Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 14(1): 30, 2017 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-28166793

RESUMEN

BACKGROUND: This study examined the development of chronic pain, a cardinal symptom of rheumatoid arthritis (RA), in mice with antigen- and collagen-induced arthritis (ACIA). Since the role of CD8+ T cells in arthritis is controversial, we investigated the consequences of CD8-depletion on arthritis development and opioid modulation of pain in this novel model of chronic autoimmune arthritis. METHODS: Disease severity in control and CD8-depleted animals was determined by histological assessment of knee-joint sections and measurement of autoantibody formation. Pain was evaluated by measuring mechanical allodynia and thermal hyperalgesia in von Frey and Hargreaves tests, respectively. The production and release of endogenous opioids and inflammatory cytokines was assessed in immunoassays. RESULTS: In ACIA, mice display persistent mechanical allodynia and thermal hyperalgesia for more than 2 months after induction of arthritis. The blockade of peripheral opioid receptors with naloxone-methiodide (NLXM) transiently increased thermal hyperalgesia, indicating that endogenous opioid peptides were released in the arthritic joint to inhibit pain. CD8+ T cell depletion did not affect autoantibody formation or severity of joint inflammation, but serum levels of the pro-inflammatory cytokines TNFα and IL-17 were increased. The release of opioid peptides from explanted arthritic knee cells and the NLXM effect were significantly reduced in the absence of CD8+ T cells. CONCLUSIONS: We have successfully modeled the development of chronic pain, a hallmark of RA, in ACIA. Furthermore, we detected a yet unknown protective role of CD8+ T cells in chronic ACIA since pro-inflammatory cytokines rose and opioid peptide release decreased in the absence of these cells.


Asunto(s)
Analgésicos Opioides/metabolismo , Artritis Experimental/complicaciones , Artritis Experimental/patología , Linfocitos T CD8-positivos/patología , Inflamación/etiología , Animales , Anticuerpos/efectos adversos , Artritis Experimental/inmunología , Antígenos CD8/inmunología , Linfocitos T CD8-positivos/metabolismo , Colágeno/toxicidad , Modelos Animales de Enfermedad , Encefalinas/metabolismo , Femenino , Adyuvante de Freund/inmunología , Adyuvante de Freund/toxicidad , Lateralidad Funcional , Hiperalgesia/etiología , Inflamación/patología , Metionina/metabolismo , Ratones , Ratones Endogámicos BALB C , Dimensión del Dolor , Umbral del Dolor/fisiología , Factores de Tiempo
2.
J Neuroimmune Pharmacol ; 12(1): 180-186, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27837406

RESUMEN

Immune cell-derived beta-endorphin (END) and other opioid peptides elicit potent and clinically relevant inhibition of pain (analgesia) in inflamed tissue by activation of peripheral opioid receptors. Pro-opiomelanocortin (POMC) is the polypeptide precursor of END and is processed by prohormone convertases (PCs). This study aims to decipher the processing of POMC in lymphocyte subsets in a rat model of unilateral painful hindpaw inflammation. Lymphocytes, isolated from popliteal lymph nodes, were separated into B-cells, T-cells, T-helper cells and cytotoxic T-cells using magnetic cell sorting, and were examined by polymerase chain reaction, immunofluorescence and radioimmunoassay. At 2 h of inflammation, POMC exon 2-3 mRNA was mostly expressed in B- but not in T-cells. Prohormone convertase 1 (PC1) mRNA and protein were upregulated in B-cells and T-helper cells. Prohormone convertase 2 (PC2) was expressed in T- and B-cells, both in inflamed and non-inflamed lymph nodes. END was expressed in B- but not in T-cells. We conclude that POMC, its processing enzymes and END are predominantly expressed in B-lymphocytes at 2 h of paw inflammation.


Asunto(s)
Linfocitos B/metabolismo , Dolor/metabolismo , Proopiomelanocortina/biosíntesis , ARN Mensajero/biosíntesis , Serina Endopeptidasas/biosíntesis , betaendorfina/biosíntesis , Animales , Linfocitos B/enzimología , Expresión Génica , Inflamación/genética , Inflamación/metabolismo , Masculino , Dolor/genética , Proopiomelanocortina/genética , Proproteína Convertasa 1/biosíntesis , Proproteína Convertasa 1/genética , Proproteína Convertasa 2/biosíntesis , Proproteína Convertasa 2/genética , ARN Mensajero/genética , Ratas , Ratas Wistar , Serina Endopeptidasas/genética , betaendorfina/genética
3.
Brain Behav Immun ; 57: 227-242, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27139929

RESUMEN

Opioids are the most powerful analgesics. As pain is driven by sensory transmission and opioid receptors couple to inhibitory G proteins, according to the classical concept, opioids alleviate pain by activating receptors on neurons and blocking the release of excitatory mediators (e.g., substance P). Here we show that analgesia can be mediated by opioid receptors in immune cells. We propose that activation of leukocyte opioid receptors leads to the secretion of opioid peptides Met-enkephalin, ß-endorphin and dynorphin A (1-17), which subsequently act at local neuronal receptors, to relieve pain. In a mouse model of neuropathic pain induced by a chronic constriction injury of the sciatic nerve, exogenous agonists of δ-, µ- and κ-opioid receptors injected at the damaged nerve infiltrated by opioid peptide- and receptor-expressing leukocytes, produced analgesia, as assessed with von Frey filaments. The analgesia was attenuated by pharmacological or genetic inactivation of opioid peptides, and by leukocyte depletion. This decrease in analgesia was restored by the transfer of wild-type, but not opioid receptor-lacking leukocytes. Ex vivo, exogenous opioids triggered secretion of opioid peptides from wild-type immune cells isolated from damaged nerves, which was diminished by blockade of Gαi/o or Gßγ (but not Gαs) proteins, by chelator of intracellular (but not extracellular) Ca(2+), by blockers of phospholipase C (PLC) and inositol 1,4,5-trisphosphate (IP3) receptors, and was partially attenuated by protein kinase C inhibitor. Similarly, the leukocyte depletion-induced decrease in exogenous opioid analgesia was re-established by transfer of immune cells ex vivo pretreated with extracellular Ca(2+) chelator, but was unaltered by leukocytes pretreated with intracellular Ca(2+) chelator or blockers of Gαi/o and Gßγ proteins. Thus, both ex vivo opioid peptide release and in vivo analgesia were mediated by leukocyte opioid receptors coupled to the Gαi/o-Gßγ protein-PLC-IP3 receptors-intracellular Ca(2+) pathway. Our findings suggest that opioid receptors in immune cells are important targets for the control of pathological pain.


Asunto(s)
Analgesia , Calcio/metabolismo , Leucocitos/metabolismo , Neuralgia/metabolismo , Péptidos Opioides/metabolismo , Receptores Opioides/metabolismo , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Ratones , Neuralgia/tratamiento farmacológico , Receptores Opioides/agonistas , Método Simple Ciego
4.
PLoS One ; 8(11): e79121, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24312175

RESUMEN

While humans and most animals respond to µ-opioid receptor (MOR) agonists with analgesia and decreased aggression, in the naked mole rat (NMR) opioids induce hyperalgesia and severe aggression. Single nucleotide polymorphisms in the human mu-opioid receptor gene (OPRM1) can underlie altered behavioral responses to opioids. Therefore, we hypothesized that the primary structure of the NMR MOR may differ from other species. Sequencing of the NMR oprm1 revealed strong homology to other mammals, but exposed three unique amino acids that might affect receptor-ligand interactions. The NMR and rat oprm1 sequences were cloned into mammalian expression vectors and transfected into HEK293 cells. Radioligand binding and 3'-5'-cyclic adenosine monophosphate (cAMP) enzyme immunoassays were used to compare opioid binding and opioid-mediated cAMP inhibition. At normalized opioid receptor protein levels we detected significantly lower [3H]DAMGO binding to NMR compared to rat MOR, but no significant difference in DAMGO-induced cAMP inhibition. Strong DAMGO-induced MOR internalization was detectable using radioligand binding and confocal imaging in HEK293 cells expressing rat or NMR receptor, while morphine showed weak or no effects. In summary, we found minor functional differences between rat and NMR MOR suggesting that other differences e.g. in anatomical distribution of MOR underlie the NMR's extreme reaction to opioids.


Asunto(s)
Receptores Opioides mu/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , AMP Cíclico/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/metabolismo , Humanos , Cinética , Ligandos , Ratas Topo , Datos de Secuencia Molecular , Morfina/metabolismo , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Receptores Opioides mu/química , Receptores Opioides mu/genética , Alineación de Secuencia
5.
JAKSTAT ; 2(4): e27638, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24470980

RESUMEN

During inflammation, several mediators directly or indirectly induce pain including pro-inflammatory cytokines and there is evidence that the JAK-STAT pathway is involved in the formation of pronociceptive cytokines. The same pathway, however, is also of importance for anti-inflammatory cytokines such as IL-4 to counteract the inflammatory reaction and-as it seems based on the current literature-nociceptive symptoms. Current therapeutic approaches targeting molecules of the JAK-STAT signaling cascade are auspicious but as this review demonstrates, more experimental and clinical studies are required to decipher the specific contribution of this pathway in the modulation of pain.

6.
Mol Pain ; 8: 83, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23146666

RESUMEN

BACKGROUND: Proopiomelanocortin (POMC)-derived beta-endorphin1-31 from immune cells can inhibit inflammatory pain. Here we investigated cytokine signaling pathways regulating POMC gene expression and beta-endorphin production in lymphocytes to augment such analgesic effects. RESULTS: Interleukin-4 dose-dependently elevated POMC mRNA expression in naïve lymph node-derived cells in vitro, as determined by real-time PCR. This effect was neutralized by janus kinase (JAK) inhibitors. Transfection of Signal Transducer and Activator of Transcription (STAT) 1/3 but not of STAT6 decoy oligonucleotides abolished interleukin-4 induced POMC gene expression. STAT3 was phosphorylated in in vitro interleukin-4 stimulated lymphocytes and in lymph nodes draining inflamed paws in vivo. Cellular beta-endorphin increased after combined stimulation with interleukin-4 and concanavalin A. Consistently, in vivo reduction of inflammatory pain by passively transferred T cells improved significantly when donor cells were pretreated with interleukin-4 plus concanavalin A. This effect was blocked by naloxone-methiodide. CONCLUSION: Interleukin-4 can amplify endogenous opioid peptide expression mediated by JAK-STAT1/3 activation in mitogen-activated lymphocytes. Transfer of these cells leads to inhibition of inflammatory pain via activation of peripheral opioid receptors.


Asunto(s)
Inflamación/metabolismo , Janus Quinasa 3/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Dolor/metabolismo , Proopiomelanocortina/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Células Cultivadas , Concanavalina A/farmacología , Inflamación/tratamiento farmacológico , Interleucina-4/farmacología , Interleucina-4/uso terapéutico , Janus Quinasa 3/genética , Masculino , Naloxona/análogos & derivados , Naloxona/farmacología , Dolor/tratamiento farmacológico , Compuestos de Amonio Cuaternario/farmacología , ARN Mensajero , Ratas , Ratas Wistar , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT3/genética
7.
Brain Behav Immun ; 24(5): 683-94, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19879349

RESUMEN

This review summarizes recent findings on neuro-immune mechanisms underlying opioid-mediated inhibition of pain. The focus is on events occurring in peripheral injured tissues that lead to the sensitization and excitation of primary afferent neurons, and on the modulation of such mechanisms by immune cell-derived opioid peptides. Primary afferent neurons are of particular interest from a therapeutic perspective because they are the initial generators of impulses relaying nociceptive information towards the spinal cord and the brain. Thus, if one finds ways to inhibit the sensitization and/or excitation of peripheral sensory neurons, subsequent central events such as wind-up, sensitization and plasticity may be prevented. This is in part achieved by endogenously released immune cell-derived opioid peptides within inflamed tissue. In addition, exogenous opioid receptor ligands that selectively modulate primary afferent function and do not cross the blood-brain barrier, avoid centrally mediated untoward side effects of conventional analgesics (e.g., opioids, anticonvulsants). This article discusses peripheral opioid receptors and their signaling pathways, opioid peptide-producing/secreting inflammatory cells and arising therapeutic perspectives.


Asunto(s)
Inflamación/metabolismo , Leucocitos/metabolismo , Péptidos Opioides/metabolismo , Dolor/metabolismo , Receptores Opioides/metabolismo , Animales , Humanos , Inflamación/inmunología , Inflamación/fisiopatología , Leucocitos/inmunología , Neuronas/inmunología , Neuronas/metabolismo , Péptidos Opioides/inmunología , Dolor/inmunología , Dolor/fisiopatología , Receptores Opioides/inmunología , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA