Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmacol Biochem Behav ; 134: 49-56, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25902407

ABSTRACT

The pathophysiology of chronic inflammatory pain remains poorly understood. In this context, we developed an experimental model in which successive daily injection of prostaglandin E2 (PGE2) for 14days into rat hind paws produces a persistent state of hypernociception (i.e. decrease in mechanical nociceptive threshold). This state persists for more than 30days after discontinuing PGE2 injection. In the present study, we investigated the participation of nuclear factor kappa B (NF-κB), in the maintenance of this process. Mechanical hypernociception was evaluated using the electronic von Frey test. Activation of NF-κB signaling was measured through the determination of NF-κB p65 subunit translocation to the nucleus of dorsal root ganglion neurons (DRG) by immunofluorescence and western blotting. Herein, we detected an increase in NF-κB p65 subunit translocation to the nucleus of DRG neurons along with persistent inflammatory hypernociception compared with controls. Intrathecal treatment with either dexamethasone or PDTC (NF-κB activation inhibitor) after ending of the induction phase of the persistent inflammatory hypernociception, curtailed the hypernociception period as well as reducing NF-κB p65 subunit translocation. Treatment with antisense oligonucleotides against the NF-κB p65 subunit for 5 consecutive days also reduced persistent inflammatory hypernociception. Inhibition of PKA and PKCε reduced persistent inflammatory hypernociception, which was associated with inhibition of NF-κB p65 subunit translocation. Together these results suggest that peripheral activation of NF-κB by PKA and PKC in primary sensory neurons plays an important role in maintaining persistent inflammatory pain.


Subject(s)
Inflammation/metabolism , NF-kappa B/metabolism , Nociceptive Pain/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Inflammation/complications , Inflammation/enzymology , Male , Nociceptive Pain/enzymology , Nociceptive Pain/etiology , Protein Kinase C-epsilon/metabolism , Rats , Rats, Wistar
2.
Proc Natl Acad Sci U S A ; 110(27): 11193-8, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776243

ABSTRACT

The activation of the satellite glial cells (SGCs) surrounding the dorsal root ganglion (DRG) neurons appears to play a role in pathological pain. We tested the hypothesis that fractalkine, which is constitutively expressed by primary nociceptive neurons, is the link between peripheral inflammation and the activation of SGCs and is thus responsible for the genesis of the inflammatory pain. The injection of carrageenin into the rat hind paw induced a decrease in the mechanical nociceptive threshold (hypernociception), which was associated with an increase in mRNA and GFAP protein expression in the DRG. Both events were inhibited by anti-fractalkine antibody administered directly into the DRG (L5) [intraganglionar (i.gl.)]. The administration of fractalkine into the DRG (L5) produced mechanical hypernociception in a dose-, time-, and CX3C receptor-1 (CX3CR1)-dependent manner. Fractalkine's hypernociceptive effect appears to be indirect, as it was reduced by local treatment with anti-TNF-α antibody, IL-1-receptor antagonist, or indomethacin. Accordingly, the in vitro incubation of isolated and cultured SGC with fractalkine induced the production/release of TNF-α, IL-1ß, and prostaglandin E2. Finally, treatment with i.gl. fluorocitrate blocked fractalkine (i.gl.)- and carrageenin (paw)-induced hypernociception. Overall, these results suggest that, during peripheral inflammation, fractalkine is released in the DRG and contributes to the genesis of inflammatory hypernociception. Fractalkine's effect appears to be dependent on the activation of the SGCs, leading to the production of TNFα, IL-1ß, and prostanoids, which are likely responsible for the maintenance of inflammatory pain. Thus, these results indicate that the inhibition of fractalkine/CX3CR1 signaling in SGCs may serve as a target to control inflammatory pain.


Subject(s)
Chemokine CX3CL1/physiology , Neuroglia/physiology , Pain/physiopathology , Satellite Cells, Perineuronal/diagnostic imaging , Animals , Cytokines/biosynthesis , Dinoprostone/biosynthesis , Ganglia, Spinal/physiopathology , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Inflammation/genetics , Inflammation/physiopathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Male , Models, Neurological , Nociception/physiology , Pain/genetics , Pain Threshold/physiology , Radiography , Rats , Rats, Wistar
3.
Mol Pain ; 8: 10, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22316281

ABSTRACT

BACKGROUND: In addition to their central effects, opioids cause peripheral analgesia. There is evidence showing that peripheral activation of kappa opioid receptors (KORs) inhibits inflammatory pain. Moreover, peripheral µ-opioid receptor (MOR) activation are able to direct block PGE(2)-induced ongoing hyperalgesia However, this effect was not tested for KOR selective activation. In the present study, the effect of the peripheral activation of KORs on PGE(2)-induced ongoing hyperalgesia was investigated. The mechanisms involved were also evaluated. RESULTS: Local (paw) administration of U50488 (a selective KOR agonist) directly blocked, PGE(2)-induced mechanical hyperalgesia in both rats and mice. This effect was reversed by treating animals with L-NMMA or N-propyl-L-arginine (a selective inhibitor of neuronal nitric oxide synthase, nNOS), suggesting involvement of the nNOS/NO pathway. U50488 peripheral effect was also dependent on stimulation of PI3Kγ/AKT because inhibitors of these kinases also reduced peripheral antinociception induced by U50488. Furthermore, U50488 lost its peripheral analgesic effect in PI3Kγ null mice. Observations made in vivo were confirmed after incubation of dorsal root ganglion cultured neurons with U50488 produced an increase in the activation of AKT as evaluated by western blot analyses of its phosphorylated form. Finally, immunofluorescence of DRG neurons revealed that KOR-expressing neurons also express PI3Kγ (≅ 43%). CONCLUSIONS: The present study indicates that activation of peripheral KORs directly blocks inflammatory hyperalgesia through stimulation of the nNOS/NO signaling pathway which is probably stimulated by PI3Kγ/AKT signaling. This study extends a previously study of our group suggesting that PI3Kγ/AKT/nNOS/NO is an important analgesic pathway in primary nociceptive neurons.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , Hyperalgesia/pathology , Inflammation/pathology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Opioid, kappa/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Dinoprostone/pharmacology , Enzyme Activation/drug effects , Hyperalgesia/complications , Hyperalgesia/enzymology , Inflammation/complications , Inflammation/enzymology , Male , Mice , Mice, Inbred C57BL , Nociception/drug effects , Peripheral Nervous System/drug effects , Peripheral Nervous System/enzymology , Peripheral Nervous System/pathology , Rats , Receptors, Opioid, kappa/agonists , Signal Transduction/drug effects
4.
Proc Natl Acad Sci U S A ; 107(9): 4442-7, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20147620

ABSTRACT

Morphine is one of the most prescribed and effective drugs used for the treatment of acute and chronic pain conditions. In addition to its central effects, morphine can also produce peripheral analgesia. However, the mechanisms underlying this peripheral action of morphine have not yet been fully elucidated. Here, we show that the peripheral antinociceptive effect of morphine is lost in neuronal nitric-oxide synthase null mice and that morphine induces the production of nitric oxide in primary nociceptive neurons. The activation of the nitric-oxide pathway by morphine was dependent on an initial stimulation of PI3Kgamma/AKT protein kinase B (AKT) and culminated in increased activation of K(ATP) channels. In the latter, this intracellular signaling pathway might cause a hyperpolarization of nociceptive neurons, and it is fundamental for the direct blockade of inflammatory pain by morphine. This understanding offers new targets for analgesic drug development.


Subject(s)
KATP Channels/metabolism , Morphine/therapeutic use , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/therapeutic use , Animals , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage , Pain/drug therapy , Pain/enzymology , Pain/metabolism , Rats , Rats, Wistar
5.
Eur J Pharmacol ; 590(1-3): 127-35, 2008 Aug 20.
Article in English | MEDLINE | ID: mdl-18585702

ABSTRACT

Hydrogen sulfide (H(2)S) is an endogenous gas involved in several biological functions, including modulation of nociception. However, the mechanisms involved in such modulation are not fully elucidated. The present study demonstrated that the pretreatment of mice with PAG, a H(2)S synthesis inhibitor, reduced LPS-induced mechanical paw hypernociception. This inhibition of hypernociception was associated with the prevention of neutrophil recruitment to the plantar tissue. Conversely, PAG had no effect on LPS-induced production of the hypernociceptive cytokines, TNF-alpha, IL-1beta and CXCL1/KC and on hypernociception induced by PGE(2), a directly acting hypernociceptive mediator. In contrast with the pro-nociceptive role of endogenous H(2)S, systemic administration of NaHS, a H(2)S donor, reduced LPS-induced mechanical hypernociception in mice. Moreover, this treatment inhibited mechanical hypernociception induced by PGE(2), suggesting a direct effect of H(2)S on nociceptive neurons. The antinociceptive mechanism of exogenous H(2)S depends on K((ATP))(+) channels since the inhibition of PGE(2) hypernociception by NaHS was prevented by glibenclamide (K((ATP))(+) channel blocker). Finally, NaHS did not alter the thermal nociceptive threshold in the hot-plate test, confirming that its effect is mainly peripheral. Taken together, these results suggest that H(2)S has a dual role in inflammatory hypernociception: 1. an endogenous pro-nociceptive effect due to up-regulation of neutrophil migration, and 2. an antinociceptive effect by direct blockade of nociceptor sensitization modulating K((ATP))(+) channels.


Subject(s)
Hydrogen Sulfide/metabolism , Inflammation/physiopathology , Pain/physiopathology , Analgesics/pharmacology , Animals , Cell Movement , KATP Channels/physiology , Male , Mice , Mice, Inbred C57BL , Neutrophils/physiology , Pain/prevention & control , Rats , Rats, Wistar
6.
Life Sci ; 81(9): 765-71, 2007 Aug 09.
Article in English | MEDLINE | ID: mdl-17706725

ABSTRACT

A considerable amount of evidence suggests that temporomandibular joint (TMJ) pain associated with temporomandibular disorder results, at least in part, from an inflammatory episode. Although histamine can cause pain, it is not clear whether this mediator induces nociception in the TMJ. In this study, we investigated the contribution of endogenous histamine to formalin-induced nociception in the TMJ of rats. We also investigated whether the administration of histamine induces nociception in the TMJ and, if so, whether this effect is mediated by an indirect action on primary afferent nociceptors. Local administration of the H1-receptor antagonist pyrilamine prevented formalin-induced nociception in the TMJ in a dose-dependent manner. Local administration of histamine (250 microg) in the TMJ induced nociceptive behavior that was inhibited by co-administration of the lidocaine N-ethyl bromide quaternary salt QX-314 (2%) or the selective H1-receptor antagonist pyrilamine (400 microg). Nociception induced by histamine was also inhibited by pre-treatment with sodium cromoglycate (800 microg) and by co-administration of the 5-HT(3) receptor antagonist tropisetron (400 mug), while pyrilamine (400 mug) did not inhibit nociception induced by 5-hydroxytryptamine (5-HT, 250 microg) in the TMJ. Furthermore, histamine, in a dose that did not induce nociception by itself, strongly enhanced 5-HT-induced nociception. Finally, the administration of a sub-threshold dose of 5-HT (100 microg), but not of histamine (100 microg), elicited nociception in the TMJ previously challenged with the inflammatory agent carrageenan (100 microg). In conclusion, these data suggest that histamine induces TMJ nociception by an indirect mechanism involving endogenous release of 5-HT and activation of 5-HT(3) receptors on sensory afferents. It is proposed that histamine activates the H1 receptor to induce the release of 5-HT which depolarizes the nociceptor by activating 5-HT(3) receptor.


Subject(s)
Histamine/pharmacology , Pain , Receptors, Histamine H1/metabolism , Temporomandibular Joint Disorders , Temporomandibular Joint , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Histamine/metabolism , Histamine H1 Antagonists/pharmacology , Male , Pain/chemically induced , Pain/drug therapy , Pain/metabolism , Pain/physiopathology , Pain Measurement , Rats , Rats, Wistar , Serotonin/metabolism , Serotonin/pharmacology , Serotonin/physiology , Serotonin Antagonists/pharmacology , Temporomandibular Joint/drug effects , Temporomandibular Joint/metabolism , Temporomandibular Joint Disorders/chemically induced , Temporomandibular Joint Disorders/drug therapy , Temporomandibular Joint Disorders/metabolism , Temporomandibular Joint Disorders/physiopathology
7.
Eur J Neurosci ; 24(4): 1175-81, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16930443

ABSTRACT

The aim of the present study was to determine the effect of pertussis toxin (PTX) on inflammatory hypernociception measured by the rat paw pressure test and to elucidate the mechanism involved in this effect. In this test, prostaglandin E(2) (PGE(2)) administered subcutaneously induces hypernociception via a mechanism associated with neuronal cAMP increase. Local intraplantar pre-treatment (30 min before), and post-treatment (5 min after) with PTX (600 ng/paw1, in 100 microL) reduced hypernociception induced by prostaglandin E(2) (100 ng/paw, in 100 microL, intraplantar). Furthermore, local intraplantar pre-treatment (30 min before) with PTX (600 ng/paw, in 100 microL) reduced hypernociception induced by DbcAMP, a stable analogue of cAMP (100 microg/paw, in 100 microL, intraplantar), which indicates that PTX may have an effect other than just G(i)/G(0) inhibition. PTX-induced analgesia was blocked by selective inhibitors of nitric oxide synthase (L-NMMA), guanylyl cyclase (ODQ), protein kinase G (KT5823) and ATP-sensitive K(+) channel (Kir6) blockers (glybenclamide and tolbutamide). In addition, PTX was shown to induce nitric oxide (NO) production in cultured neurons of the dorsal root ganglia. In conclusion, this study shows a peripheral antinociceptive effect of pertussis toxin, resulting from the activation of the arginine/NO/cGMP/PKG/ATP-sensitive K(+) channel pathway.


Subject(s)
Analgesics/metabolism , Arginine/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Nitric Oxide/metabolism , Pertussis Toxin/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Adenosine Triphosphate/metabolism , Analgesia , Animals , Bucladesine/metabolism , Carbazoles/metabolism , Cells, Cultured , Dinoprostone/administration & dosage , Dinoprostone/immunology , Enzyme Inhibitors/metabolism , Ganglia, Spinal/cytology , Glyburide/metabolism , Indoles/metabolism , KATP Channels , Male , Neurons/cytology , Neurons/metabolism , Oxadiazoles/metabolism , Pain/metabolism , Pain Measurement , Quinoxalines/metabolism , Rats , Rats, Wistar , Signal Transduction/physiology , Tolbutamide/metabolism , omega-N-Methylarginine/metabolism
8.
Eur J Pharmacol ; 534(1-3): 258-63, 2006 Mar 18.
Article in English | MEDLINE | ID: mdl-16612844

ABSTRACT

The aim of this study was to investigate the effect of melatonin on the inflammatory increase in vascular permeability. Vascular permeability was stimulated by a nonspecific pro-inflammatory agent (carrageenan), by drugs that disrupt endothelial cells junction (histamine, serotonin or bradykinin) or drugs that promote neutrophil recruitment (leukotriene B4 or N-formyl-methionyl-leucyl-phenylalanine fMLP). Vascular permeability was measured by Evan's blue dye extravasation after simultaneous injection of melatonin and the pro-inflammatory drugs in rat dorsal skin. Melatonin only reduced the increase in vascular permeability induced by leukotriene B4, which activates both neutrophil and endothelial cells. The neutrophil expression of CD18 induced by leukotriene B4 or fMLP was not changed by melatonin. On the other hand, melatonin inhibited the leukotriene B4-induced endothelial cells hyperadhesiveness. Our findings suggest that vascular permeability reduction induced by local melatonin injection is mediated by a reduction of endothelial cells ability to interact with neutrophils.


Subject(s)
Capillary Permeability , Endothelial Cells/drug effects , Melatonin/pharmacology , Animals , Cell Adhesion , Cells, Cultured , Endothelial Cells/metabolism , Leukotriene B4/pharmacology , Male , Neutrophil Infiltration/drug effects , Neutrophils/metabolism , Rats , Rats, Wistar
9.
Mol Cell Endocrinol ; 249(1-2): 32-9, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-16490300

ABSTRACT

Deficiency of adrenal hormones promotes exacerbated neutrophil influx into inflammatory sites. We investigated the effect of adrenal deficiency on neutrophil mobilization comparing adrenalectomized (ADX) male Wistar rats to sham-operated (SO) or non-manipulated (N) animals, as controls. Seven days after surgeries, the number of neutrophils in peripheral blood was increased in ADX rats, by accelerating neutrophil maturation steps in the bone marrow. The investigation of adhesive properties on neutrophil membranes indicated reduced and increased expressions of L-selectin on cells present in the bone marrow and circulating blood, respectively. Similar levels of L-selectin mRNA in both cells from ADX or non-manipulated rats suggest that these effects do not depend on gene expression. Even though no differences in the expression of beta(2) integrin by neutrophils were detected, modulation on subsequent PMN activation may occur by adrenal hormones, since circulating neutrophils from ADX exhibit lower in vitro adherence to the endothelium. We conclude that adrenal hormones control the adhesive interactions of neutrophils with the bone marrow microenvironment and with the vascular endothelium chiefly by modulation of L-selectin on PMN membrane in a mechanism independent of L-selectin gene expression.


Subject(s)
Adrenal Cortex Hormones/deficiency , Neutrophils/physiology , Adrenalectomy , Animals , Bone Marrow Cells/metabolism , Cell Adhesion/physiology , Corticosterone/deficiency , Corticosterone/metabolism , Integrin beta Chains/metabolism , L-Selectin/metabolism , Male , Neutrophils/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...