Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Neuroinflammation ; 13(1): 132, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27245576

RESUMEN

BACKGROUND: T cell-derived opioids play a key role in the control of inflammatory pain. However, the nature of opioids produced by T cells is still matter of debate in mice. Whereas ß-endorphin has been found in T lymphocytes by using antibody-based methods, messenger RNA (mRNA) quantification shows mainly mRNA encoding for enkephalins. The objective of the study is to elucidate the nature of T cell-derived opioids responsible for analgesia and clarify discrepancy of the results at the protein and genetic levels. METHODS: CD4(+) T lymphocytes were isolated from wild-type and enkephalin-deficient mice. mRNA encoding for ß-endorphin and enkephalin was quantified by RT-qPCR. The binding of commercially available polyclonal anti-endorphin antibodies to lymphocytes from wild-type or enkephalin knockout mice was assessed by cytofluorometry. Opioid-mediated analgesic properties of T lymphocytes from wild-type and enkephalin-deficient mice were compared in a model of inflammation-induced somatic pain by measuring sensitivity to mechanical stimuli using calibrated von Frey filaments. RESULTS: CD4(+) T lymphocytes expressed high level of mRNA encoding for enkephalins but not for ß-endorphin in mice. Anti-ß-endorphin polyclonal IgG antibodies are specific for ß-endorphin but cross-react with enkephalins. Anti-ß-endorphin polyclonal antibodies bound to wild-type but not enkephalin-deficient CD4(+) T lymphocytes. Endogenous regulation of inflammatory pain by wild-type T lymphocytes was completely abolished when T lymphocytes were deficient in enkephalins. Pain behavior of immune-deficient (i.e., without B and T lymphocytes) mice was superimposable to that of mice transferred with enkephalin-deficient lymphocytes. CONCLUSIONS: Rabbit polyclonal anti-ß-endorphin serum IgG bind to CD4(+) T lymphocytes because of their cross-reactivity towards enkephalins. Thus, staining of T lymphocytes by anti-ß-endorphin polyclonal IgG reported in most of studies in mice is because of their binding to enkephalins. In mice, CD4(+) T lymphocytes completely lose their analgesic opioid-mediated activity when lacking enkephalins.


Asunto(s)
Analgesia/métodos , Linfocitos T CD4-Positivos/metabolismo , Encefalinas/metabolismo , Dimensión del Dolor/métodos , Dolor/metabolismo , Dolor/prevención & control , Secuencia de Aminoácidos , Animales , Linfocitos T CD4-Positivos/inmunología , Encefalinas/genética , Encefalinas/inmunología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Dolor/inmunología , Conejos , Distribución Aleatoria
2.
Gastroenterology ; 146(1): 166-75, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24055279

RESUMEN

BACKGROUND & AIMS: A dysregulated response of CD4(+) T cells against the microbiota contributes to the development of inflammatory bowel disease. Effector CD4(+) T cells, generated in response to microbe-derived antigens, can reduce somatic inflammatory pain through the local release of opioids. We investigated whether colitogenic CD4(+) T cells that accumulate in the inflamed colon also produce opioids and are able to counteract inflammation-induced visceral pain in mice. METHODS: Colitis was induced via transfer of naive CD4(+)CD45RB(high) T cells to immune-deficient mice or by administration of dextran sulfate sodium. Mice without colitis were used as controls. Samples of colon tissue were collected, and production of opioids by immune cells from inflamed intestine was assessed by quantitative polymerase chain reaction and cytofluorometry analyses. The role of intestinal opioid tone in inflammation-induced visceral hypersensitivity was assessed by colorectal distention. RESULTS: In mice with T cell- or dextran sulfate sodium-induced colitis, colitogenic CD4(+) T cells (T-helper 1 and Th17 cells) accumulated in the inflamed intestine and expressed a high level of endogenous opioids. In contrast, macrophages and epithelial cells did not express opioids; opioid synthesis in the myenteric plexus was not altered on induction of inflammation. In mice with colitis, the local release of opioids by colitogenic CD4(+) T cells led to significant reduction of inflammation-associated visceral hypersensitivity. CONCLUSIONS: In mice, colitogenic Th1 and Th17 cells promote intestinal inflammation and colonic tissue damage but have simultaneous opioid-mediated analgesic activity, thereby reducing abdominal pain.


Asunto(s)
Colitis/inmunología , Colon/inmunología , Plexo Mientérico/inmunología , Péptidos Opioides/inmunología , Células TH1/inmunología , Células Th17/inmunología , Dolor Visceral/inmunología , Animales , Colitis/inducido químicamente , Colitis/patología , Colon/inervación , Colon/patología , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Inmunidad Mucosa , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Plexo Mientérico/fisiología , Péptidos Opioides/fisiología
3.
J Hepatol ; 58(5): 984-92, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23333450

RESUMEN

BACKGROUND & AIMS: Nutrients influence non-alcoholic fatty liver disease. Essential fatty acids deficiency promotes various syndromes, including hepatic steatosis, through increased de novo lipogenesis. The mechanisms underlying such increased lipogenic response remain unidentified. METHODS: We used wild type mice and mice lacking Liver X Receptors to perform a nutrigenomic study that aimed at examining the role of these transcription factors. RESULTS: We showed that, in the absence of Liver X Receptors, essential fatty acids deficiency does not promote steatosis. Consistent with this, Liver X Receptors are required for the elevated expression of genes involved in lipogenesis in response to essential fatty acids deficiency. CONCLUSIONS: This work identifies, for the first time, the central role of Liver X Receptors in steatosis induced by essential fatty acids deficiency.


Asunto(s)
Ácidos Grasos Esenciales/deficiencia , Hígado Graso/fisiopatología , Expresión Génica/fisiología , Lipogénesis/genética , Lipogénesis/fisiología , Receptores Nucleares Huérfanos/fisiología , Animales , Colesterol/metabolismo , Enfermedades Carenciales/fisiopatología , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Hígado/metabolismo , Receptores X del Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores Nucleares Huérfanos/deficiencia , Receptores Nucleares Huérfanos/genética , Factores de Transcripción/fisiología , Triglicéridos/metabolismo , Regulación hacia Arriba/fisiología
4.
J Immunol ; 186(9): 5078-84, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21422247

RESUMEN

Pain is an inherent component of inflammation often accompanying immune response. A large spectrum of molecules released within the inflamed tissue induces pain by stimulating primary afferent neurons in situ. Activity of primary sensitive fibers can be counteracted by local opioid release by leukocytes. In this study, we investigated the endogenous regulation of CFA-induced inflammatory pain in the context of adaptive T cell immune response. The nociceptive response to mechanical stimuli was studied using von Frey filaments in mice immunized with OVA in CFA. The nociceptive response of nude versus wild-type mice was dramatically increased, demonstrating T cell deficiency associated with increased pain sensitivity. Based on adoptive transfer experiments of OVA-specific CD4(+) T lymphocytes into nude mice, we show that Ag-specific activated, but not resting T lymphocytes are responsible for the spontaneous relief of inflammation-induced pain following Ag challenge. The analgesia was dependent on opioid release by Ag-primed CD4(+) T lymphocytes at the inflammatory site. Indeed, T cell-mediated analgesia was inhibited by local injection of an opioid receptor antagonist, unable to cross the blood-brain barrier. Notably, we found opioid precursor mRNA to be >7-fold increased in Ag-specific activated CD4(+) T lymphocytes, as compared with resting T lymphocytes in vivo. Taken together, our results show that CD4(+) T lymphocytes acquire antinociceptive effector properties when specifically primed by Ag and point out analgesia as a property linked to the effector phase of adaptive T cell response.


Asunto(s)
Inmunidad Adaptativa/inmunología , Linfocitos T CD4-Positivos/inmunología , Péptidos Opioides/inmunología , Dolor/inmunología , Traslado Adoptivo , Animales , Separación Celular , Citometría de Flujo , Inmunohistoquímica , Inflamación/complicaciones , Activación de Linfocitos/inmunología , Ratones , Ratones Desnudos , Dolor/etiología , Reacción en Cadena de la Polimerasa
5.
J Neuroimmunol ; 197(1): 21-8, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18486241

RESUMEN

Endogenous opioid peptides are locally produced at the inflammatory site where antigens are captured and processed by dendritic cells (DCs). Subsequently, maturing DCs migrate towards draining lymph nodes to initiate T cell response. Given the primordial role of DCs in adaptive immune response, we examined whether opioids may affect the migratory response of DCs. We found that the delta opioid receptor (DOR) mRNA was expressed at low level in bone marrow-derived immature DCs and up-regulated upon DC maturation. Moreover, DOR agonists triggered DC chemotaxis in vitro. In vivo, enkephalins prevented the egress of mature DCs injected into the peritoneal cavity of normal mice. This effect was inhibited by blocking opioid receptors on mature DCs. The cross-talk between CCR7 and DOR receptors that are both up-regulated during DC maturation was then examined. Whereas opioids did not alter the migratory responsiveness to CCR7 ligands, DOR-mediated mobilization of mature DCs was inhibited by CCL19 and CCL21 suggesting that the opioid chemotactic activity decreases as the concentration of the chemokines increases.


Asunto(s)
Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Quimiotaxis de Leucocito/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Receptores Opioides delta/fisiología , Animales , Células de la Médula Ósea/citología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Cultivadas , Quimiocina CCL19/fisiología , Quimiocina CCL21/fisiología , Células Dendríticas/citología , Células Dendríticas/trasplante , Encefalina D-Penicilamina (2,5)/agonistas , Encefalina D-Penicilamina (2,5)/biosíntesis , Encefalina D-Penicilamina (2,5)/fisiología , Mediadores de Inflamación/fisiología , Ratones , Ratones Endogámicos BALB C , Receptor Cross-Talk/inmunología , Receptores CCR7/fisiología , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides delta/biosíntesis , Regulación hacia Arriba/inmunología
6.
J Gastroenterol ; 53(2): 215-226, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28424989

RESUMEN

BACKGROUND: Endogenous opioids, including enkephalins, are fundamental regulators of pain. In inflammatory conditions, the local release of opioids by leukocytes at the inflammatory site inhibits nociceptor firing, thereby inducing analgesia. Accordingly, in chronic intestinal Th1/Th17-associated inflammation, enkephalins released by colitogenic CD4+ T lymphocytes relieve inflammation-induced visceral pain. The present study aims to investigate whether mucosal T-cell-derived enkephalins also exhibit a potent anti-inflammatory activity as described for exogenous opioid drugs in Th1/Th17-associated colitis. METHODS: The anti-inflammatory effects of endogenous opioids were investigated in both Th1/Th17-associated (transfer of CD4+CD45RBhigh T lymphocytes) and Th2-associated (oxazolone) colitis models in mice. Inflammation-induced colonic damage and CD4+ T cell subsets were compared in mice treated or not treated with naloxone methiodide, a peripheral antagonist of opioid receptors. The anti-inflammatory activity of T-cell-derived enkephalins was further estimated by comparison of colitis severity in immunodeficient mice into which naïve CD4+CD45RBhigh T lymphocytes originating from wild-type or enkephalin-knockout mice had been transferred. RESULTS: Peripheral opioid receptor blockade increases the severity of Th1/Th17-induced colitis and attenuates Th2 oxazolone colitis. The opposite effects of naloxone methiodide treatment in these two models of intestinal inflammation are dependent on the potency of endogenous opioids to promote a Th2-type immune response. Accordingly, the transfer of enkephalin-deficient CD4+CD45RBhigh T lymphocytes into immunodeficient mice exacerbates inflammation-induced colonic injury. CONCLUSIONS: Endogenous opioids, including T-cell-derived enkephalins, promote a Th2-type immune response, which, depending on the context, may either attenuate (Th1/Th17-associated) or aggravate (Th2-associated) intestinal inflammation.


Asunto(s)
Colitis/inmunología , Encefalinas/inmunología , Células TH1/inmunología , Células Th17/inmunología , Animales , Linfocitos T CD4-Positivos/trasplante , Colitis/tratamiento farmacológico , Inmunidad Mucosa , Transfusión de Linfocitos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Naloxona/uso terapéutico , Subgrupos de Linfocitos T/inmunología , Dolor Visceral/inmunología
7.
Pain ; 159(2): 331-341, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29140925

RESUMEN

T lymphocytes play a pivotal role in endogenous regulation of inflammatory visceral pain. The analgesic activity of T lymphocytes is dependent on their production of opioids, a property acquired on antigen activation. Accordingly, we investigated whether an active recruitment of T lymphocytes within inflamed colon mucosa via a local vaccinal strategy may counteract inflammation-induced visceral pain in mice. Mice were immunized against ovalbumin (OVA). One month after immunization, colitis was induced by adding 3% (wt/vol) dextran sulfate sodium into drinking water containing either cognate antigen OVA or control antigen bovine serum albumin for 5 days. Noncolitis OVA-primed mice were used as controls. Visceral sensitivity was then determined by colorectal distension. Oral administration of OVA but not bovine serum albumin significantly reduced dextran sulfate sodium-induced abdominal pain without increasing colitis severity in OVA-primed mice. Analgesia was dependent on local release of enkephalins by effector anti-OVA T lymphocytes infiltrating the inflamed mucosa. The experiments were reproduced with the bacillus Calmette-Guerin vaccine as antigen. Similarly, inflammatory visceral pain was dramatically alleviated in mice vaccinated against bacillus Calmette-Guerin and then locally administered with live Mycobacterium bovis. Together, these results show that the induction of a secondary adaptive immune response against vaccine antigens in inflamed mucosa may constitute a safe noninvasive strategy to relieve from visceral inflammatory pain.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Colitis/complicaciones , Colitis/etiología , Inmunización/efectos adversos , Membrana Mucosa/patología , Dolor Visceral , Animales , Antígenos CD11/genética , Antígenos CD11/metabolismo , Colitis/patología , Modelos Animales de Enfermedad , Encefalinas/deficiencia , Encefalinas/genética , Encefalinas/farmacología , Adyuvante de Freund/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina/efectos adversos , Precursores de Proteínas/deficiencia , Precursores de Proteínas/genética , Estadísticas no Paramétricas , Dolor Visceral/etiología , Dolor Visceral/inmunología , Dolor Visceral/patología
8.
Inflamm Bowel Dis ; 20(10): 1870-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24846722

RESUMEN

Painful sensation is a hallmark of microbe-induced inflammation. This inflammatory pain is downregulated a few days after infection by opioids locally released by effector T lymphocytes generated in response to microbe-derived antigens. This review focuses on the endogenous regulation of inflammatory pain associated with adaptive T-cell response and puts in perspective the clinical consequences of the opioid-mediated analgesic activity of colitogenic T lymphocytes in inflammatory bowel disease.


Asunto(s)
Inflamación/complicaciones , Péptidos Opioides/uso terapéutico , Dolor/tratamiento farmacológico , Linfocitos T/inmunología , Humanos , Inflamación/inmunología , Dolor/etiología , Pronóstico , Linfocitos T/metabolismo
9.
PLoS One ; 7(9): e46348, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029489

RESUMEN

G-protein coupled receptors (GPCRs) play a major role in a number of physiological and pathological processes. Thus, GPCRs have become the most frequent targets for development of new therapeutic drugs. In this context, the availability of highly specific antibodies may be decisive to obtain reliable findings on localization, function and medical relevance of GPCRs. However, the rapid and easy generation of highly selective anti-GPCR antibodies is still a challenge. Herein, we report that highly specific antibodies suitable for detection of GPCRs in native and unfolded forms can be elicited by immunizing animals against purified full length denatured recombinant GPCRs. Contrasting with the currently admitted postulate, our study shows that an active and well-folded GPCR is not required for the production of specific anti-GPCR antibodies. This new immunizing strategy validated with three different human GPCR (µ-opioid, κ-opioid, neuropeptide FF2 receptors) might be generalized to other members of the GPCR family.


Asunto(s)
Anticuerpos/inmunología , Inmunoglobulina G/biosíntesis , Receptores de Neuropéptido/inmunología , Receptores Opioides kappa/inmunología , Receptores Opioides mu/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos/aislamiento & purificación , Humanos , Inmunización , Inmunoglobulina G/inmunología , Inmunoglobulina G/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Pichia/genética , Desnaturalización Proteica , Pliegue de Proteína , Receptores de Neuropéptido/administración & dosificación , Receptores de Neuropéptido/genética , Receptores Opioides kappa/administración & dosificación , Receptores Opioides kappa/genética , Receptores Opioides mu/administración & dosificación , Receptores Opioides mu/genética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología
10.
Pain ; 153(2): 485-493, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22188867

RESUMEN

Effector CD4(+) T lymphocytes generated in response to antigens produce endogenous opioids. Thus, in addition to their critical role in host defenses against pathogens, effector CD4(+) T lymphocytes contribute to relieving inflammatory pain. In this study, we investigated mechanisms of opioid release by antigen-experienced effector CD4(+) T cells that leave draining lymph nodes and come back into the inflammatory site. Effector antigen-primed CD4(+) T lymphocytes generated in vitro were intravenously injected into nude mice previously immunized with either cognate or irrelevant antigens in complete Freund adjuvant (CFA). CFA-induced mechanical hyperalgesia was only reduced in mice immunized with cognate antigen. Thus, antinociceptive activity of effector CD4(+) T cells requires the presence of the antigen for which they are specific within the inflammatory site. Accordingly, analgesia was inhibited by neutralizing cognate T cell receptor-mediated interaction between effector CD4(+) T lymphocytes and antigen-presenting cells at the site of inflammation. Analgesia was observed by transferring effector CD4(+) T lymphocytes with Th1 or Th2 phenotype, suggesting that antinociceptive activity is a fundamental property of effector CD4(+) T lymphocytes irrespective of their effector functions. Based on the use of agonists and antagonists selective for each of the opioid receptor subclasses, we showed that analgesia induced by T cell-derived opioids is elicited via activation of δ-type opioid receptors in the periphery. Thus, the antinociceptive activity is a fundamental property associated with the effector phase of adaptive immunity, which is driven by recognition of the cognate antigen by effector CD4(+) T lymphocytes at the inflammatory site.


Asunto(s)
Analgesia/métodos , Linfocitos T CD4-Positivos/metabolismo , Péptidos Opioides/metabolismo , Dolor/inmunología , Inmunidad Adaptativa , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Células Cultivadas , Encefalinas/sangre , Encefalinas/genética , Encefalinas/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Péptidos Opioides/sangre , Péptidos Opioides/genética , Dolor/metabolismo , Dolor/patología , Precursores de Proteínas/sangre , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo
11.
Sci Transl Med ; 4(158): 158ra144, 2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23115353

RESUMEN

Elafin, a natural protease inhibitor expressed in healthy intestinal mucosa, has pleiotropic anti-inflammatory properties in vitro and in animal models. We found that mucosal expression of Elafin is diminished in patients with inflammatory bowel disease (IBD). This defect is associated with increased elastolytic activity (elastase-like proteolysis) in colon tissue. We engineered two food-grade strains of lactic acid bacteria (LAB) to express and deliver Elafin to the site of inflammation in the colon to assess the potential therapeutic benefits of the Elafin-expressing LAB. In mouse models of acute and chronic colitis, oral administration of Elafin-expressing LAB decreased elastolytic activity and inflammation and restored intestinal homeostasis. Furthermore, when cultures of human intestinal epithelial cells were treated with LAB secreting Elafin, the inflamed epithelium was protected from increased intestinal permeability and from the release of cytokines and chemokines, both of which are characteristic of intestinal dysfunction associated with IBD. Together, these results suggest that oral delivery of LAB secreting Elafin may be useful for treating IBD in humans.


Asunto(s)
Bacterias/metabolismo , Colon/inmunología , Colon/microbiología , Elafina/metabolismo , Inflamación/prevención & control , Animales , Bacterias/genética , Humanos , Técnicas In Vitro , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal , Ratones
12.
J Invest Dermatol ; 130(5): 1337-44, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20107485

RESUMEN

Endogenous opioid peptides mainly produced by neurons are also released by immune cells. They bind to mu- (mu-opioid receptor, MOR), delta-, and kappa-opioid receptors. On the basis of studies on mice showing that MOR is the main mediator of the deleterious effects of opioids on immunity, we wondered whether MOR, absent under normal conditions, is expressed in some pathological situations such as lymphomas. mRNA expression for all three opioid receptors was examined in lymph node biopsy samples from patients with non-Hodgkin's B-cell and T-cell lymphomas. We found that MOR and one of its ligands (enkephalin) are simultaneously expressed almost exclusively in lymph nodes from patients with Sézary cutaneous T cell lymphoma. As MOR was undetectable in circulating malignant T lymphocytes and in normal immune cells, we hypothesized that tumor-released cytokines might induce MOR expression in non-neoplastic lymph node cells. The correlation between mRNA levels of MOR and interleukin-13 (IL-13) within lymph nodes from Sézary patients led us to investigate the ability of IL-13 to upregulate MOR expression in normal immune cell subsets. We found that IL-13 upregulates MOR in activated Langerhans cells. Thus, our data suggest that, under pathological conditions, IL-13 overexpression might allow immune-derived endogenous opioids to down-modulate immune response.


Asunto(s)
Interleucina-13/genética , Receptores Opioides mu/genética , Síndrome de Sézary/inmunología , Síndrome de Sézary/fisiopatología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/fisiopatología , Biopsia , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/fisiología , Células Cultivadas , Células Dendríticas/patología , Células Dendríticas/fisiología , Expresión Génica/inmunología , Humanos , Tolerancia Inmunológica/fisiología , Interleucina-13/inmunología , Células de Langerhans/patología , Células de Langerhans/fisiología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Monocitos/patología , Monocitos/fisiología , ARN Mensajero/metabolismo , Receptores Opioides mu/inmunología , Síndrome de Sézary/patología , Neoplasias Cutáneas/patología
13.
Pain ; 146(1-2): 121-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19674841

RESUMEN

Serine proteases such as thrombin, trypsin and mast cell tryptase can act on different cell types through protease-activated receptors (PARs). These receptors have been shown to be implicated in several phenomena such as inflammation, platelet activation, immune response and atherosclerosis. Several studies recently reported PARs expression on neurons and some of them demonstrated that these receptors could interfere with nociception. The contribution of PAR(1) to inflammatory pain and the mechanism involved in this phenomenon were investigated. Intraplantar injection of PAR(1) agonist increased withdrawal latency and reduced response frequency to von Frey filaments, thus inhibiting nociceptive response to both mechanical and thermal stimuli in mice. PAR(1) agonist also reduced carrageenan-induced inflammatory hyperalgesia. The anti-nociceptive effects of PAR(1) agonist were mediated by endogenous opioids, as this effect was inhibited by local injection of naloxone methiodide, and because intraplantar injection of PAR(1) agonist increased mRNA expression of the endogenous opioid precursor proenkephalin. However, PAR(1) agonist was not able to inhibit calcium signals in isolated sensory neurons exposed to pro-nociceptive agents. Finally, despite similar inflammatory parameters, PAR(1)-deficient mice showed a strong potentiation of inflammatory hyperalgesia induced by the intraplantar injection of either formalin or carrageenan, or in the chronic model of collagen-induced arthritis, compared to wild-type mice. This study highlights a previously unknown endogenous mechanism of analgesia, showing a central role for the thrombin receptor PAR(1) in the regulation of inflammatory pain and as an activator of opioid pathways.


Asunto(s)
Inflamación/fisiopatología , Péptidos Opioides/fisiología , Dolor/fisiopatología , Receptores de Trombina/fisiología , Animales , Calcio/metabolismo , Pie , Hiperalgesia/inducido químicamente , Hiperalgesia/patología , Inmunohistoquímica , Inflamación/inducido químicamente , Inflamación/complicaciones , Inyecciones , Masculino , Ratones , Ratones Endogámicos C57BL , Nociceptores/efectos de los fármacos , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacología , Dolor/inducido químicamente , Dolor/etiología , Dimensión del Dolor/efectos de los fármacos , Receptor PAR-1/agonistas , Receptor PAR-1/fisiología , Receptores Opioides/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Receptoras Sensoriales/efectos de los fármacos , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA