Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuropharmacology ; 205: 108907, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34856203

RESUMO

Pioglitazone, an agonist at peroxisome proliferator-activated receptor gamma, is FDA-approved for the treatment of insulin resistance in type 2 diabetes. Numerous studies in male rodents suggest that pioglitazone inhibits inflammatory and neuropathic pain, but few included female subjects. To address this gap, we compared the effects of pioglitazone in both sexes in the intraplantar methylglyoxal model (MG) model of chemical pain and painful diabetic neuropathy (PDN), the plantar incision model (PIM) of postoperative pain, the spared nerve injury (SNI) model of traumatic nerve injury, and the ZDF rat and db/db mouse models of PDN. We administered pioglitazone by one-time intrathecal or intraperitoneal injection or by adding it to chow for 6 weeks, followed by measurement of hypersensitivity to non-noxious mechanical, noxious mechanical, heat, and/or cold stimuli. In all mouse models, injection of pioglitazone decreased pain-like behaviors with greater potency and/or efficacy in females as compared to males: heat and mechanical hypersensitivity in the MG model (0.1-10 mg/kg); mechanical hypersensitivity in the PIM model (10 µg); mechanical and cold hypersensitivity in the SNI model (100 mg/kg); and heat hypersensitivity in the db/db model (100 mg/kg). Furthermore, co-administration of low doses of morphine (1 mg/kg) and pioglitazone (10 mg/kg) decreased SNI-induced mechanical and cold hypersensitivity in female but not male mice. In the ZDF rat, pioglitazone (100 mg/kg) decreased heat and mechanical hypersensitivity with no sex difference. In the db/db model, pioglitazone had no effect when given into chow for 6 weeks at 0.3, 3 or 30 mg/kg doses. We conclude that females exhibit greater anti-hyperalgesic responses to pioglitazone in mouse models of chemical-induced nociception, postsurgical pain, neuropathic pain, and PDN. These findings set the stage for clinical trials to determine whether pioglitazone has analgesic properties across a broad spectrum of chronic pain conditions, particularly in women.


Assuntos
Analgésicos/farmacologia , Hiperalgesia/tratamento farmacológico , Neuralgia/tratamento farmacológico , Dor Nociceptiva/tratamento farmacológico , PPAR gama/agonistas , Dor Pós-Operatória/tratamento farmacológico , Pioglitazona/farmacologia , Analgésicos/administração & dosagem , Animais , Neuropatias Diabéticas/complicações , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Morfina/farmacologia , Neuralgia/etiologia , Dor Nociceptiva/induzido quimicamente , Dor Pós-Operatória/etiologia , Traumatismos dos Nervos Periféricos/complicações , Pioglitazona/administração & dosagem , Aldeído Pirúvico/farmacologia , Caracteres Sexuais
2.
Science ; 341(6152): 1394-9, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-24052307

RESUMO

Opioid receptor antagonists increase hyperalgesia in humans and animals, which indicates that endogenous activation of opioid receptors provides relief from acute pain; however, the mechanisms of long-term opioid inhibition of pathological pain have remained elusive. We found that tissue injury produced µ-opioid receptor (MOR) constitutive activity (MOR(CA)) that repressed spinal nociceptive signaling for months. Pharmacological blockade during the posthyperalgesia state with MOR inverse agonists reinstated central pain sensitization and precipitated hallmarks of opioid withdrawal (including adenosine 3',5'-monophosphate overshoot and hyperalgesia) that required N-methyl-D-aspartate receptor activation of adenylyl cyclase type 1. Thus, MOR(CA) initiates both analgesic signaling and a compensatory opponent process that generates endogenous opioid dependence. Tonic MOR(CA) suppression of withdrawal hyperalgesia may prevent the transition from acute to chronic pain.


Assuntos
Dor Crônica/metabolismo , Hiperalgesia/metabolismo , Dor Nociceptiva/metabolismo , Receptores Opioides mu/metabolismo , Dor Aguda/metabolismo , Monofosfato de Adenosina/metabolismo , Adenilil Ciclases/metabolismo , Animais , Modelos Animais de Doenças , Adjuvante de Freund/farmacologia , Hiperalgesia/induzido quimicamente , Isoflurano/farmacologia , Masculino , Camundongos , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo
3.
Neuropharmacology ; 70: 236-46, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23415633

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARγ) is emerging as a new pharmacotherapeutic target for chronic pain. When oral (3-30 mg/kg/day in chow for 7 wk) or twice-daily intraperitoneal (1-10 mg/kg/day for 2 wk) administration began before spared nerve injury (SNI), pioglitazone, a PPARγ agonist, dose-dependently prevented multiple behavioral signs of somatosensory hypersensitivity. The highest dose of intraperitoneal pioglitazone did not produce ataxia or reductions in transient mechanical and heat nociception, indicating that inhibitory effects on hypersensitivity were not secondary to adverse drug-induced behaviors or antinociception. Inhibitory effects on hypersensitivity persisted at least one week beyond cessation of pioglitazone administration, suggestive of long-lasting effects on gene expression. Blockade of PPARγ with GW9662, an irreversible and selective PPARγ antagonist, dose-dependently reduced the inhibitory effect of pioglitazone on hypersensitivity, indicating a PPARγ-dependent action. Remarkably, a single preemptive injection of pioglitazone 15 min before SNI attenuated hypersensitivity for at least 2 weeks; this was enhanced with a second injection delivered 12 h after SNI. Pioglitazone injections beginning after SNI also reduced hypersensitivity, albeit to a lesser degree than preemptive treatment. Intraperitoneal pioglitazone significantly reduced the nerve injury-induced up-regulation of cd11b, GFAP, and p-p38 in the dorsal horn, indicating a mechanism of action involving spinal microglia and/or astrocyte activation. Oral pioglitazone significantly reduced touch stimulus-evoked phospho-extracellular signal-related kinase (p-ERK) in lamina I-II, indicating a mechanism of action involving inhibition of central sensitization. We conclude that pioglitazone reduces spinal glial and stimulus-evoked p-ERK activation and that PPARγ activation blocks the development of and reduces established neuropathic pain.


Assuntos
Neuralgia/fisiopatologia , PPAR gama/fisiologia , Tiazolidinedionas/uso terapêutico , Anilidas/farmacologia , Animais , Ataxia/induzido quimicamente , Antígeno CD11b/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Hiperalgesia/prevenção & controle , Masculino , Neuralgia/tratamento farmacológico , Neuralgia/prevenção & controle , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , Pioglitazona , Células do Corno Posterior/efeitos dos fármacos , Células do Corno Posterior/metabolismo , Ratos , Tiazolidinedionas/administração & dosagem , Tiazolidinedionas/antagonistas & inibidores , Tiazolidinedionas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Neuropharmacology ; 58(2): 337-45, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19891980

RESUMO

Systemic administration of thiazolidinediones reduces peripheral inflammation in vivo, presumably by acting at peroxisome proliferator-activated receptor gamma (PPARgamma) in peripheral tissues. Based on a rapidly growing body of literature indicating the CNS as a functional target of PPARgamma actions, we postulated that brain PPARgamma modulates peripheral edema and the processing of inflammatory pain signals in the dorsal horn of the spinal cord. To test this in the plantar carrageenan model of inflammatory pain, we measured paw edema, heat hyperalgesia, and dorsal horn expression of the immediate-early gene c-fos after intracerebroventricular (ICV) administration of PPARgamma ligands or vehicle. We found that ICV rosiglitazone (0.5-50 microg) or 15d-PGJ(2) (50-200 microg), but not vehicle, dose-dependently reduced paw thickness, paw volume and behavioral withdrawal responses to noxious heat. These anti-inflammatory and anti-hyperalgesia effects result from direct actions in the brain and not diffusion to other sites, because intraperitoneal and intrathecal administration of rosiglitazone (50 microg) and 15d-PGJ(2) (200 microg) had no effect. PPARgamma agonists changed neither overt behavior nor motor coordination, indicating that non-specific behavioral effects do not contribute to PPAR ligand-induced anti-hyperalgesia. ICV administration of structurally dissimilar PPARgamma antagonists (either GW9662 or BADGE) reversed the anti-inflammatory and anti-hyperalgesic actions of both rosiglitazone and 15d-PGJ(2). To evaluate the effects of PPARgamma agonists on a classic marker of noxious stimulus-evoked gene expression, we quantified Fos protein expression in the dorsal horn. The number of carrageenan-induced Fos-like immunoreactive profiles was less in rosiglitazone-treated rats as compared to vehicle controls. We conclude that pharmacological activation of PPARgamma in the brain rapidly inhibits local edema and the spinal transmission of noxious inflammatory signals.


Assuntos
Encéfalo/metabolismo , Inflamação/metabolismo , PPAR gama/metabolismo , Dor/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Medula Espinal/metabolismo , Anilidas/farmacologia , Animais , Compostos Benzidrílicos , Encéfalo/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/farmacologia , Modelos Animais de Doenças , Edema/tratamento farmacológico , Edema/genética , Edema/metabolismo , Compostos de Epóxi/farmacologia , Expressão Gênica/efeitos dos fármacos , Inflamação/tratamento farmacológico , Inflamação/genética , Masculino , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , Dor/tratamento farmacológico , Dor/etiologia , Dor/genética , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacologia , Proteínas Proto-Oncogênicas c-fos/genética , Ratos , Ratos Sprague-Dawley , Rosiglitazona , Medula Espinal/efeitos dos fármacos , Tiazolidinedionas/farmacologia
5.
Brain Res ; 897(1-2): 131-8, 2001 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-11282366

RESUMO

The present study investigated the effect of lesions of the anterior cingulate cortex (ACC) on mechanical allodynia/hyperalgesia after L5 ligation or on inflammatory nociceptive responses following formalin injection in the rat. For both the neuropathic and inflammatory pain models, three groups of animals were used. The control groups consisted of a group of sham lesioned animals and a group of animals that had unilateral damage to the ACC or unilateral/bilateral damage to surrounding cortical tissue. The third group consisted of animals that had at least 75% bilateral damage of the ACC. Subjects received L5 ligation or a 0.05-ml injection of 1% formalin into the plantar surface of the hindpaw. In contrast to the control groups, bilateral ACC lesions significantly decreased inflammatory nociceptive responses during the prolonged, tonic portion of the formalin test (20-35 min). The difference between the groups was most prevalent in the amount of time spent licking the paw. However, ACC lesions did not significantly attenuate the enhanced mechanical paw withdrawal threshold in the neuropathic nociceptive model. These results suggest a differential role of the ACC in the modulation of different types of pain conditions.


Assuntos
Giro do Cíngulo/imunologia , Giro do Cíngulo/fisiopatologia , Hiperalgesia/imunologia , Hiperalgesia/fisiopatologia , Nociceptores/imunologia , Doença Aguda , Animais , Comportamento Animal , Doença Crônica , Denervação , Ligadura , Masculino , Inflamação Neurogênica/imunologia , Inflamação Neurogênica/fisiopatologia , Medição da Dor , Ratos , Ratos Sprague-Dawley , Tempo de Reação , Nervos Espinhais/imunologia , Nervos Espinhais/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA