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










Base de datos
Intervalo de año de publicación
1.
Mol Ther Oncolytics ; 12: 124-137, 2019 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-30740516

RESUMEN

Use of chimeric antigen receptors (CARs) as the basis of targeted adoptive T cell therapies has enabled dramatic efficacy against multiple hematopoietic malignancies, but potency against bulky and solid tumors has lagged, potentially due to insufficient CAR-T cell expansion and persistence. To improve CAR-T cell efficacy, we utilized a potent activation switch based on rimiducid-inducible MyD88 and CD40 (iMC)-signaling elements. To offset potential toxicity risks by this enhanced CAR, an orthogonally regulated, rapamycin-induced, caspase-9-based safety switch (iRC9) was developed to allow in vivo elimination of CAR-T cells. iMC costimulation induced by systemic rimiducid administration enhanced CAR-T cell proliferation, cytokine secretion, and antitumor efficacy in both in vitro assays and xenograft tumor models. Conversely, rapamycin-mediated iRC9 dimerization rapidly induced apoptosis in a dose-dependent fashion as an approach to mitigate therapy-related toxicity. This novel, regulatable dual-switch system may promote greater CAR-T cell expansion and prolonged persistence in a drug-dependent manner while providing a safety switch to mitigate toxicity concerns.

2.
Mol Ther ; 25(9): 2176-2188, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28697888

RESUMEN

Anti-tumor efficacy of T cells engineered to express chimeric antigen receptors (CARs) is dependent on their specificity, survival, and in vivo expansion following adoptive transfer. Toll-like receptor (TLR) and CD40 signaling in T cells can improve persistence and drive proliferation of antigen-specific CD4+ and CD8+ T cells following pathogen challenge or in graft-versus-host disease (GvHD) settings, suggesting that these costimulatory pathways may be co-opted to improve CAR-T cell persistence and function. Here, we present a novel strategy to activate TLR and CD40 signaling in human T cells using inducible MyD88/CD40 (iMC), which can be triggered in vivo via the synthetic dimerizing ligand, rimiducid, to provide potent costimulation to CAR-modified T cells. Importantly, the concurrent activation of iMC (with rimiducid) and CAR (by antigen recognition) is required for interleukin (IL)-2 production and robust CAR-T cell expansion and may provide a user-controlled mechanism to amplify CAR-T cell levels in vivo and augment anti-tumor efficacy.


Asunto(s)
Antígenos CD28/metabolismo , Antígenos CD40/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Antígenos CD28/genética , Antígenos CD40/genética , Proliferación Celular , Supervivencia Celular , Análisis por Conglomerados , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Inmunoterapia Adoptiva/métodos , Leucemia/genética , Leucemia/inmunología , Leucemia/metabolismo , Leucemia/terapia , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal , Linfocitos T/efectos de los fármacos , Receptores Toll-Like/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
PLoS One ; 7(12): e52666, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23285141

RESUMEN

Activation of complement is one of the earliest immune responses to exogenous threats, resulting in various cleavage products including anaphylatoxin C3a. In addition to its contribution to host defense, C3a has been shown to mediate Th2 responses in animal models of asthma. However, the role of C3a on pulmonary Th17 responses during allergic inflammation remains unclear. Here, we show that mice deficient in C3a receptor (C3aR) exhibited (i) higher percentages of endogenous IL-17-producing CD4(+) T cells in the lungs, (ii) higher amounts of IL-17 in the bronchoalveolar lavage fluid, and (iii) more neutrophils in the lungs than wild-type mice when challenged with intranasal allergens. Moreover, adoptive transfer experiments showed that the frequencies of antigen-specific IL-17-producing CD4(+) T cells were significantly higher in the lungs and bronchial lymph nodes of C3aR-deficient recipients than those of wild-types recipients. Bone-marrow reconstitution study indicated that C3aR-deficiency on hematopoietic cells was required for the increased Th17 responses. Furthermore, C3aR-deficient mice exhibited increased percentages of Foxp3(+) regulatory T cells; however, depletion of these cells minimally affected the induction of antigen-specific Th17 cell population in the lungs. Neutralization of IL-17 significantly reduced the number of neutrophils in bronchoalveolar lavage fluid of C3aR-deficient mice. Our findings demonstrate that C3a signals negatively regulate antigen-specific Th17 responses during allergic lung inflammation and the size of Foxp3(+) regulatory T cell population in the periphery.


Asunto(s)
Anafilatoxinas/inmunología , Complemento C3a/inmunología , Hipersensibilidad/inmunología , Neumonía/inmunología , Células Th17/inmunología , Alérgenos/administración & dosificación , Alérgenos/inmunología , Animales , Complemento C3a/metabolismo , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/metabolismo , Hipersensibilidad/metabolismo , Interleucina-17/biosíntesis , Ratones , Ratones Noqueados , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Neumonía/genética , Receptores de Complemento/genética , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
4.
J Immunol ; 186(2): 1097-106, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21149612

RESUMEN

Adenosine is an extracellular signaling molecule that is generated in response to cell injury where it orchestrates tissue protection and repair. Whereas adenosine is best known for promoting anti-inflammatory activities during acute injury responses, prolonged elevations can enhance destructive tissue remodeling processes associated with chronic disease states. The generation of adenosine and the subsequent activation of the adenosine 2B receptor (A(2B)R) is an important processes in the regulation of both acute and chronic lung disease. The goal of this study was to examine the contribution of the A(2B)R in models of bleomycin-induced lung injury that exhibit varying degrees of acute and chronic injury. Intratracheal bleomycin exposure results in substantial acute lung injury followed by progressive fibrosis. In this model, genetic removal of the A(2B)R resulted in enhanced loss of barrier function and increased pulmonary inflammation, with few differences in indexes of pulmonary fibrosis. These results support an anti-inflammatory role for this receptor in this model of acute lung injury. In contrast, systemic exposure of mice to bleomycin resulted in modest acute lung injury together with progressive pulmonary fibrosis. In this model, the effects of A(2B)R removal on acute lung injury were negligible; however, there were substantial reductions in pulmonary fibrosis, supporting a profibrotic role for this receptor. A(2B)R-dependent regulation of IL-6 production was identified as a potential mechanism involved in the diminished pulmonary fibrosis seen in A(2B)R knockout mice exposed to i.p. bleomycin. These studies highlight the distinct roles of A(2B)R signaling during acute and chronic stages of lung injury.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Bleomicina/toxicidad , Receptor de Adenosina A2B/fisiología , Enfermedad Aguda , Lesión Pulmonar Aguda/patología , Animales , Enfermedad Crónica , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Intubación Intratraqueal , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Receptor de Adenosina A2B/deficiencia , Receptor de Adenosina A2B/genética
5.
Am J Respir Cell Mol Biol ; 42(5): 564-71, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19556606

RESUMEN

Pharmacologic evidence suggests that activation of A(2B) adenosine receptors results in proinflammatory effects relevant to the progression of asthma, a chronic lung disease associated with elevated interstitial adenosine concentrations in the lung. This concept has been challenged by the finding that genetic removal of A(2B) receptors leads to exaggerated responses in models of acute inflammation. Therefore, the goal of our study was to determine the effects of A(2B) receptor gene ablation in the context of ovalbumin-induced chronic pulmonary inflammation. We found that repetitive airway allergen challenge induced a significant increase in adenosine levels in fluid recovered by bronchoalveolar lavage. Genetic ablation of A(2B) receptors significantly attenuated allergen-induced chronic pulmonary inflammation, as evidenced by a reduction in the number of bronchoalveolar lavage eosinophils and in peribronchial eosinophilic infiltration. The most striking difference in the pulmonary inflammation induced in A(2B) receptor knockout (A(2B)KO) and wild-type mice was the lack of allergen-induced IL-4 release in the airways of A(2B)KO animals, in line with a significant reduction in IL-4 protein and mRNA levels in lung tissue. In addition, attenuation of allergen-induced transforming growth factor-beta release in airways of A(2B)KO mice correlated with reduced airway smooth muscle and goblet cell hyperplasia/hypertrophy. In conclusion, genetic removal of A(2B) adenosine receptors in mice leads to inhibition of allergen-induced chronic pulmonary inflammation and airway remodeling. These findings are in agreement with previous pharmacologic studies suggesting a deleterious role for A(2B) receptor signaling in chronic lung inflammation.


Asunto(s)
Neumonía/metabolismo , Neumonía/prevención & control , Receptor de Adenosina A2B/deficiencia , Adenosina/metabolismo , Alérgenos/inmunología , Animales , Bronquios/inmunología , Bronquios/metabolismo , Bronquios/patología , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Enfermedad Crónica , Modelos Animales de Enfermedad , Eosinófilos/patología , Eliminación de Gen , Regulación de la Expresión Génica , Hipertrofia , Interleucina-4/biosíntesis , Metaplasia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Moco/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
6.
Handb Exp Pharmacol ; (193): 215-69, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19639284

RESUMEN

Extracellular adenosine is produced in a coordinated manner from cells following cellular challenge or tissue injury. Once produced, it serves as an autocrine- and paracrine-signaling molecule through its interactions with seven-membrane-spanning G-protein-coupled adenosine receptors. These signaling pathways have widespread physiological and pathophysiological functions. Immune cells express adenosine receptors and respond to adenosine or adenosine agonists in diverse manners. Extensive in vitro and in vivo studies have identified potent anti-inflammatory functions for all of the adenosine receptors on many different inflammatory cells and in various inflammatory disease processes. In addition, specific proinflammatory functions have also been ascribed to adenosine receptor activation. The potent effects of adenosine signaling on the regulation of inflammation suggest that targeting specific adenosine receptor activation or inactivation using selective agonists and antagonists could have important therapeutic implications in numerous diseases. This review is designed to summarize the current status of adenosine receptor signaling in various inflammatory cells and in models of inflammation, with an emphasis on the advancement of adenosine-based therapeutics to treat inflammatory disorders.


Asunto(s)
Inflamación/tratamiento farmacológico , Inflamación/etiología , Receptores Purinérgicos P1/fisiología , Adenosina/metabolismo , Animales , Células Dendríticas/fisiología , Células Endoteliales/fisiología , Humanos , Linfocitos/fisiología , Neutrófilos/fisiología , Antagonistas de Receptores Purinérgicos P1 , Receptores Purinérgicos P1/genética
7.
J Immunol ; 182(12): 8037-46, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494329

RESUMEN

Adenosine is a signaling nucleoside that is generated in response to cellular injury and orchestrates the balance between tissue protection and the progression to pathological tissue remodeling. Adenosine deaminase (ADA)-deficient mice develop progressive airway inflammation and remodeling in association with adenosine elevations, suggesting that adenosine can promote features of chronic lung disease. Furthermore, pharmacological studies in ADA-deficient mice demonstrate that A(2B)R antagonism can attenuate features of chronic lung disease, implicating this receptor in the progression of chronic lung disease. This study examines the contribution of A(2B)R signaling in this model by generating ADA/A(2B)R double-knockout mice. Our hypothesis was that genetic removal of the A(2B)R from ADA-deficient mice would lead to diminished pulmonary inflammation and damage. Unexpectedly, ADA/A(2B)R double-knockout mice exhibited enhanced pulmonary inflammation and airway destruction. Marked loss of pulmonary barrier function and excessive airway neutrophilia are thought to contribute to the enhanced tissue damage observed. These findings support an important protective role for A(2B)R signaling during acute stages of lung disease.


Asunto(s)
Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/metabolismo , Neumonía/inmunología , Neumonía/metabolismo , Receptor de Adenosina A2B/metabolismo , Adenosina Desaminasa/genética , Animales , Moléculas de Adhesión Celular/metabolismo , Colágeno/biosíntesis , Citocinas/biosíntesis , Citocinas/inmunología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/genética , Neumonía/patología , Receptor de Adenosina A2B/deficiencia , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/inmunología
8.
Am J Respir Cell Mol Biol ; 39(6): 697-705, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18587054

RESUMEN

Adenosine is a signaling molecule produced during conditions that cause cellular stress or damage. This signaling pathway is implicated in the regulation of pulmonary disorders through the selective engagement of adenosine receptors. The goal of this study was to examine the involvement of the A(3) adenosine receptor (A(3)R) in a bleomycin model of pulmonary inflammation and fibrosis. Results demonstrated that A(3)R-deficient mice exhibit enhanced pulmonary inflammation that included an increase in eosinophils. Accordingly, there was a selective up-regulation of eosinophil-related chemokines and cytokines in the lungs of A(3)R-deficient mice exposed to bleomycin. This increase in eosinophil numbers was accompanied by a decrease in the amount of extracellular eosinophil peroxidase activity in lavage fluid from A(3)R-deficient mice exposed to bleomycin, an observation suggesting that the A(3)R is necessary for eosinophil degranulation in this model. Despite an increase in inflammatory metrics associated with A(3)R-deficient mice treated with bleomycin, there was little difference in the degree of pulmonary fibrosis. Examination of fibrotic mediators demonstrated enhanced transforming growth factor (TGF)-beta1 expression, but not a concomitant increase in TGF-beta1 activity. This was associated with the loss of expression of matrix metalloprotease 9, an activator of TGF-beta1, in alveolar macrophages and airway mast cells in the lungs of A(3)R-deficient mice. Together, these results suggest that the A(3)R serves antiinflammatory functions in the bleomycin model, and is also involved in regulating the production of mediators that can impact fibrosis.


Asunto(s)
Neumonía/complicaciones , Neumonía/metabolismo , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/metabolismo , Receptor de Adenosina A3/metabolismo , Transducción de Señal , Animales , Bleomicina , Movimiento Celular , Eosinófilos/patología , Femenino , Regulación de la Expresión Génica , Pulmón/enzimología , Pulmón/patología , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
9.
J Clin Invest ; 116(8): 2173-2182, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16841096

RESUMEN

Adenosine has been implicated in the pathogenesis of chronic lung diseases such as asthma and chronic obstructive pulmonary disease. In vitro studies suggest that activation of the A2B adenosine receptor (A2BAR) results in proinflammatory and profibrotic effects relevant to the progression of lung diseases; however, in vivo data supporting these observations are lacking. Adenosine deaminase-deficient (ADA-deficient) mice develop pulmonary inflammation and injury that are dependent on increased lung adenosine levels. To investigate the role of the A2BAR in vivo, ADA-deficient mice were treated with the selective A2BAR antagonist CVT-6883, and pulmonary inflammation, fibrosis, and airspace integrity were assessed. Untreated and vehicle-treated ADA-deficient mice developed pulmonary inflammation, fibrosis, and enlargement of alveolar airspaces; conversely, CVT-6883-treated ADA-deficient mice showed less pulmonary inflammation, fibrosis, and alveolar airspace enlargement. A2BAR antagonism significantly reduced elevations in proinflammatory cytokines and chemokines as well as mediators of fibrosis and airway destruction. In addition, treatment with CVT-6883 attenuated pulmonary inflammation and fibrosis in wild-type mice subjected to bleomycin-induced lung injury. These findings suggest that A2BAR signaling influences pathways critical for pulmonary inflammation and injury in vivo. Thus in chronic lung diseases associated with increased adenosine, antagonism of A2BAR-mediated responses may prove to be a beneficial therapy.


Asunto(s)
Adenosina/efectos adversos , Enfermedades Pulmonares/fisiopatología , Lesión Pulmonar , Receptor de Adenosina A2B/fisiología , Animales , Inflamación/inducido químicamente , Inflamación/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Enfermedades Pulmonares/inducido químicamente , Ratones , Ratones Noqueados , Purinas/farmacología , Pirazoles/farmacología , Receptor de Adenosina A2B/deficiencia , Receptor de Adenosina A2B/genética , Transducción de Señal , Transcripción Genética
10.
Eur J Pharmacol ; 498(1-3): 195-202, 2004 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-15363995

RESUMEN

We examined the involvement of endogenous vasopressin and platelet-activating factor (PAF) in the pathogenesis of two types of experimental angina in urethane-anaesthetised male Wistar rats. In the first model, epinephrine (10 microg kg(-1)) was injected into the tail vein, followed at the development of the maximum blood pressure response, i.e., 30 s later, by phentolamine (15 mg kg(-1)). In the second model, the vasopressin V1 receptor agonist ornithine-vasopressin (ornipressin; 0.5 IU kg(-1), i.v.) was administered. The heart rate, mean arterial blood pressure and surface electrocardiogram (ECG, standard lead II) were registered simultaneously. As a measure of myocardial ischaemia, at 1 min after phentolamine or ornipressin administration, we found significant ST-segment depression, lasting for more than 10 or 5 min, respectively. Pretreatment (15 min, s.c.) with the vasopressin V1 receptor antagonist Mca1,Tyr(Me)2AVP (the Manning peptide; 0.02-0.2 microg kg(-1)) or the platelet-activating factor receptor antagonist ginkgolide B (BN 52021; 0.25-2.5 mg kg(-1)) alone caused a dose-dependent reduction of the ST-segment depression. Concurrent administration of the two antagonists in their threshold doses (0.02 microg kg(-1) and 0.25 mg kg(-1)) also attenuated the ST-segment depression in both models. Neither antagonist affected the blood pressure or heart rate changes throughout the studies. Our results suggest that endogenous vasopressin and platelet-activating factor interact synergistically in provoking myocardial ischaemia in vivo in experimental angina in the rat.


Asunto(s)
Angina de Pecho/metabolismo , Arginina Vasopresina/análogos & derivados , Factor de Activación Plaquetaria/metabolismo , Vasopresinas/metabolismo , Angina de Pecho/etiología , Animales , Antagonistas de los Receptores de Hormonas Antidiuréticas , Arginina Vasopresina/administración & dosificación , Arginina Vasopresina/farmacología , Presión Sanguínea/efectos de los fármacos , Diterpenos/administración & dosificación , Diterpenos/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Electrocardiografía , Epinefrina/administración & dosificación , Epinefrina/farmacología , Ginkgólidos , Frecuencia Cardíaca/efectos de los fármacos , Antagonistas de Hormonas/administración & dosificación , Antagonistas de Hormonas/farmacología , Inyecciones Intravenosas , Lactonas/administración & dosificación , Lactonas/farmacología , Masculino , Isquemia Miocárdica/inducido químicamente , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/fisiopatología , Ornipresina/administración & dosificación , Ornipresina/farmacología , Fentolamina/administración & dosificación , Fentolamina/farmacología , Factor de Activación Plaquetaria/antagonistas & inhibidores , Ratas , Ratas Wistar , Receptores de Vasopresinas/agonistas , Factores de Tiempo , Vasoconstrictores/administración & dosificación , Vasoconstrictores/farmacología
11.
Eur J Pharmacol ; 495(2-3): 179-84, 2004 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-15249168

RESUMEN

We studied the effects of a 2-week period of oral raloxifene therapy on the cardiac level of nitric oxide (NO) and on the susceptibility to angina in ovariectomized rats. Ovariectomy decreased the activity of Ca2+-dependent nitric oxide synthase (NOS) in the left ventricle, an effect restored by raloxifene (0.2-5 mg kg(-1) day(-1)) or 17beta-oestradiol (0.3 mg kg(-1) day(-1)). Ovariectomy led to a significant ST segment depression after the injection of (1) ornithine-vasopressin (0.5 IU kg(-1), i.v.) or (2) epinephrine (10 microg kg(-1), i.v.), followed 30 s later by phentolamine (15 mg kg(-1), i.v.); both effects were reversed by raloxifene or 17beta-oestradiol treatment. Inhibition of nitric oxide synthase (with NG-nitro-L-arginine methyl ester [L-NAME]; 5 mg kg(-1), s.c.) augmented the ST segment depression in the ovariectomized rat and abolished the anti-ischaemic effect of 17beta-oestradiol or raloxifene. Thus, an oestrogen deficiency down-regulates the cardiac constitutive nitric oxide synthase, which increases the susceptibility of the heart to ishaemia because both actions can be blocked by exogenous administration of the natural oestrogen 17beta-oestradiol or the selective oestrogen-receptor modulator (SERM) raloxifene. In the present in vivo system, raloxifene exerts oestrogen-agonist properties.


Asunto(s)
Isquemia Miocárdica/prevención & control , Miocardio/metabolismo , Óxido Nítrico/biosíntesis , Clorhidrato de Raloxifeno/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Administración Oral , Animales , Presión Sanguínea/efectos de los fármacos , Calcio/metabolismo , Inhibidores Enzimáticos/farmacología , Epinefrina/farmacología , Estradiol/farmacología , Femenino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Ornipresina/farmacología , Ovariectomía , Ratas , Ratas Wistar , Factores de Tiempo
12.
Pancreas ; 29(2): 157-61, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15257108

RESUMEN

OBJECTIVES: Ischemia-reperfusion injury can be involved in the pathophysiology of acute necrotizing pancreatitis. The aim of our study was to determine the production of cytokines, tumor necrosis factor (TNF) and interleukin-6 (IL-6), the activation of the inducible nitric oxide synthase (iNOS), and the development of apoptosis during this pathologic process. METHODS: Acute pancreatitis was produced in male Wistar rats by injection of 200 microL of 6% taurocholic acid into the main pancreatic duct in combination with the temporary (15 minutes) occlusion of the inferior splenic artery. Six and 24 hours later, the histologic damage was evaluated, and serum amylase, TNF, IL-6 levels, and iNOS and apoptotic activity from pancreatic and pulmonary tissues were determined. RESULTS: Twenty-four hours after the induction of pancreatitis, the mortality rate was 63%. During this period, the serum TNF and IL-6 levels were permanently high (50 +/- 12 and 58 +/- 10 U/mL and 7083 +/- 1610 and 6790 +/- 850 U/mL after 6 and 24 hours, respectively). The iNOS activity showed an increasing tendency in the pancreas, and a decrease following an initial increase in the lung (from 4.2 +/- 0.6 to 5 +/- 0.4 and from 6.8 +/- 0.6 to 3.8 +/- 0.5 pmol/min/mg protein after 6 and 24 hours, respectively). Histologic examination confirmed severe necrotizing pancreatitis. In the pancreas, the apoptotic activity increased significantly (from 4 +/- 4 to 27 +/- 5/mm at 6 and 24 hours), while in the lungs, following an initial increase it declined during the course of necrotizing pancreatitis (from 49 +/- 4 to 11 +/- 6/mm at 6 and 24 hours). CONCLUSION: Our results indicate that intraductal taurocholic acid and ischemia-reperfusion provokes severe acute necrotizing pancreatitis with a high mortality rate and leads to systemic inflammatory reaction, which appears to be the consequence of the activation of the cytokine cascade and iNOS. The degree of NO overproduction by iNOS corresponds with the apoptotic process in the pancreas and the lung.


Asunto(s)
Apoptosis , Citocinas/fisiología , Óxido Nítrico/fisiología , Pancreatitis Aguda Necrotizante/fisiopatología , Animales , Interleucina-6/sangre , Isquemia/complicaciones , Isquemia/fisiopatología , Pulmón/metabolismo , Pulmón/patología , Masculino , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Páncreas/irrigación sanguínea , Páncreas/metabolismo , Páncreas/patología , Pancreatitis Aguda Necrotizante/inducido químicamente , Pancreatitis Aguda Necrotizante/etiología , Pancreatitis Aguda Necrotizante/metabolismo , Ratas , Ratas Wistar , Daño por Reperfusión/fisiopatología , Síndrome de Respuesta Inflamatoria Sistémica/etiología , Síndrome de Respuesta Inflamatoria Sistémica/fisiopatología , Ácido Taurocólico/metabolismo , Ácido Taurocólico/toxicidad , Factor de Necrosis Tumoral alfa/análisis
13.
Pancreas ; 25(3): 277-82, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12370539

RESUMEN

INTRODUCTION: Nitric oxide (NO) has been implicated in the regulation of the pancreatic circulation, the promotion of the capillary integrity, and the inhibition of leukocyte adhesion. AIMS: To investigate the rates of changes in the pancreatic constitutive NO synthase (cNOS) and inducible NOS (iNOS) activities and the role of NO in the vascular permeability changes during the development of L-arginine (Arg)-induced acute pancreatitis. METHODOLOGY: Acute pancreatitis was induced in male Wistar rats by injecting 250 mg/100 g body weight of Arg i.p. twice at an interval of 1 hour, as a 20% solution in 0.15 NaCl (group I). The control rats received the same quantity of glycine (group II). In group III, 30 mg/kg N -nitro-L-arginine methyl ester (L-NAME) was injected i.p. 19 hours after the first Arg injection. The rats were killed at 6, 12, 24, or 48 hours following Arg administration, and the plasma amylase concentration and the pancreatic weight/body weight (pw/bw) ratios were evaluated. NOS activity was determined via the conversion of L- C-Arg monohydrochloride to C-citrulline. The vascular permeability was examined by means of the extravasation of Evans blue dye (20 mg/kg bw) into the pancreatic tissue. RESULTS: The serum amylase level was already increased at 6 hours in group I animals, peaked at 12 hours after the Arg injection (11.800 +/- 590 versus 6.618 +/- 252 U/L in group II), and returned to the control level at 48 hours. The pw/bw ratio peaked at 24 hours in group I (6.63 +/- 0.52 versus 4.02 +/- 0.22 mg/g in group II) and returned to the control level at 48 hours. The cNOS activity was depleted at 6 hours in group I (0.02 +/- 0.003 versus 0.23 +/- 0.02 pmol/min/mg protein in group II); it then gradually increased to a level significantly higher than that in group II and decreased thereafter (0.45 +/- 0.03 and 0.13 +/- 0.01 pmol/min/mg protein at 24 and 48 hours). The iNOS activity was significantly increased at 24 and 48 hours versus that in group II (0.15 +/- 0.05 and 0.07 +/- 0.01 versus 0.04 +/- 0.01 pmol/min/mg protein). The pancreatic concentration of Evans blue dye was significantly higher in group I than in group II (138.59 +/- 11.04 versus 43.57 +/- 2.67 (g/dry weight). Treatment with L-NAME significantly reduced the amylase activity, pw/bw, Evans blue concentration, and cNOS activity of the pancreas but did not exert any beneficial effect on the histologic score at 24 hours after the onset of pancreatitis, as compared with those values in group I (6.528 +/- 673 U/L, 4.56 +/- 0.65 mg/g, 86.84 +/- 3.9 (g/dry weight, 0.14 +/- 0.04 pmol/min/mg protein). CONCLUSION: Endogenous NO is involved in the formation of pancreatic edema in Arg-induced acute pancreatitis by increasing the vascular permeability and protein extravasation. L-NAME treatment decreased the cNOS activity and edema formation but did not prevent the histologic damage in Arg-induced acute pancreatitis.


Asunto(s)
Edema/etiología , Óxido Nítrico/fisiología , Pancreatitis/etiología , Enfermedad Aguda , Amilasas/sangre , Animales , Arginina , Permeabilidad Capilar , Colorantes/metabolismo , Edema/enzimología , Edema/metabolismo , Edema/patología , Inhibidores Enzimáticos/farmacología , Azul de Evans/metabolismo , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Pancreatitis/inducido químicamente , Pancreatitis/enzimología , Pancreatitis/metabolismo , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA