Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Publication year range
1.
Eur J Pain ; 15(2): 169.e1-169.e12, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20594879

ABSTRACT

The N-methyl-d-aspartate receptor (NMDAR) contributes to central sensitization in the spinal cord, a phenomenon which comprises various pathophysiological mechanisms responsible for neuropathic pain-like signs in animal models. NMDAR function is modulated by post-translational modifications including phosphorylation, and this is proposed to underlie its involvement in the production of pain hypersensitivity. As in diabetic patients, streptozotocin-induced diabetic rats exhibit or not somatic mechanical hyperalgesia; these rats were named DH and DNH respectively. At three weeks of diabetes, we present evidence that somatic mechanical hyperalgesia was correlated with an enhanced phosphorylation of the NMDAR NR1 subunit (pNR1) in the rat spinal cord. This increase was not found in normal and DNH rats, suggesting that this regulation was specific to hyperalgesia. Double immunofluorescence studies revealed that the numbers of pNR1-immunoreactive neurons and microglial cells were significantly increased in all laminae (I-II and III-VI) of the dorsal horn from DH animals. Western-blots analysis showed no change in NR1 protein levels, whatever the behavioural and glycemic status of the animals. Chronic intrathecal treatment (5µg/rat/day for 7days) by U0126 and MK801, which blocked MEK (an upstream kinase of extracellular signal-regulated protein kinase: ERK) and the NMDAR respectively, simultaneously suppressed somatic mechanical hyperalgesia developed by diabetic rats and decreased pNR1. These results indicate for the first time that increased expression of pNR1 is regulated by ERK and the NMDAR via a feedforward mechanism in spinal neurons and microglia and represents one mechanism involved in central sensitization and somatic mechanical hyperalgesia after diabetes.


Subject(s)
Diabetic Neuropathies/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Microglia/metabolism , Phosphorylation/physiology , Posterior Horn Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/metabolism , Analysis of Variance , Animals , Blotting, Western , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Enzyme Inhibitors/pharmacology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Immunohistochemistry , Male , Microglia/drug effects , Phosphorylation/drug effects , Posterior Horn Cells/drug effects , Posterior Horn Cells/physiopathology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/drug effects , Spinal Cord/physiopathology
2.
J Pharmacol Exp Ther ; 322(3): 1237-45, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17551093

ABSTRACT

Agmatine, an endogenous cationic amine resulting from the decarboxylation of L-arginine, produces antihyperalgesic and antiallodynic effects in animal models of chronic neuropathic and inflammatory pain. We examined the effect of agmatine on tactile and thermal allodynia and on mechanical hyperalgesia in streptozocin-induced diabetic rats. To determine its mechanism of action and the potential interest of some of its combinations, the antihyperalgesic effect of agmatine was challenged with alpha(2)-adrenergic imidazoline and opioid-receptor antagonists, and its interaction with the opioid-receptor agonist morphine, the competitive N-methyl-D-aspartate receptor antagonist D-CPP [R(-)-3-(2-carboxypiperazine-4-yl)-propyl-1-phosphonic acid], and the nitric-oxide synthase inhibitor L-NAME (L-N(G)-nitro-L-arginine methyl ester) were examined. When intrathecally (i.t.) injected (4.4 to 438 nmol/rat), agmatine was ineffective in normal rats but suppressed tactile allodynia (von Frey hair test), thermal allodynia (tail immersion test), and mechanical hyperalgesia (paw-pressure test) in diabetic rats. This spinal antihyperalgesic effect was suppressed by idazoxan (40 micromol/rat i.t.) but not by yohimbine (40 micromol/rat i.t.) or naloxone (0.69 micromol/rat i.v.). In diabetic rats, an isobolographic analysis showed that combinations of i.t. agmatine with i.v. L-NAME or with i.t. morphine resulted in an additive antihyperalgesic effect, whereas the agmatine/D-CPP i.t. combination was superadditive. In summary, the present findings reveal that spinal agmatine produces antiallodynic and antihyperalgesic effects in diabetic neuropathic pain involving, at least for its antihyperalgesic effect, the imidazoline receptors. Moreover, agmatine combined with D-CPP produces an antinociceptive synergy in experimental neuropathy, opening opportunities in the development of new strategies for pain therapy.


Subject(s)
Agmatine/pharmacology , Hyperalgesia/drug therapy , Pain/drug therapy , Piperazines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Agmatine/therapeutic use , Animals , Anticonvulsants/pharmacology , Diabetes Mellitus, Experimental/complications , Drug Interactions , Excitatory Amino Acid Antagonists/pharmacology , Pain/etiology , Rats , Streptozocin
4.
Mol Pharmacol ; 70(4): 1246-54, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16868181

ABSTRACT

Molecular mechanisms underlying diabetes-induced painful neuropathy are poorly understood. We have demonstrated, in rats with streptozotocin-induced diabetes, that mechanical hyperalgesia, a common symptom of diabetic neuropathy, was correlated with an early increase in extracellular signal-regulated protein kinase (ERK), p38, and c-Jun N-terminal kinase (JNK) phosphorylation in the spinal cord and dorsal root ganglion at 3 weeks after induction of diabetes. This change was specific to hyperalgesia because nonhyperalgesic rats failed to have such an increase. Immunoblot analysis showed no variation of protein levels, suggesting a post-translational regulation of the corresponding kinases. In diabetic hyperalgesic rats, immunocytochemistry revealed that all phosphorylated mitogen-activated protein kinases (MAPKs) colocalized with both the neuronal (NeuN) and microglial (OX42) cell-specific markers but not with the astrocyte marker [glial fibrillary acidic protein (GFAP)] in the superficial dorsal horn-laminae of the spinal cord. In these same rats, a 7-day administration [5 microg/rat/day, intrathecal (i.t.)] of 1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)butadiene (U0126), 4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole (SB203580), and anthra(1,9-cd)pyrazol-6(2H)-one (SP600125), which inhibited MAPK kinase, p38, and JNK, respectively, suppressed mechanical hyperalgesia, and decreased phosphorylation of the kinases. To characterize the cellular events upstream of MAPKs, we have examined the role of the NMDA receptor known to be implicated in pain hypersensitivity. The prolonged blockade of this receptor during 7 days by (5R, 10S)-(+)-5-methyl-10, 11-dihydro-5H-dibenzo[a,d]-cyclohepten-5-10-imine hydrogen maleate (MK801; 5 microg/rat/day, i.t.), a noncompetitive NMDA receptor antagonist, reversed hyperalgesia developed by diabetic rats and blocked phosphorylation of all MAPKs. These results demonstrate for the first time that NMDA receptor-dependent phosphorylation of MAPKs in spinal cord neurons and microglia contribute to the establishment and longterm maintenance of painful diabetic hyperalgesia and that these kinases represent potential targets for pain therapy.


Subject(s)
Diabetes Mellitus, Experimental/complications , Hyperalgesia/etiology , Microglia/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Enzyme Activation , Hyperalgesia/metabolism , MAP Kinase Signaling System , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation , Rats , Rats, Sprague-Dawley , Streptozocin
5.
Pain ; 110(1-2): 236-45, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15275773

ABSTRACT

Nociceptin/orphanin FQ (noci/OFQ), the endogenous ligand for the orphan ORL1 (opioid receptor-like1), has been shown to be anti- or pronociceptive and modify morphine analgesia in rats after central administration. We comparatively examined the effect of noci/OFQ on hyperalgesia and morphine analgesia in two experimental models of neuropathic pain: diabetic (D) and mononeuropathic (MN) rats. Noci/OFQ, when intrathecally (i.t.) injected (0.1, 0.3, or 1, to 10 microg/rat) was ineffective in normal rats, but reduced and suppressed mechanical hyperalgesia (paw-pressure test) in D and MN rats, respectively. This spinal inhibitory effect was suppressed by naloxone (10 microg/rat, i.t.) in both models. Combinations of systemic morphine with spinal noci/OFQ resulted in a strong potentiation of analgesia in D rats. In MN rats, an isobolographic analysis showed that the morphine+noci/OFQ association (i.t.) suppressed mechanical hyperalgesia in a superadditive manner. In summary, the present findings reveal that spinal noci/OFQ produces a differential antinociception in diabetic and traumatic neuropathic pain according to the etiology of neuropathy, an effect possibly mediated by opioid receptors. Moreover, noci/OFQ combined with morphine produces antinociceptive synergy in experimental neuropathy, opening new opportunities in the treatment of neuropathic pain.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Mononeuropathies/physiopathology , Opioid Peptides/physiology , Pain/physiopathology , Receptors, Opioid/metabolism , Animals , Behavior, Animal , Body Weight , Diabetes Mellitus, Experimental/chemically induced , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Hyperalgesia/physiopathology , Male , Mononeuropathies/metabolism , Morphine/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pain/metabolism , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Receptors, Opioid/agonists , Sciatic Nerve/injuries , Streptozocin , Time Factors , Vocalization, Animal/drug effects , Nociceptin Receptor , Nociceptin
6.
Neurotoxicology ; 24(6): 797-805, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14637374

ABSTRACT

Using doses close to those used clinically, we have developed an animal model of vincristine-induced nociceptive sensory neuropathy after repeated intravenous injection in male Sprague-Dawley rats. In order to validate the model, three different doses (50, 100 and 150 microg/kg) of vincristine were injected every 2nd day until five injections had been given. The sensory behavioural assessment revealed mechanical hyperalgesia and allodynia associated with cold thermal hyperalgesia and allodynia. With regard to electrophysiological evaluation, we observed a decrease in the nerve conduction velocity in the highest dose group. Morphological studies revealed few degenerated fibers in the sciatic nerve and many degenerated myelinated axons in the fine nerve fibers of the subcutaneous paw tissue. Finally, to develop an animal model, we chose the 150 microg/kg dose because of the good general clinical status of the rats without motor function changes associated with severe sensation disorders like hyperalgesia and allodynia. This model of vincristine-induced painful neuropathy will be used to explore physiopathological mechanisms implied in the genesis of neuropathic pain and also to test new analgesic and neuroprotective drugs.


Subject(s)
Disease Models, Animal , Pain Measurement/drug effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/pathology , Vincristine/toxicity , Animals , Male , Pain Measurement/methods , Rats
7.
Exp Neurol ; 182(1): 12-20, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12821373

ABSTRACT

We report the assessment of motor and sensory behaviors using an electrophysiologic and an histologic approach, in a rat model of cisplatin peripheral neuropathy. Cisplatin was injected intraperitoneally one (3 mg/ kg), two (2 mg/kg), or three (1 mg/kg) times a week up to a cumulative dose of 15 or 20 mg/kg. With regard to nociceptive signs, we observed mechanical and thermal (cold stimuli) hyperalgesia and allodynia associated with minor motor disorders for the 3 mg/kg dose. Peripheral nerve conduction velocities were decreased in the cisplatin-(3 mg/kg) treated group. In addition, the histologic approach revealed that large axons were more frequently affected than the small ones, and nonmyelinated axons were unaffected. However, even in the most severe cases, myelin sheaths remained within normal limits. This animal model of nociceptive neuropathy would be suitable to study the pathophysiologic mechanisms of neuropathic pain and to test potential neuroprotective agents.


Subject(s)
Antineoplastic Agents , Cisplatin , Pain/chemically induced , Peripheral Nervous System Diseases/chemically induced , Animals , Axons/pathology , Behavior, Animal/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Electrophysiology , Hand Strength , Hot Temperature , In Vitro Techniques , Lumbosacral Region , Male , Neural Conduction/drug effects , Pain/complications , Pain/physiopathology , Pain Measurement , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/physiopathology , Rats , Rats, Sprague-Dawley , Reaction Time , Sciatic Nerve/pathology , Sciatic Nerve/physiopathology , Spinal Cord/pathology
8.
Pain ; 88(1): 15-22, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11098095

ABSTRACT

Since evidence points to the involvement of cholecystokinin (CCK) in nociception, we examined the effect of intrathecal CI-988, an antagonist of the CCK-B receptors, on mechanical hyperalgesia and allodynia in normal, mononeuropathic and diabetic rats,. Owing to the anti-opioid activity of CCK, it has been suggested that hyperactivity in the spinal CCK system is responsible for the low sensitivity of neuropathic pain to opioids. We therefore also evaluated the effect of the combination of i.t. CI-988 + i.v. morphine on mechanical hyperalgesia in diabetic and mononeuropathic rats using isobolographic analysis. Although ineffective in normal rats, CI-988 induced antinociceptive effects in diabetic (290 +/- 20 g with a cut-off of 750 g) and mononeuropathic (117 +/- 16 g; cut-off 750 g) rats, suggesting an involvement of the CCKergic system in neurogenic pain conditions. The combination of CI-988 and morphine showed a superadditive interaction in the diabetic rats only (477 +/- 16 g; cut-off 750 g), in comparison with the antinociceptive effect of each drug. In addition, CI-988 exhibited a weak anti-allodynic effect in mononeuropathic rats, and no anti-allodynic effect in diabetic rats. These results show the CCK-B receptor blockade-mediated antinociceptive effects and reveals the antinociceptive action of morphine in diabetic rats after CCKergic system inhibition.


Subject(s)
Analgesia , Analgesics, Opioid/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Hormone Antagonists/pharmacology , Hyperalgesia/physiopathology , Indoles/pharmacology , Meglumine/analogs & derivatives , Meglumine/pharmacology , Morphine/pharmacology , Nervous System Diseases/physiopathology , Pain Threshold/drug effects , Receptors, Cholecystokinin/antagonists & inhibitors , Animals , Drug Synergism , Injections, Intravenous , Injections, Spinal , Male , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin B , Touch
SELECTION OF CITATIONS
SEARCH DETAIL