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.
Brain Behav Immun ; 23(2): 240-50, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18938237

ABSTRACT

Morphine-induced glial proinflammatory responses have been documented to contribute to tolerance to opioid analgesia. Here, we examined whether drugs previously shown to suppress glial proinflammatory responses can alter other clinically relevant opioid effects; namely, withdrawal or acute analgesia. AV411 (ibudilast) and minocycline, drugs with distinct mechanisms of action that result in attenuation of glial proinflammatory responses, each reduced naloxone-precipitated withdrawal. Analysis of brain nuclei associated with opioid withdrawal revealed that morphine altered expression of glial activation markers, cytokines, chemokines, and a neurotrophic factor. AV411 attenuated many of these morphine-induced effects. AV411 also protected against spontaneous withdrawal-induced hyperactivity and weight loss recorded across a 12-day timecourse. Notably, in the spontaneous withdrawal study, AV411 treatment was delayed relative to the start of the morphine regimen so to also test whether AV411 could still be effective in the face of established morphine dependence, which it was. AV411 did not simply attenuate all opioid effects, as co-administering AV411 with morphine or oxycodone caused three-to-five-fold increases in acute analgesic potency, as revealed by leftward shifts in the analgesic dose response curves. Timecourse analyses revealed that plasma morphine levels were not altered by AV411, suggestive that potentiated analgesia was not simply due to prolongation of morphine exposure or increased plasma concentrations. These data support and extend similar potentiation of acute opioid analgesia by minocycline, again providing converging lines of evidence of glial involvement. Hence, suppression of glial proinflammatory responses can significantly reduce opioid withdrawal, while improving analgesia.


Subject(s)
Analgesia/psychology , Analgesics, Opioid/adverse effects , Brain/metabolism , Pyridines/pharmacology , Substance Withdrawal Syndrome/physiopathology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Brain/drug effects , Brain/immunology , Bronchodilator Agents/administration & dosage , Bronchodilator Agents/pharmacology , Dose-Response Relationship, Drug , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Injections, Intraperitoneal , Male , Minocycline/administration & dosage , Minocycline/pharmacology , Morphine/adverse effects , Naloxone/adverse effects , Opioid-Related Disorders/etiology , Opioid-Related Disorders/metabolism , Opioid-Related Disorders/physiopathology , Opioid-Related Disorders/psychology , Oxycodone/adverse effects , Pain/physiopathology , Pain/psychology , Pain Measurement , Pyridines/administration & dosage , Rats , Rats, Sprague-Dawley , Substance Withdrawal Syndrome/etiology , Substance Withdrawal Syndrome/metabolism , Weight Loss/drug effects
2.
Brain Behav Immun ; 22(8): 1248-56, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18706994

ABSTRACT

Recent data suggest that opioids can activate immune-like cells of the central nervous system (glia). This opioid-induced glial activation is associated with decreased analgesia, owing to the release of proinflammatory mediators. Here, we examine in rats whether the putative microglial inhibitor, minocycline, may affect morphine-induced respiratory depression and/or morphine-induced reward (conditioned place preference). Systemic co-administration of minocycline significantly attenuated morphine-induced reductions in tidal volume, minute volume, inspiratory force, and expiratory force, but did not affect morphine-induced reductions in respiratory rate. Minocycline attenuation of respiratory depression was also paralleled with significant attenuation by minocycline of morphine-induced reductions in blood oxygen saturation. Minocycline also attenuated morphine conditioned place preference. Minocycline did not simply reduce all actions of morphine, as morphine analgesia was significantly potentiated by minocycline co-administration. Lastly, morphine dose-dependently increased cyclooxygenase-1 gene expression in a rat microglial cell line, an effect that was dose-dependently blocked by minocycline. Together, these data support that morphine can directly activate microglia in a minocycline-suppressible manner and suggest a pivotal role for minocycline-sensitive processes in the mechanisms of morphine-induced respiration depression, reward, and pain modulation.


Subject(s)
Analgesia , Minocycline/pharmacology , Morphine/pharmacology , Respiratory Insufficiency/drug therapy , Reward , Analysis of Variance , Animals , Cell Line , Cells, Cultured , Conditioning, Operant/drug effects , Cyclooxygenase 1/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Male , Microglia/drug effects , Microglia/metabolism , Minocycline/therapeutic use , Narcotics/pharmacology , Pain/drug therapy , Pain Measurement , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Respiratory Insufficiency/chemically induced , Reverse Transcriptase Polymerase Chain Reaction , Spatial Behavior/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...