanical nociception, or the effects of sigma-1 antagonism on local morphine antinociception in this type of stimulus. However, it impaired the responses to heat stimuli and the effect of local morphine on heat nociception. In conclusion, peripheral opioid antinociception to mechanical stimuli is limited by sigma-1 tonic inhibitory actions, whereas peripheral opioid antinociception to heat stimuli (produced in TRPV1-expressing neurons) is not. Therefore, sigma-1 receptors contribute to the modality-specific peripheral effects of opioid analgesics.


Assuntos
Analgésicos Opioides/farmacologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Receptores Opioides mu/agonistas , Receptores sigma/metabolismo , Animais , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Temperatura Alta , Hiperalgesia/patologia , Camundongos Knockout , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Nociceptores/patologia , Distribuição Aleatória , Receptores Opioides mu/metabolismo , Receptores sigma/agonistas , Receptores sigma/antagonistas & inibidores , Receptores sigma/genética , Canais de Cátion TRPV/metabolismo , Tato , Receptor Sigma-1
6.
Pharmacol Res ; 131: 224-230, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29454675

RESUMO

Immune cells have a known role in pronociception, since they release a myriad of inflammatory algogens which interact with neurons to facilitate pain signaling. However, these cells also produce endogenous opioid peptides with analgesic potential. The sigma-1 receptor is a ligand-operated chaperone that modulates neurotransmission by interacting with multiple protein partners, including the µ-opioid receptor. We recently found that sigma-1 antagonists are able to induce opioid analgesia by enhancing the action of endogenous opioid peptides of immune origin during inflammation. This opioid analgesia is seen only at the inflamed site, where immune cells naturally accumulate. In this article we review the difficulties of targeting the opioid system for selective pain relief, and discuss the dual role of immune cells in pain and analgesia. Our discussion creates perspectives for possible novel therapeutic uses of sigma-1 antagonists as agents able to maximize the analgesic potential of the immune system.


Assuntos
Analgésicos Opioides/uso terapêutico , Terapia de Alvo Molecular/métodos , Dor/tratamento farmacológico , Receptores sigma/antagonistas & inibidores , Analgesia/métodos , Analgésicos Opioides/farmacologia , Animais , Humanos , Inflamação/complicações , Inflamação/tratamento farmacológico , Inflamação/imunologia , Dor/complicações , Dor/imunologia , Receptores sigma/imunologia , Receptor Sigma-1
7.
J Med Chem ; 61(1): 372-384, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29220177

RESUMO

Methoxycarbonyl-1-phenyl-2-cyclopropylmethyl based derivatives cis-(+)-1a [cis-(+)-MR200], cis-(-)-1a [cis-(-)-MR201], and trans-(±)-1a [trans-(±)-MR204], have been identified as new potent sigma (σ) receptor ligands. In the present paper, novel enantiomerically pure analogues were synthesized and optimized for their σ receptor affinity and selectivity. Docking studies rationalized the results obtained in the radioligand binding assay. Absolute stereochemistry was unequivocally established by X-ray analysis of precursor trans-(+)-5a as camphorsulfonyl derivative 9. The most promising compound, trans-(+)-1d, showed remarkable selectivity over a panel of more than 15 receptors as well as good chemical and enzymatic stability in human plasma. An in vivo evaluation evidenced that trans-(+)-1d, in contrast to trans-(-)-1d, cis-(+)-1d, or cis-(-)-1d, which behave as σ1 antagonists, exhibited a σ1 agonist profile. These data clearly demonstrated that compound trans-(+)-1d, due to its σ1 agonist activity and favorable receptor selectivity and stability, provided an useful tool for the study of σ1 receptors.


Assuntos
Analgésicos/química , Analgésicos/metabolismo , Ciclopropanos/química , Ciclopropanos/metabolismo , Piperidinas/química , Piperidinas/metabolismo , Receptores sigma/metabolismo , Analgésicos/farmacologia , Animais , Ciclopropanos/farmacologia , Feminino , Ligantes , Camundongos , Modelos Moleculares , Simulação de Acoplamento Molecular , Piperidinas/farmacologia , Conformação Proteica , Receptores sigma/química , Solubilidade , Estereoisomerismo , Relação Estrutura-Atividade , Especificidade por Substrato , Água/química
8.
Neuropharmacology ; 125: 231-242, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28760650

RESUMO

Grip strength deficit is a measure of pain-induced functional disability in rheumatic disease. We tested whether this parameter and tactile allodynia, the standard pain measure in preclinical studies, show parallels in their response to analgesics and basic mechanisms. Mice with periarticular injections of complete Freund's adjuvant (CFA) in the ankles showed periarticular immune infiltration and synovial membrane alterations, together with pronounced grip strength deficits and tactile allodynia measured with von Frey hairs. However, inflammation-induced tactile allodynia lasted longer than grip strength alterations, and therefore did not drive the functional deficits. Oral administration of the opioid drugs oxycodone (1-8 mg/kg) and tramadol (10-80 mg/kg) induced a better recovery of grip strength than acetaminophen (40-320 mg/kg) or the nonsteroidal antiinflammatory drugs ibuprofen (10-80 mg/kg) or celecoxib (40-160 mg/kg); these results are consistent with their analgesic efficacy in humans. Functional impairment was generally a more sensitive indicator of drug-induced analgesia than tactile allodynia, as drug doses that attenuated grip strength deficits showed little or no effect on von Frey thresholds. Finally, ruthenium red (a nonselective TRP antagonist) or the in vivo ablation of TRPV1-expressing neurons with resiniferatoxin abolished tactile allodynia without altering grip strength deficits, indicating that the neurobiology of tactile allodynia and grip strength deficits differ. In conclusion, grip strength deficits are due to a distinct type of pain that reflects an important aspect of the human pain experience, and therefore merits further exploration in preclinical studies to improve the translation of new analgesics from bench to bedside.


Assuntos
Artrite/diagnóstico , Força da Mão , Hiperalgesia/diagnóstico , Força Muscular , Medição da Dor , Doenças Reumáticas/diagnóstico , Acetaminofen/farmacologia , Analgésicos/farmacologia , Animais , Artrite/tratamento farmacológico , Artrite/patologia , Artrite/fisiopatologia , Celecoxib/farmacologia , Modelos Animais de Doenças , Diterpenos/farmacologia , Feminino , Adjuvante de Freund , Hiperalgesia/tratamento farmacológico , Hiperalgesia/patologia , Hiperalgesia/fisiopatologia , Ibuprofeno/farmacologia , Inflamação/diagnóstico , Inflamação/tratamento farmacológico , Inflamação/patologia , Inflamação/fisiopatologia , Força Muscular/efeitos dos fármacos , Nociceptores/efeitos dos fármacos , Nociceptores/metabolismo , Nociceptores/patologia , Oxicodona/farmacologia , Medição da Dor/métodos , Doenças Reumáticas/tratamento farmacológico , Doenças Reumáticas/patologia , Doenças Reumáticas/fisiopatologia , Rutênio Vermelho/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo , Tarso Animal , Tato , Tramadol/farmacologia
9.
Proc Natl Acad Sci U S A ; 114(31): 8396-8401, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28716934

RESUMO

Sigma-1 antagonism potentiates the antinociceptive effects of opioid drugs, so sigma-1 receptors constitute a biological brake to opioid drug-induced analgesia. The pathophysiological role of this process is unknown. We aimed to investigate whether sigma-1 antagonism reduces inflammatory pain through the disinhibition of the endogenous opioidergic system in mice. The selective sigma-1 antagonists BD-1063 and S1RA abolished mechanical and thermal hyperalgesia in mice with carrageenan-induced acute (3 h) inflammation. Sigma-1-mediated antihyperalgesia was reversed by the opioid antagonists naloxone and naloxone methiodide (a peripherally restricted naloxone analog) and by local administration at the inflamed site of monoclonal antibody 3-E7, which recognizes the pan-opioid sequence Tyr-Gly-Gly-Phe at the N terminus of most endogenous opioid peptides (EOPs). Neutrophils expressed pro-opiomelanocortin, the precursor of ß-endorphin (a known EOP), and constituted the majority of the acute immune infiltrate. ß-endorphin levels increased in the inflamed paw, and this increase and the antihyperalgesic effects of sigma-1 antagonism were abolished by reducing the neutrophil load with in vivo administration of an anti-Ly6G antibody. The opioid-dependent sigma-1 antihyperalgesic effects were preserved 5 d after carrageenan administration, where macrophages/monocytes were found to express pro-opiomelanocortin and to now constitute the majority of the immune infiltrate. These results suggest that immune cells harboring EOPs are needed for the antihyperalgesic effects of sigma-1 antagonism during inflammation. In conclusion, sigma-1 receptors curtail immune-driven peripheral opioid analgesia, and sigma-1 antagonism produces local opioid analgesia by enhancing the action of EOPs of immune origin, maximizing the analgesic potential of immune cells that naturally accumulate in painful inflamed areas.


Assuntos
Analgesia/métodos , Analgésicos Opioides/farmacologia , Morfolinas/farmacologia , Naloxona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Receptores sigma/antagonistas & inibidores , Animais , Antígenos Ly/imunologia , Carragenina/toxicidade , Feminino , Inflamação/tratamento farmacológico , Inflamação/patologia , Macrófagos/metabolismo , Camundongos , Naloxona/farmacologia , Neutrófilos/metabolismo , Oligopeptídeos/metabolismo , Dor/tratamento farmacológico , Piperazinas/farmacologia , Pró-Opiomelanocortina/biossíntese , Pirazóis/farmacologia , Compostos de Amônio Quaternário/farmacologia , Receptores sigma/metabolismo , Receptor Sigma-1
10.
EMBO Mol Med ; 7(5): 670-87, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25802402

RESUMO

Primary coenzyme Q10 (CoQ10) deficiency is due to mutations in genes involved in CoQ biosynthesis. The disease has been associated with five major phenotypes, but a genotype-phenotype correlation is unclear. Here, we compare two mouse models with a genetic modification in Coq9 gene (Coq9(Q95X) and Coq9(R239X)), and their responses to 2,4-dihydroxybenzoic acid (2,4-diHB). Coq9(R239X) mice manifest severe widespread CoQ deficiency associated with fatal encephalomyopathy and respond to 2,4-diHB increasing CoQ levels. In contrast, Coq9(Q95X) mice exhibit mild CoQ deficiency manifesting with reduction in CI+III activity and mitochondrial respiration in skeletal muscle, and late-onset mild mitochondrial myopathy, which does not respond to 2,4-diHB. We show that these differences are due to the levels of COQ biosynthetic proteins, suggesting that the presence of a truncated version of COQ9 protein in Coq9(R239X) mice destabilizes the CoQ multiprotein complex. Our study points out the importance of the multiprotein complex for CoQ biosynthesis in mammals, which may provide new insights to understand the genotype-phenotype heterogeneity associated with human CoQ deficiency and may have a potential impact on the treatment of this mitochondrial disorder.


Assuntos
Ataxia/patologia , Variação Genética , Genótipo , Doenças Mitocondriais/patologia , Debilidade Muscular/patologia , Ubiquinona/deficiência , Animais , Modelos Animais de Doenças , Hidroxibenzoatos/administração & dosagem , Hidroxibenzoatos/toxicidade , Mamíferos , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Ubiquinona/genética
11.
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
12.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...