Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Neuropharmacology ; 70: 236-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23415633

ABSTRACT

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.


Subject(s)
Neuralgia/physiopathology , PPAR gamma/physiology , Thiazolidinediones/therapeutic use , Anilides/pharmacology , Animals , Ataxia/chemically induced , CD11b Antigen/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Extracellular Signal-Regulated MAP Kinases/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/physiopathology , Hyperalgesia/prevention & control , Male , Neuralgia/drug therapy , Neuralgia/prevention & control , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Pioglitazone , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Rats , Thiazolidinediones/administration & dosage , Thiazolidinediones/antagonists & inhibitors , Thiazolidinediones/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Neuropharmacology ; 58(2): 337-45, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19891980

ABSTRACT

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.


Subject(s)
Brain/metabolism , Inflammation/metabolism , PPAR gamma/metabolism , Pain/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Spinal Cord/metabolism , Anilides/pharmacology , Animals , Benzhydryl Compounds , Brain/drug effects , Central Nervous System Agents/pharmacology , Disease Models, Animal , Edema/drug therapy , Edema/genetics , Edema/metabolism , Epoxy Compounds/pharmacology , Gene Expression/drug effects , Inflammation/drug therapy , Inflammation/genetics , Male , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Pain/drug therapy , Pain/etiology , Pain/genetics , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Rats , Rats, Sprague-Dawley , Rosiglitazone , Spinal Cord/drug effects , Thiazolidinediones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...