Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 41(13): 2870-2882, 2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33593854

RESUMO

Interleukin-4 (IL-4) is an anti-inflammatory cytokine, which can be protective in inflammatory and neurologic disorders, and can alleviate pain. Classically, IL-4 diminishes pain by blocking the production of proinflammatory cytokines. Here, we uncovered that IL-4 induces acute antinociception by IL-4 receptor α (IL-4Rα)-dependent release of opioid peptides from M1 macrophages at injured nerves. As a model of pathologic pain, we used a chronic constriction injury (CCI) of the sciatic nerve in male mice. A single application of IL-4 at the injured nerves (14 d following CCI) attenuated mechanical hypersensitivity evaluated by von Frey filaments, which was reversed by co-injected antibody to IL-4Rα, antibodies to opioid peptides such as Met-enkephalin (ENK), ß-endorphin and dynorphin A 1-17, and selective antagonists of δ-opioid, µ-opioid, and κ-opioid receptors. Injured nerves were predominately infiltrated by proinflammatory M1 macrophages and IL-4 did not change their numbers or the phenotype, assessed by flow cytometry and qRT-PCR, respectively. Macrophages isolated from damaged nerves by immunomagnetic separation (IMS) and stimulated with IL-4 dose dependently secreted all three opioid peptides measured by immunoassays. The IL-4-induced release of ENK was diminished by IL-4Rα antibody, intracellular Ca2+ chelator, and inhibitors of protein kinase A (PKA), phosphoinositide 3-kinase (PI3K), and ryanodine receptors. Together, we identified a new opioid mechanism underlying the IL-4-induced antinociception that involves PKA-mediated, PI3K-mediated, ryanodine receptor-mediated, and intracellular Ca2+-mediated release from M1 macrophages of opioid peptides, which activate peripheral opioid receptors in injured tissue.SIGNIFICANCE STATEMENT Interleukin-4 (IL-4) is an anti-inflammatory cytokine, which can ameliorate pain. The IL-4-mediated effects are considered to mostly result from the inhibition of the production of proinflammatory mediators (e.g., IL-1ß, tumor necrosis factor, prostaglandin E2). Here, we found that IL-4 injected at the injured nerves attenuates pain by releasing opioid peptides from the infiltrating macrophages in mice. The opioids were secreted by IL-4 in the intracellular Ca2+-dependent manner and activated local peripheral opioid receptors. These actions represent a novel mode of IL-4 action, since its releasing properties have not been so far reported. Importantly, our findings suggest that the IL-4-opioid system should be targeted in the peripheral damaged tissue, since this can be devoid of central and systemic side effects.


Assuntos
Interleucina-4/farmacologia , Macrófagos/metabolismo , Neuralgia/metabolismo , Neuralgia/patologia , Peptídeos Opioides/metabolismo , Animais , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/metabolismo , Neuropatia Ciática/metabolismo , Neuropatia Ciática/patologia
2.
Sci Rep ; 10(1): 18599, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33122720

RESUMO

The newly designed fentanyl derivative [( ±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide] (NFEPP) was recently shown to produce analgesia selectively via peripheral mu-opioid receptors (MOR) at acidic pH in rat inflamed tissues. Here, we examined the pH-dependency of NFEPP binding to brain MOR and its effects on bone cancer-induced pain in mice. The IC50 of NFEPP to displace bound [3H]-DAMGO was significantly higher compared to fentanyl at pH 7.4, but no differences were observed at pH 5.5 or 6.5. Intravenous NFEPP (30-100 nmol/kg) or fentanyl (17-30 nmol/kg) inhibited heat hyperalgesia in mice inoculated with B16-F10 melanoma cells. The peripherally-restricted opioid receptor antagonist naloxone-methiodide reversed the effect of NFEPP (100 nmol/kg), but not of fentanyl (30 nmol/kg). The antihyperalgesic effect of NFEPP was abolished by a selective MOR- (cyprodime), but not delta- (naltrindole) or kappa- (nor-binaltorphimine) receptor antagonists. Ten-fold higher doses of NFEPP than fentanyl induced maximal antinociception in mice without tumors, which was reversed by the non-restricted antagonist naloxone, but not by naloxone-methiodide. NFEPP also reduced heat hyperalgesia produced by fibrosarcoma- (NCTC 2472) or prostate cancer-derived (RM1) cells. These data demonstrate the increased affinity of NFEPP for murine MOR at low pH, and its ability to inhibit bone cancer-induced hyperalgesia through peripheral MOR. In mice, central opioid receptors may be activated by ten-fold higher doses of NFEPP.


Assuntos
Analgésicos Opioides/farmacologia , Dor do Câncer/tratamento farmacológico , Piperidinas/farmacologia , Receptores Opioides mu/metabolismo , Animais , Neoplasias Ósseas/complicações , Neoplasias Ósseas/metabolismo , Dor do Câncer/metabolismo , Linhagem Celular Tumoral , Fentanila/farmacologia , Concentração de Íons de Hidrogênio , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Ligantes , Masculino , Melanoma Experimental/complicações , Melanoma Experimental/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Morfinanos/farmacologia , Naloxona/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia
3.
Pain ; 161(12): 2798-2804, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32639370

RESUMO

Currently, opioids targeting mu-opioid receptors are the most potent drugs for acute and cancer pain. However, opioids produce adverse side effects such as constipation, respiratory depression, or addiction potential. We recently developed (±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenyl propionamide (NFEPP), a compound that does not evoke central or intestinal side effects due to its selective activation of mu-opioid receptors at low pH in peripheral injured tissues. Although we demonstrated that NFEPP effectively abolishes injury-induced pain, hyperalgesia, and allodynia in rodents, the efficacy of NFEPP in nonevoked ongoing pain remains to be established. Here, we examined reward, locomotor activity, and defecation in rats with complete Freund's adjuvant-induced paw inflammation to compare fentanyl's and NFEPP's potentials to induce side effects and to inhibit spontaneous pain. We demonstrate that low, but not higher, doses of NFEPP produce conditioned place preference but not constipation or motor disturbance, in contrast to fentanyl. Using a peripherally restricted antagonist, we provide evidence that NFEPP-induced place preference is mediated by peripheral opioid receptors. Our results indicate that a low dose of NFEPP produces reward by abolishing spontaneous inflammatory pain.


Assuntos
Analgésicos Opioides , Dor , Analgésicos Opioides/uso terapêutico , Animais , Concentração de Íons de Hidrogênio , Hiperalgesia/tratamento farmacológico , Dor/tratamento farmacológico , Ratos , Receptores Opioides , Receptores Opioides mu/genética
4.
Front Immunol ; 11: 300, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194554

RESUMO

Opioid receptors comprise µ (MOP), δ (DOP), κ (KOP), and nociceptin/orphanin FQ (NOP) receptors. Opioids are agonists of MOP, DOP, and KOP receptors, whereas nociceptin/orphanin FQ (N/OFQ) is an agonist of NOP receptors. Activation of all four opioid receptors in neurons can induce analgesia in animal models, but the most clinically relevant are MOP receptor agonists (e.g., morphine, fentanyl). Opioids can also affect the function of immune cells, and their actions in relation to immunosuppression and infections have been widely discussed. Here, we analyze the expression and the role of opioid receptors in peripheral immune cells and glia in the modulation of pain. All four opioid receptors have been identified at the mRNA and protein levels in immune cells (lymphocytes, granulocytes, monocytes, macrophages) in humans, rhesus monkeys, rats or mice. Activation of leukocyte MOP, DOP, and KOP receptors was recently reported to attenuate pain after nerve injury in mice. This involved intracellular Ca2+-regulated release of opioid peptides from immune cells, which subsequently activated MOP, DOP, and KOP receptors on peripheral neurons. There is no evidence of pain modulation by leukocyte NOP receptors. More good quality studies are needed to verify the presence of DOP, KOP, and NOP receptors in native glia. Although still questioned, MOP receptors might be expressed in brain or spinal cord microglia and astrocytes in humans, mice, and rats. Morphine acting at spinal cord microglia is often reported to induce hyperalgesia in rodents. However, most studies used animals without pathological pain and/or unconventional paradigms (e.g., high or ultra-low doses, pain assessment after abrupt discontinuation of chronic morphine treatment). Therefore, the opioid-induced hyperalgesia can be viewed in the context of dependence/withdrawal rather than pain management, in line with clinical reports. There is convincing evidence of analgesic effects mediated by immune cell-derived opioid peptides in animal models and in humans. Together, MOP, DOP, and KOP receptors, and opioid peptides in immune cells can ameliorate pathological pain. The relevance of NOP receptors and N/OFQ in leukocytes, and of all opioid receptors, opioid peptides and N/OFQ in native glia for pain control is yet to be clarified.


Assuntos
Leucócitos/fisiologia , Macrófagos/fisiologia , Neuroglia/fisiologia , Dor/fisiopatologia , Receptores Opioides/fisiologia , Animais , Humanos , Receptor de Nociceptina
5.
JCI Insight ; 5(4)2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-32102987

RESUMO

IL-4 is a pleiotropic antiinflammatory cytokine, which can be neuroprotective after nervous system injury. The beneficial actions of IL-4 are thought to result from the blunting of action of inflammatory mediators, such as proinflammatory cytokines. Here, we demonstrate that IL-4 induces M2 macrophages to continuously produce opioid peptides and ameliorate pain. IL-4 application at injured nerves in mice shifted F4/80+ macrophages from the proinflammatory M1 to the antiinflammatory M2 phenotype, which synthesized opioid peptides (Met-enkephalin, ß-endorphin, and dynorphin A 1-17). These effects were accompanied by a long-lasting attenuation of neuropathy-induced mechanical hypersensitivity, beyond the IL-4 treatment. This IL-4-induced analgesia was decreased by opioid peptide antibodies and opioid receptor (δ, µ, κ) antagonists applied at injured nerves, which confirms the involvement of the local opioid system. The participation of M2 macrophages was supported by analgesia in recipient mice injected at injured nerves with F4/80+ macrophages from IL-4-treated donors. Together, IL-4-induced M2 macrophages at injured nerves produced opioid peptides, which activated peripheral opioid receptors to diminish pain. Fostering the opioid-mediated actions of intrinsic M2 macrophages may be a strategy to tackle pathological pain.


Assuntos
Analgesia , Interleucina-4/farmacologia , Macrófagos/efeitos dos fármacos , Peptídeos Opioides/biossíntese , Animais , Temperatura Alta , Interleucina-4/uso terapêutico , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/tratamento farmacológico , Peptídeos Opioides/fisiologia , Tempo de Reação/efeitos dos fármacos , Receptores de Interleucina-4/antagonistas & inibidores , Receptores de Interleucina-4/fisiologia
6.
J Neuroinflammation ; 13(1): 262, 2016 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-27717401

RESUMO

BACKGROUND: During the inflammation which occurs following nerve damage, macrophages are recruited to the site of injury. Phenotypic diversity is a hallmark of the macrophage lineage and includes pro-inflammatory M1 and anti-inflammatory M2 populations. Our aim in this study was to investigate the ability of polarized M0, M1, and M2 macrophages to secrete opioid peptides and to examine their relative contribution to the modulation of neuropathic pain. METHODS: Mouse bone marrow-derived cells were cultured as unstimulated M0 macrophages or were stimulated into an M1 phenotype using lipopolysaccharide and interferon-γ or into an M2 phenotype using interleukin-4. The macrophage phenotypes were verified using flow cytometry for surface marker analysis and cytokine bead array for cytokine profile assessment. Opioid peptide levels were measured by radioimmunoassay and enzyme immunoassay. As a model of neuropathic pain, a chronic constriction injury (CCI) of the sciatic nerve was employed. Polarized M0, M1, and M2 macrophages (5 × 105 cells) were injected perineurally twice, on days 14 and 15 following CCI or sham surgery. Mechanical and heat sensitivity were measured using the von Frey and Hargreaves tests, respectively. To track the injected macrophages, we also transferred fluorescently stained polarized cells and analyzed the surface marker profile of endogenous and injected cells in the nerves ex vivo. RESULTS: Compared to M0 and M1 cells, M2 macrophages contained and released higher amounts of opioid peptides, including Met-enkephalin, dynorphin A (1-17), and ß-endorphin. M2 cells transferred perineurally at the nerve injury site reduced mechanical, but not heat hypersensitivity following the second injection. The analgesic effect was reversed by the perineurally applied opioid receptor antagonist naloxone methiodide. M2 cells did not affect sensitivity following sham surgery. Neither M0 nor M1 cells altered mechanical and heat sensitivity in CCI or sham-operated animals. Tracing the fluorescently labeled M0, M1, and M2 cells ex vivo showed that they remained in the nerve and preserved their phenotype. CONCLUSIONS: Perineural transplantation of M2 macrophages resulted in opioid-mediated amelioration of neuropathy-induced mechanical hypersensitivity, while M1 macrophages did not exacerbate pain. Therefore, rather than focusing on macrophage-induced pain generation, promoting opioid-mediated M2 actions may be more relevant for pain control.


Assuntos
Transferência Adotiva/métodos , Citocinas/metabolismo , Macrófagos/fisiologia , Neuralgia/imunologia , Neuralgia/patologia , Peptídeos Opioides/metabolismo , Aciltransferases/metabolismo , Animais , Polaridade Celular/efeitos dos fármacos , Modelos Animais de Doenças , Dinorfinas/metabolismo , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/metabolismo , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Lipopolissacarídeos/farmacologia , Macrófagos/classificação , Macrófagos/efeitos dos fármacos , Macrófagos/transplante , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Limiar da Dor/fisiologia , Estimulação Física , beta-Endorfina/metabolismo
7.
FASEB J ; 26(12): 5161-71, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22923332

RESUMO

Inflammatory pain can be controlled by endogenous opioid peptides. Here we blocked the degradation of opioids in peripheral injured tissue to locally augment this physiological system. In rats with hindpaw inflammation, inhibitors of aminopeptidase N (APN; bestatin) or neutral endopeptidase (NEP; thiorphan), and a dual inhibitor, NH(2)-CH-Ph-P(O)(OH)CH(2)-CH-CH(2)Ph(p-Ph)-CONH-CH-CH(3)-COOH (P8B), were applied to injured paws. Combined bestatin (1.25-5 mg)/thiorphan (0.2-0.8 mg) or P8B (0.0625-1 mg) alone elevated mechanical nociceptive thresholds to 307 and 227% of vehicle-treated controls, respectively. This analgesia was abolished by antibodies to methionine-enkephalin, leucine-enkephalin, and dynorphin A 1-17, by peripherally restricted and by selective µ-, δ-, and κ-opioid receptor antagonists. Flow cytometry and photospectrometry revealed expression and metabolic activity of APN and NEP on macrophages, granulocytes, and sciatic nerves from inflamed tissue. Radioimmunoassays showed that inhibition of leukocytic APN and NEP by bestatin (5-500 µM)/thiorphan (1-100 µM) combinations or by P8B (1-100 µM) prevented the degradation of enkephalins. Blockade of neuronal peptidases by bestatin (0.5-10 mM)/thiorphan (0.1-5 mM) or by P8B (0.1-10 mM) additionally hindered dynorphin A 1-17 catabolism. Thus, leukocytes and peripheral nerves are important sources of APN and NEP in inflamed tissue, and their blockade promotes peripheral opioid analgesia.


Assuntos
Antígenos CD13/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Inflamação/prevenção & controle , Neprilisina/antagonistas & inibidores , Dor/prevenção & controle , Alanina/análogos & derivados , Alanina/farmacologia , Sequência de Aminoácidos , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Antígenos CD13/metabolismo , Relação Dose-Resposta a Droga , Dinorfinas/imunologia , Dinorfinas/metabolismo , Dinorfinas/farmacologia , Encefalina Leucina/imunologia , Encefalina Leucina/metabolismo , Encefalina Leucina/farmacologia , Encefalina Metionina/imunologia , Encefalina Metionina/metabolismo , Encefalina Metionina/farmacologia , Citometria de Fluxo , Membro Posterior/efeitos dos fármacos , Membro Posterior/inervação , Membro Posterior/fisiopatologia , Inflamação/complicações , Inflamação/enzimologia , Leucina/análogos & derivados , Leucina/farmacologia , Leucócitos/efeitos dos fármacos , Leucócitos/enzimologia , Masculino , Antagonistas de Entorpecentes , Neprilisina/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Peptídeos Opioides/imunologia , Peptídeos Opioides/metabolismo , Peptídeos Opioides/farmacologia , Dor/complicações , Dor/enzimologia , Limiar da Dor/efeitos dos fármacos , Ácidos Fosfínicos/farmacologia , Ratos , Ratos Wistar , Receptores Opioides/metabolismo , Tiorfano/farmacologia
8.
Mol Pain ; 5: 72, 2009 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-20003437

RESUMO

BACKGROUND: Leukocytes infiltrating inflamed tissue produce and release opioid peptides such as beta-endorphin, which activate opioid receptors on peripheral terminals of sensory nerves resulting in analgesia. Gene therapy is an attractive strategy to enhance continuous production of endogenous opioids. However, classical viral and plasmid vectors for gene delivery are hampered by immunogenicity, recombination, oncogene activation, anti-bacterial antibody production or changes in physiological gene expression. Non-viral, non-plasmid minimalistic, immunologically defined gene expression (MIDGE) vectors may overcome these problems as they carry only elements needed for gene transfer. Here, we investigated the effects of a nuclear localization sequence (NLS)-coupled MIDGE encoding the beta-endorphin precursor proopiomelanocortin (POMC) on complete Freund's adjuvant-induced inflammatory pain in rats. RESULTS: POMC-MIDGE-NLS injected into inflamed paws appeared to be taken up by leukocytes resulting in higher concentrations of beta-endorphin in these cells. POMC-MIDGE-NLS treatment reversed enhanced mechanical sensitivity compared with control MIDGE-NLS. However, both effects were moderate, not always statistically significant or directly correlated with each other. Also, the anti-hyperalgesic actions could not be increased by enhancing beta-endorphin secretion or by modifying POMC-MIDGE-NLS to code for multiple copies of beta-endorphin. CONCLUSION: Although MIDGE vectors circumvent side-effects associated with classical viral and plasmid vectors, the current POMC-MIDGE-NLS did not result in reliable analgesic effectiveness in our pain model. This was possibly associated with insufficient and variable efficacy in transfection and/or beta-endorphin production. Our data point at the importance of the reproducibility of gene therapy strategies for the control of chronic pain.


Assuntos
Manejo da Dor , beta-Endorfina/metabolismo , Animais , Citometria de Fluxo , Vetores Genéticos , Imuno-Histoquímica , Inflamação/terapia , Masculino , Camundongos , Modelos Biológicos , Pró-Opiomelanocortina/genética , Radioimunoensaio , Ratos , Ratos Wistar , beta-Endorfina/genética
9.
J Clin Invest ; 119(2): 278-86, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19139563

RESUMO

The analgesic effects of leukocyte-derived opioids have been exclusively demonstrated for somatic inflammatory pain, for example, the pain associated with surgery and arthritis. Neuropathic pain results from injury to nerves, is often resistant to current treatments, and can seriously impair a patient's quality of life. Although it has been recognized that neuronal damage can involve inflammation, it is generally assumed that immune cells act predominately as generators of neuropathic pain. However, in this study we have demonstrated that leukocytes containing opioids are essential regulators of pain in a mouse model of neuropathy. About 30%-40% of immune cells that accumulated at injured nerves expressed opioid peptides such as beta-endorphin, Met-enkephalin, and dynorphin A. Selective stimulation of these cells by local application of corticotropin-releasing factor led to opioid peptide-mediated activation of opioid receptors in damaged nerves. This ultimately abolished tactile allodynia, a highly debilitating heightened response to normally innocuous mechanical stimuli, which is symptomatic of neuropathy. Our findings suggest that selective targeting of opioid-containing immune cells promotes endogenous pain control and offers novel opportunities for management of painful neuropathies.


Assuntos
Leucócitos/metabolismo , Peptídeos Opioides/fisiologia , Dor/prevenção & controle , Doenças do Sistema Nervoso Periférico/prevenção & controle , Animais , Hormônio Liberador da Corticotropina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Opioides/fisiologia
10.
Anesthesiology ; 101(1): 204-11, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15220792

RESUMO

BACKGROUND: Opioid-containing leukocytes migrate to peripheral sites of inflammation. On exposure to stress, opioid peptides are released, bind to opioid receptors on peripheral sensory neurons, and induce endogenous antinociception. In later stages of Freund's complete adjuvant-induced local inflammation, monocytes/macrophages are a major opioid-containing leukocyte subpopulation, but these cells also produce proalgesic cytokines. In this study, the role of tissue monocytes/macrophages in hyperalgesia and in peripheral opioid-mediated antinociception was investigated. METHODS: After intraplantar injection of Freund's adjuvant, leukocyte subpopulations and opioid-containing leukocytes were analyzed by flow cytometry in the inflamed paw in the presence or absence of monocyte/macrophage depletion by intraplantar injection of clodronate-containing liposomes (phosphate-buffered saline and empty liposomes served as controls). Paw volume was measured with a plethysmometer. Hyperalgesia was determined by measuring heat-induced paw withdrawal latency and paw pressure threshold. Paw pressure threshold was also measured after swim stress and injection of fentanyl. RESULTS: At 48 and 96 h of inflammation, it was found that (1). monocytes/macrophages were the largest leukocyte subpopulation (> 55% of all leukocytes) and the predominant producers of opioid peptides (71-77% of all opioid-containing leukocytes in the paw), (2). clodronate-containing liposomes depleted monocytes/macrophages by 30-35% (P < 0.05), (3). hyperalgesia was unaltered by liposome injection (P > 0.05), and (4) opioid-containing leukocytes and swim stress but not fentanyl-induced antinociception were significantly decreased by clodronate-containing liposomes (P < 0.05, P > 0.05, all by t test; opioid-containing cells and swim stress-induced increase of paw pressure threshold were reduced by 35-42% and 20%, respectively). CONCLUSION: Partial depletion of tissue monocytes/macrophages impairs peripheral endogenous opioid-mediated antinociception without affecting hyperalgesia.


Assuntos
Analgésicos Opioides/farmacologia , Fentanila/farmacologia , Hiperalgesia/induzido quimicamente , Inflamação/patologia , Macrófagos/patologia , Monócitos/patologia , Analgésicos não Narcóticos/farmacocinética , Analgésicos não Narcóticos/farmacologia , Analgésicos Opioides/administração & dosagem , Animais , Ácido Clodrônico/farmacocinética , Ácido Clodrônico/farmacologia , Fentanila/administração & dosagem , Citometria de Fluxo , Pé/patologia , Adjuvante de Freund , Temperatura Alta , Hiperalgesia/patologia , Hiperalgesia/psicologia , Inflamação/induzido quimicamente , Injeções , Lipossomos , Masculino , Medição da Dor/efeitos dos fármacos , Pressão , Ratos , Ratos Wistar
11.
Br J Pharmacol ; 142(4): 772-80, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15159283

RESUMO

1. Control of inflammatory pain can result from activation of opioid receptors on peripheral sensory nerves by opioid peptides secreted from leukocytes in response to stress (e.g. experimental swim stress or surgery). The extravasation of immunocytes to injured tissues involves rolling, adhesion and transmigration through the vessel wall, orchestrated by various adhesion molecules. 2. Here we evaluate the relative contribution of selectins, integrins alpha(4) and beta(2), and platelet-endothelial cell adhesion molecule-1 (PECAM-1) to the opioid-mediated inhibition of inflammatory pain. 3. We use flow cytometry, double immunofluorescence and nociceptive (paw pressure) testing in rats with unilateral hind paw inflammation induced by complete Freund's adjuvant. 4. In inflamed tissue, 43-58% of hematopoietic cells (CD45(+)) expressed opioid peptides. L-selectin and beta(2) were coexpressed by 7 and 98% of opioid-containing leukocytes, respectively. Alpha(4) integrin was expressed in low levels by the majority of leukocytes. Opioid-containing cells, vascular P- and E-selectin and PECAM-1 were simultaneously upregulated. 5. Swim stress produced potent opioid-mediated antinociception in inflamed tissue, unaffected by blockade of PECAM-1. However, blockade of L- and P-selectins by fucoidin, or of alpha(4) and beta(2) by monoclonal antibodies completely abolished peripheral stress-induced antinociception. This coincided with a 40% decrease in the migration of opioid-containing leukocytes to inflamed tissue. 6. These findings establish selectins and integrins alpha(4) and beta(2), but not PECAM-1, as important molecules involved in stress-induced opioid-mediated antinociception in inflammation. They point to a cautious use of anti-inflammatory treatments applying anti-selectin, anti-alpha(4) and anti-beta(2) strategies because they may impair intrinsic pain inhibition.


Assuntos
Antígenos CD18/fisiologia , Inflamação/prevenção & controle , Integrina alfa4/fisiologia , Dor/prevenção & controle , Selectinas/fisiologia , Molécula 1 de Adesão de Célula Vascular/fisiologia , Animais , Artrite Experimental , Plaquetas/química , Plaquetas/fisiologia , Antígenos CD18/efeitos dos fármacos , Movimento Celular/fisiologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Endorfinas/fisiologia , Células Endoteliais/química , Células Endoteliais/fisiologia , Endotélio Vascular/química , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Adjuvante de Freund/efeitos adversos , Expressão Gênica/efeitos dos fármacos , Alemanha , Inflamação/complicações , Integrina alfa4/efeitos dos fármacos , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/imunologia , Masculino , Dor/complicações , Molécula-1 de Adesão Celular Endotelial a Plaquetas/fisiologia , Polissacarídeos/farmacologia , Ratos , Ratos Wistar , Selectinas/classificação , Selectinas/efeitos dos fármacos
12.
Anesthesiology ; 100(1): 149-57, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14695736

RESUMO

BACKGROUND: Leukocytes can control inflammatory pain by secretion of opioid peptides, stimulated by cold-water swimming or local injection of corticotropin-releasing factor, and subsequent activation of opioid receptors on peripheral sensory neurons. This study investigated whether mobilization of polymorphonuclear cells (PMN) by granulocyte colony-stimulating factor (G-CSF) and stem cell factor (SCF) enhances immigration of opioid-containing PMN and peripheral opioid analgesia in rats with Freund complete adjuvant-induced hind paw inflammation. METHODS: In circulating PMN of rats treated with G-CSF+SCF and sham-treated rats, opioid peptide content was measured by radioimmunoassay. Expression of adhesion molecules (CD62L, CD49d, CD18), in vitro migration in the Boyden chamber, and infiltrating leukocytes were analyzed by flow cytometry. Chemokine messenger RNA transcription was quantified by LightCycler polymerase chain reaction. Paw pressure threshold was measured at baseline, after cold-water swimming, and after injection of corticotropin-releasing factor. RESULTS: G-CSF+SCF treatment increased circulating PMN (11-fold, P < 0.05). Mobilized PMN had decreased content of beta-endorphin but not of Met-enkephalin per cell, down-regulation of CD62L, up-regulation of CD49d (but no change in CD18), and reduced migration toward higher chemokine concentrations (all P < 0.05). In the paw, one of four chemokine messenger RNAs was significantly expressed during the first 2 h of inflammation (P < 0.05), immigration of PMN and opioid-containing cells was slightly increased (1.5-fold, P < 0.05), and baseline paw pressure threshold, as well as paw pressure threshold increases induced by corticotropin-releasing factor and cold-water swimming, were unchanged (P > 0.05). CONCLUSIONS: G-CSF+SCF mobilized circulating opioid-containing PMN but had a minor influence on cell migration and peripheral analgesia, probably because of the low expression of chemokines in the inflamed paw and one of the decreased beta-endorphin content in PMN.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Inflamação/complicações , Entorpecentes/metabolismo , Entorpecentes/farmacologia , Neutrófilos/metabolismo , Dor/tratamento farmacológico , Fator de Células-Tronco/farmacologia , Animais , Moléculas de Adesão Celular/farmacologia , Separação Celular , Quimiocinas/biossíntese , Quimiotaxia de Leucócito/efeitos dos fármacos , Primers do DNA , Citometria de Fluxo , Membro Posterior/fisiologia , Luz , Masculino , Dor/fisiopatologia , Medição da Dor/efeitos dos fármacos , RNA Mensageiro/biossíntese , Radioimunoensaio , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
J Neuroimmunol ; 126(1-2): 5-15, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12020952

RESUMO

Recently, two novel highly selective mu-opioid receptor (MOR) agonists, endomorphin-1 and endomorphin-2, have been isolated from bovine as well as human brains and were proposed to be the endogenous ligand for MOR. Later, endomorphin-1 and endomorphin-2 have been detected in the immune system of rats and humans using radioimmunoassay in combination with reverse-high-phase-liquid chromatography. In the present study, we analyzed the expression of endomorphin-1, endomorphin-2 and MOR by immunohistochemistry in a model of Freund's complete adjuvant (FCA)-induced painful inflammation. While MOR was upregulated on peripheral and central nerve terminals, inflammation did not alter endomorphin-2 expression in nerve fibers either in the dorsal horn of the spinal cord or in subcutaneous tissue. Endomorphin-1 and endomorphin-2 were expressed in immune cells (macrophage/monocytes) in the medullary region of the popliteal lymph nodes. The proportion of immunocytes (macrophage/monocytes, lymphocytes) containing endomorphin-1 and endomorphin-2 was increased in inflamed lymph nodes and subcutaneous paw tissue of animals with local inflammatory pain. Taken together, the upregulation of MOR and of its endogenous ligands endomorphin-1 and endomorphin-2 in immunocytes suggests an involvement of these opioid peptides in the peripheral control of inflammatory pain.


Assuntos
Linfonodos/citologia , Oligopeptídeos/análise , Dor/imunologia , Células do Corno Posterior/química , Animais , Adjuvante de Freund , Membro Posterior , Imuno-Histoquímica , Linfócitos/química , Macrófagos/química , Masculino , Monócitos/química , Dor/induzido quimicamente , Ratos , Ratos Wistar , Pele/química , Pele/imunologia , Pele/inervação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA