Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 657(1-3): 89-96, 2011 Apr 25.
Article in English | MEDLINE | ID: mdl-21300053

ABSTRACT

The long-lasting post-surgical changes in nociceptive thresholds in mice, indicative of latent pain sensitization, were studied. The contribution of kappa opioid and N-methyl-d-aspartate (NMDA) receptors was assessed by the administration of nor-binaltorphimine or MK-801; dynorphin levels in the spinal cord were also determined. Animals underwent a plantar incision and/or a subcutaneous infusion of remifentanil (80µg/kg), and mechanical thresholds (von Frey) were evaluated at different times. On day 21, after complete recovery of mechanical thresholds and healing of the wound, one of the following drugs was administered subcutaneously: (-)-naloxone (1mg/kg), (+)-naloxone (1mg/kg), naloxone-methiodide (3mg/kg), or nor-binaltorphimine (5mg/kg). Another group received subcutaneous MK-801 (0.15mg/kg) before nor-binaltorphimine administration. Dynorphin on day 21 was determined in the spinal cord by immunoassay. In mice receiving remifentanil during surgery, the administration of (-)-naloxone or nor-binaltorphimine induced significant hyperalgesia even 5months after manipulation. Nociceptive thresholds remained unaltered after (+)-naloxone or naloxone-methiodide. On day 21 after manipulation, the administration of MK-801 prevented nor-binaltorphimine-induced hyperalgesia. No changes in dynorphin levels were observed before or after opioid antagonist administration. In conclusion, surgery produced latent pain sensitization evidenced by opioid antagonist-precipitated hyperalgesia. The effect was stereospecific, centrally originated, and mediated by kappa opioid receptors. The blockade of nor-binaltorphimine-induced hyperalgesia by MK-801, suggests that NMDA receptors are also involved. Our results show for the first time that surgery induces latent, long-lasting changes in the processing of nociceptive information that can be induced by non-nociceptive stimuli such as the administration of opioid antagonists.


Subject(s)
Narcotic Antagonists/administration & dosage , Narcotic Antagonists/pharmacology , Pain Threshold/drug effects , Pain/physiopathology , Postoperative Complications/physiopathology , Reaction Time/drug effects , Animals , Dizocilpine Maleate/administration & dosage , Dizocilpine Maleate/pharmacology , Dynorphins/metabolism , Dynorphins/pharmacology , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Male , Mice , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Pain/chemically induced , Pain/metabolism , Pain Threshold/physiology , Piperidines/administration & dosage , Piperidines/pharmacology , Postoperative Complications/chemically induced , Postoperative Complications/metabolism , Reaction Time/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Opioid, kappa/metabolism , Remifentanil , Spinal Cord/drug effects , Spinal Cord/metabolism
2.
Electron. j. biotechnol ; 13(5): 19-20, Sept. 2010. ilus, tab
Article in English | LILACS | ID: lil-591901

ABSTRACT

Nucleic-acid based methods for bacterial identification are extremely useful in diagnostic applications due to their specificity and sensitivity. However, they require an optimal purification of the target molecules. As part of the development of a new diagnostic method for the detection of bacterial RNA in cow milk, we have compared four commercially available RNA extraction kits for the isolation of bacterial RNA from spiked UHT milk samples. The kits were compared in terms of extraction efficiency and RNA purity using two bacterial species, the Gram negative Escherichia coli and the Gram positive Staphylococcus aureus. Two kits are based in silica-matrix extraction, and the other two in the guanidinium thiocyanate-phenol-chloroform extraction. In our hands, the RNeasy Protect Bacteria Mini kit from QIAGEN was the best performing in terms of RNA yield, quality, reproducibility and consumable needs, under the conditions here described.


Subject(s)
RNA, Bacterial/isolation & purification , Escherichia coli/isolation & purification , Milk/microbiology , Staphylococcus aureus/isolation & purification
3.
Mol Pharmacol ; 77(2): 185-94, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19917879

ABSTRACT

In humans, remifentanil anesthesia enhances nociceptive sensitization in the postoperative period. We hypothesized that activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) and the expression of c-Fos, prodynorphin (mRNA), and dynorphin in the spinal cord could participate in the molecular mechanisms underlying postoperative opioid-induced sensitization. In a mouse model of incisional pain, we evaluated thermal (Hargreaves test) and mechanical (von Frey) hyperalgesia during the first 21 postoperative days. Moreover, prodynorphin (mRNA, real-time polymerase chain reaction), dynorphin (enzymatic immunoassay), c-Fos expression, and ERK1/2 phosphorylation (both by immunohistochemistry) in the lumbar spinal cord were assessed. Surgery performed under remifentanil anesthesia induced a maximal decrease in nociceptive thresholds between 4 h and 2 days postoperatively (p < 0.001) that lasted 10 to 14 days compared with noninjured animals. In the same experimental conditions, a significant increase in prodynorphin mRNA expression (at 2 and 4 days) followed by a sustained increase of dynorphin (days 2 to 10) in the spinal cord was observed. We also identified an early expression of c-Fos immunoreactivity in the superficial laminae of the dorsal horn of the spinal cord (peak at 4 h; p < 0.001), together with a partial activation of ERK1/2 (4 h; p < 0.001). These findings suggest that activated ERK1/2 could induce c-Fos expression and trigger the transcription of prodynorphin in the spinal cord. This in turn would result in long-lasting increased levels of dynorphin that, in our model, could participate in the persistence of pain but not in the manifestation of first pain.


Subject(s)
Dynorphins/biosynthesis , Genes, fos/physiology , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Pain, Postoperative/metabolism , Piperidines/therapeutic use , Anesthetics, Intravenous/pharmacology , Anesthetics, Intravenous/therapeutic use , Animals , Male , Mice , Pain Measurement/drug effects , Pain Measurement/methods , Pain, Postoperative/prevention & control , Piperidines/pharmacology , Remifentanil , Spinal Cord/drug effects , Spinal Cord/metabolism
4.
Fundam Clin Pharmacol ; 24(3): 317-23, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19751225

ABSTRACT

The implication of inducible nitric-oxide synthase (iNOS) on peripheral tolerance to morphine was evaluated in wild-type (WT) and iNOS knockout mice. Chronic inflammation was induced by subplantar (s.p.) injection of Complete Freund's Adjuvant (CFA), and morphine tolerance by subcutaneous implantation of a 75 mg morphine-pellet. Withdrawal was assessed after the intraperitoneal injection of 2 mg/kg naloxone. Antinociception was assessed (Randall-Selitto test) 5 min after a fixed dose of s.p. morphine (16 microg). In the absence of inflammation, s.p. morphine did not induce antinociception, while during CFA-inflammation produced 47.4 +/- 0.8 and 38.8 +/- 2.7% inhibitions respectively, in each genotype (P < 0.05). In morphine-tolerant mice with CFA-inflammation, no antinociception could be elicited in WT mice (2.4 +/- 0.3% inhibition); however, iNOS knockout mice showed significant antinociception (33.1 +/- 0.9%) (P < 0.001). Thus, iNOS gene deletion partially prevented tolerance to the peripheral effects of morphine, and significantly attenuated withdrawal-induced hyperactivity.


Subject(s)
Arthritis, Experimental/enzymology , Disease Models, Animal , Drug Tolerance/physiology , Gene Deletion , Morphine/administration & dosage , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Chronic Disease , Inflammation/chemically induced , Inflammation/enzymology , Inflammation/genetics , Inflammation Mediators/administration & dosage , Inflammation Mediators/adverse effects , Inflammation Mediators/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/adverse effects , Naloxone/administration & dosage , Nitric Oxide Synthase Type II/physiology , Pain Measurement/drug effects , Pain Measurement/methods , Substance Withdrawal Syndrome/enzymology , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/pathology
5.
Brain Res ; 1248: 31-9, 2009 Jan 12.
Article in English | MEDLINE | ID: mdl-19026993

ABSTRACT

Tolerance to peripheral antinociception after chronic exposure to systemic morphine was assessed in mice with chronic CFA-inflammation; cross-tolerance to locally administered mu, delta and kappa-opioid agonists and levels of beta-arrestins in the injured paw, were also evaluated. Tolerance was induced by the subcutaneous implantation of a 75 mg morphine-pellet, and antinociception evaluated with the Randall-Selitto test, 5 min after the subplantar injection of morphine, fentanyl, buprenorphine, DPDPE, U-50488H or CRF. Experiments were performed in the absence and presence of CFA-inflammation, in animals implanted with a morphine or placebo pellet. Beta-arrestin protein levels were determined by western blot. In mice without inflammation, subplantar opioids did not induce antinociception, while during CFA-inflammation, all drugs generated dose-response curves with an order of potency of: U-50488H < DPDPE < morphine < buprenorphine < fentanyl << CRF. During CFA-inflammation plus morphine-pellet, the potency of fentanyl decreased 1.25 times, while that of DPDPE, U-50488H and CRF diminished approximately 2.5-4.3 times. For each drug, the ratio between the ED(50)'s in tolerant and naive animals, was significantly higher than 1 (except for buprenorphine and fentanyl), demonstrating partial cross-tolerance to systemic morphine. Inflammation induced a twofold increase in beta-arrestin expression (p<0.01), and the levels decreased after acute morphine exposure (p<0.05). Tolerance did not alter beta-arrestins, but partially prevented the increase induced by inflammation. The results suggest that peripheral beta-arrestins could facilitate peripheral OR-desensitization and tolerance development. Clinically, the experiments could be useful to establish the effectiveness of local opioid administration in patients with musculoskeletal pain, chronically receiving morphine analgesia.


Subject(s)
Analgesics, Opioid/administration & dosage , Arrestins/metabolism , Pain/drug therapy , Analgesics, Opioid/pharmacology , Animals , Blotting, Western , Dose-Response Relationship, Drug , Drug Tolerance , Extremities , Inflammation/physiopathology , Male , Mice , Morphine/administration & dosage , Morphine/pharmacology , Pain Measurement , Random Allocation , Receptors, Opioid/metabolism , beta-Arrestins
6.
Naunyn Schmiedebergs Arch Pharmacol ; 379(3): 315-23, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18839150

ABSTRACT

Opioids are used in humans in the treatment of chronic osteoarticular pain, but the development of tolerance to the analgesic effects after continuous administration is still not well understood. The aim of the present study was to evaluate the expression of phospho-ERK 1/2 and phospho-p38 in mice with monoarthritis chronically exposed to morphine as a possible explanation for the development of tolerance. Inflammation was induced by intraplantar injection of complete Freund's adjuvant (CFA) and the tolerance by implantation of 75 mg morphine pellets. The results of the present study show that ERKs phosphorylation is unaltered by inflammation or morphine tolerance, each one individually, in the plantar tissue. In contrast, phospho-p38 is similarly decreased by inflammation or morphine tolerance. In naïve but not in tolerant animals, acute injection of morphine induces significant increase in phospho-p38 without any changes in phospho-ERK 1/2 expression. During inflammation, the acute injection of morphine induces a significant increase in the expression of ERK 1/2, but not in phospho-p38, in naïve animals. Phospho-ERK 1/2 expression was significantly decreased in the presence of inflammation plus tolerance. In contrast, no significant differences in phospho-p38 expression were observed between naïve and tolerant animals acutely injected with saline or morphine in presence of CFA inflammation. These results suggest that ERK but not p38 could be implicated in the development of morphine tolerance during peripheral inflammation. These experiments could contribute to establish the mechanisms implicated in the development of morphine tolerance in presence of inflammatory pain.


Subject(s)
Analgesics, Opioid/therapeutic use , Arthritis, Experimental/drug therapy , Drug Tolerance , Mitogen-Activated Protein Kinase 1/biosynthesis , Morphine/therapeutic use , p38 Mitogen-Activated Protein Kinases/biosynthesis , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Arthritis, Experimental/enzymology , Blotting, Western , Chronic Disease , Dose-Response Relationship, Drug , Freund's Adjuvant , Male , Mice , Morphine/administration & dosage , Morphine/pharmacology , Pain Threshold/drug effects , Phosphorylation
7.
Pain ; 141(1-2): 88-96, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19058913

ABSTRACT

The ultra-short-acting mu-opioid receptor (MOR) agonist remifentanil enhances postsurgical pain when used as main anesthetic in animal models and man. Although the mechanism/s involved are poorly characterized, changes in opioid receptor expression could be a relevant feature. Using a mouse model of postoperative pain, we assessed the expression of MOR and delta opioid receptors (DORs) and the efficacy of Herpes Simplex vector-mediated proenkephalin release (SHPE) preventing postoperative nociceptive sensitization induced by remifentanil or surgical incision. We determined MOR and DOR expressions in the dorsal root ganglia and the spinal cord after remifentanil or surgery in CD1 mice, using real-time PCR and Western blotting. We also assessed the effect of SHPE on nociception induced by remifentanil, surgery, and their combination (2 and 7 days after manipulation), using thermal and mechanical tests. Both remifentanil and surgery decreased DOR mRNA levels (up to days 2 and 4, respectively) in the dorsal root ganglia, but not in the spinal cord. No changes were observed in MOR mRNA, or in receptor-protein levels (Western) of either receptor. Pre-treatment with SHPE 7 days before manipulation prevented remifentanil-induced thermal hyperalgesia and mechanical allodynia and the increase in incisional pain observed when surgery was performed under remifentanil anesthesia. SHPE also prevented surgically induced allodynia but not hyperalgesia, which was blocked by the additional administration of RB101, an enkephalinase inhibitor. The study suggests that down-regulation of DOR contributes to remifentanil and surgery-induced nociception, and that postoperative pain is completely reversed by increasing enkephalin levels in the spinal cord and the periphery.


Subject(s)
Down-Regulation/physiology , Enkephalins/metabolism , Pain, Postoperative/metabolism , Pain, Postoperative/prevention & control , Protein Precursors/metabolism , RNA, Messenger/metabolism , Receptors, Opioid, delta/genetics , Analgesics/administration & dosage , Analgesics/adverse effects , Animals , Disease Models, Animal , Disulfides/administration & dosage , Down-Regulation/drug effects , Enkephalins/genetics , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Male , Methyl Ethers/administration & dosage , Mice , Pain Measurement/drug effects , Pain Measurement/methods , Pain Threshold , Pain, Postoperative/etiology , Pain, Postoperative/pathology , Phenylalanine/administration & dosage , Phenylalanine/analogs & derivatives , Piperidines/adverse effects , Protein Precursors/genetics , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Remifentanil , Sevoflurane , Spinal Cord/drug effects , Spinal Cord/metabolism , Time Factors
8.
J Pharmacol Exp Ther ; 322(1): 360-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17468301

ABSTRACT

Opioids are used in humans in the management of chronic osteoarticular pains, but the development of tolerance to the analgesic effects after continuous administration is still not well understood. Our aim was to characterize morphine tolerance in a murine model of arthritis that mimics the sequence of events occurring in humans. Inflammation was induced by the intraplantar injection of complete Freund's adjuvant (CFA) and tolerance by the implantation of a 75-mg morphine pellet. We assessed the antihyperalgesic (plantar and Randall-Selitto tests), antiallodynic (Von Frey test), and antiexudative (Evans blue) effects of morphine, the mu-opioid receptor (MOR) mRNA levels in dorsal root ganglia (DRG), and MOR protein levels in DRG and plantar tissue. Inflammation induced plasma extravasation, and it significantly increased the antihyperalgesic effects of morphine (p < 0.05). Morphine pellet implantation decreased morphine potency in all tests. ED(50) values decreased 4.4 and 7.3 times in the absence and presence of inflammation in the plantar test and 2.7 and 5.3 times in the Randall-Selitto test, whereas plasma extravasation decreased 4.2 times. MOR mRNA levels in the DRG were not affected 7 days after inflammation, whereas chronic morphine administration induced a discrete increase (p < 0.05). MOR protein in the DRG or the paw was unchanged. The results show that inflammation enhances the development of tolerance to the antihyperalgesic and antiexudative effects of morphine. At the molecular level, our results suggest that these effects are not mediated by changes in MOR expression but by other changes in receptor activation/internalization.


Subject(s)
Analgesics, Opioid/therapeutic use , Arthritis, Experimental/drug therapy , Exudates and Transudates/drug effects , Morphine/therapeutic use , Animals , Dose-Response Relationship, Drug , Drug Tolerance , Freund's Adjuvant , Ganglia, Spinal/metabolism , Male , Mice , Morphine/blood , Naloxone/analogs & derivatives , Naloxone/pharmacology , Quaternary Ammonium Compounds/pharmacology , RNA, Messenger/analysis
9.
J Neurosci Res ; 79(1-2): 166-71, 2005.
Article in English | MEDLINE | ID: mdl-15573411

ABSTRACT

There is an increasing body of evidence demonstrating that inhibition of cytochrome c oxidase by nitric oxide (NO) may be one more step in a signaling cascade involved in the physiologic regulation of cell functions. For example, in both astrocytes and neurons the inhibition of mitochondrial respiration by endogenously produced NO induces transient and modest decreases in cellular ATP concentrations. This mitochondrial impairment may serve as a cellular sensor of energy charges, hence modulating metabolic pathways, such as glycolysis, through AMP-activated protein kinase (AMPK) in astrocytes. In neurons, the NO derivative peroxynitrite anion triggers signaling pathways leading to glucose oxidation through the pentose-phosphate pathway to form reducing equivalents in the form of NADPH. The modulation of these metabolic pathways by nitric oxide or its derivatives may be important for understanding the mechanisms by which this free radical affects neuronal death or survival.


Subject(s)
Glucose/metabolism , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Nitric Oxide/physiology , Respiration/drug effects , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/physiology , Brain/cytology , Homeostasis/physiology , Humans , Mitochondria/metabolism , Models, Biological , Nitric Oxide/biosynthesis , Peroxynitrous Acid/pharmacology , Peroxynitrous Acid/physiology
10.
Mol Aspects Med ; 25(1-2): 61-73, 2004.
Article in English | MEDLINE | ID: mdl-15051317

ABSTRACT

Following brain inflammatory stimuli, astrocytes actively synthesize nitric oxide and peroxynitrite. These nitrogen-derived species trigger a repertoire of biochemical effects, including alteration of mitochondrial function and redox status both in astrocytes and neighboring neurons. Furthermore, under such nitrosative stress astrocytes show remarkable resistance in spite of having their mitochondria impaired, whereas the neighboring neurons show vulnerability. In this review, we discuss recent evidence strongly suggesting that nitrogen-derived species modulate key regulatory steps of glucose metabolism. These involve up-regulation of high-affinity glucose transporter, stimulation of glycolysis at 6-phosphofructo-1-kinase, and activation of pentose-phosphate pathway at glucose-6-phosphate dehydrogenase. We conclude that the orchestrated stimulation of glucose-metabolising pathways by nitric oxide would be a transient attempt of certain neural cells to compensate for the impaired energy status and oxidised glutathione and thus emerge from an otherwise neuropathological outcome.


Subject(s)
Glucose/metabolism , Nervous System Physiological Phenomena , Oxidative Stress/physiology , Animals , Humans , Nitric Oxide/physiology , Nitrosation , Pentose Phosphate Pathway/physiology , Peroxynitrous Acid/physiology
11.
Curr Pharm Des ; 10(8): 867-77, 2004.
Article in English | MEDLINE | ID: mdl-15032690

ABSTRACT

Peroxynitrite, the product of the reaction between nitric oxide and superoxide, is spontaneouly formed within most mammalian cells under physiological conditions. Initial work addressing the pathophysiology of peroxynitrite afforded the generally accepted notion that this compound would be the long-term neurotoxic nitric oxide-derivative. However, over the past six years a number of interesting studies have reported direct in vivo and in vitro evidence that, at nanomolar-low micromolar concentrations, peroxynitrite is actively involved in triggering cellular survival signals. Most such evidence came from studies demonstrating protection against myocardial ischemia-reperfusion injury and neuronal apoptosis. Although full elucidation of the precise mechanism responsible for such protection still requires further research, peroxynitrite has been shown to promote the nitration and/or phosphorylation of regulatory sites at tyrosine kinase receptors coupled to well-known antiapoptotic pathways, such as those involving phosphoinositide 3-kinase/Akt or mitogen-activated protein kinases. In addition, peroxynitrite-mediated transient protection of neurons against apoptotic death is associated with rapid stimulation of glucose metabolism and glutathione regeneration. In view of the potential cytoprotective function of peroxynitrite, further studies specifically focused on elucidating the possible therapeutic potential of peroxynitrite are sure to appear.


Subject(s)
Neurons/cytology , Peroxynitrous Acid/metabolism , Animals , Cell Death/physiology , Cell Survival/physiology , Cytoprotection/physiology , Glutathione/metabolism , Humans , Neurons/metabolism , Peroxynitrous Acid/biosynthesis , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Sulfhydryl Compounds/metabolism , Tyrosine/metabolism , Up-Regulation
12.
J Biol Chem ; 278(2): 864-74, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12414804

ABSTRACT

Peroxynitrite is thought to be a nitric oxide-derived neurotoxic effector molecule involved in the disruption of key energy-related metabolic targets. To assess the consequences of such interference in cellular glucose metabolism and viability, we studied the possible modulatory role played by peroxynitrite in glucose oxidation in neurons and astrocytes in primary culture. Here, we report that peroxynitrite triggered rapid stimulation of pentose phosphate pathway (PPP) activity and the accumulation of NADPH, an essential cofactor for glutathione regeneration. In contrast to peroxynitrite, nitric oxide elicited NADPH depletion, glutathione oxidation, and apoptotic cell death in neurons, but not in astrocytes. These events were noticeably counteracted by pretreatment of neurons with peroxynitrite. In an attempt to elucidate the mechanism responsible for this PPP stimulation and neuroprotection, we found evidence consistent with both exogenous and endogenous peroxynitrite-mediated activation of glucose-6-phosphate dehydrogenase (G6PD), an enzyme that catalyzes the first rate-limiting step in the PPP. Moreover, functional overexpression of the G6PD gene in stably transformed PC12 cells induced NADPH accumulation and offered remarkable resistance against nitric oxide-mediated apoptosis, whereas G6PD gene-targeted antisense inhibition depleted NADPH levels and exacerbated cellular vulnerability. In light of these results, we suggest that G6PD activation represents a novel role for peroxynitrite in neuroprotection against nitric oxide-mediated apoptosis.


Subject(s)
Apoptosis , Glucosephosphate Dehydrogenase/physiology , Neurons/pathology , Nitric Oxide/physiology , Peroxynitrous Acid/physiology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Cells, Cultured , Glucosephosphate Dehydrogenase/genetics , Glutathione/metabolism , NADP/analysis , Neurons/drug effects , Neuroprotective Agents/pharmacology , PC12 Cells , Pentose Phosphate Pathway , Peroxynitrous Acid/pharmacology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...