Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 309(4): L378-87, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26071556

RESUMEN

Pulmonary hypertension is a serious condition that can lead to premature death. The mechanisms involved are incompletely understood although a role for the immune system has been suggested. Inflammasomes are part of the innate immune system and consist of the effector caspase-1 and a receptor, where nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) is the best characterized and interacts with the adaptor protein apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC). To investigate whether ASC and NLRP3 inflammasome components are involved in hypoxia-induced pulmonary hypertension, we utilized mice deficient in ASC and NLRP3. Active caspase-1, IL-18, and IL-1ß, which are regulated by inflammasomes, were measured in lung homogenates in wild-type (WT), ASC(-/-), and NLRP3(-/-) mice, and phenotypical changes related to pulmonary hypertension and right ventricular remodeling were characterized after hypoxic exposure. Right ventricular systolic pressure (RVSP) of ASC(-/-) mice was significantly lower than in WT exposed to hypoxia (40.8 ± 1.5 mmHg vs. 55.8 ± 2.4 mmHg, P < 0.001), indicating a substantially reduced pulmonary hypertension in mice lacking ASC. Magnetic resonance imaging further supported these findings by demonstrating reduced right ventricular remodeling. RVSP of NLRP3(-/-) mice exposed to hypoxia was not significantly altered compared with WT hypoxia. Whereas hypoxia increased protein levels of caspase-1, IL-18, and IL-1ß in WT and NLRP3(-/-) mice, this response was absent in ASC(-/-) mice. Moreover, ASC(-/-) mice displayed reduced muscularization and collagen deposition around arteries. In conclusion, hypoxia-induced elevated right ventricular pressure and remodeling were attenuated in mice lacking the inflammasome adaptor protein ASC, suggesting that inflammasomes play an important role in the pathogenesis of pulmonary hypertension.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Hipertensión Pulmonar/metabolismo , Inflamasomas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Arterias/patología , Proteínas Adaptadoras de Señalización CARD , Hipoxia de la Célula , Colágeno/metabolismo , Expresión Génica , Hipertrofia Ventricular Derecha/metabolismo , Interleucina-18/sangre , Leucocitos/inmunología , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Remodelación Ventricular
2.
Exp Lung Res ; 38(8): 406-19, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22908875

RESUMEN

Interleukin (IL)-18 is a pro-inflammatory cytokine suggested to be involved in the development of pulmonary emphysema and inflammation. Studies involving immunology and cancer have revealed that IL-18 can have synergistic effects with IL-12. We have studied the presence of IL-18 and IL-12 receptors (IL-18R/IL-12R) in the lungs and whether IL-18 and IL-12, alone or in combination, have the ability to initiate the induction of mediators related to the development of emphysema and inflammation. The expression of the IL-18R was abundant in lungs compared to other organs (heart, liver, and spleen), and the IL-12R was also expressed in lung tissue. Mice treated with i.p. injection of recombinant murine IL-18 or IL-12 expressed significantly higher pulmonary mRNA levels of the matrix degrading enzymes metalloproteinase (MMP) 12 and cathepsin S, in addition to interferon-γ, tumor necrosis factor-α, and CXC chemokine ligand 9 (CXCL9) (all P < .05) than controls (received PBS). Treatment with IL-18 and IL-12 in combination showed an even more pronounced induction of these mediators, as well as a significant increase in MMP-9, IL-6, IL-1ß, and transforming growth factor-ß (P < .05). Furthermore, cellular apoptosis in lung tissue was induced. Immunohistochemical analysis revealed T-cell infiltration in pulmonary vessels following co-stimulation. In summary, IL-18 and IL-12 exert a synergistic effect on the lungs by inducing MMPs, cathepsins S, and pro-inflammatory cytokines, which may promote pulmonary emphysema and inflammation. The synergy between IL-18 and IL-12 involves infiltration of T-cells in the lungs, possibly induced by the T-cell chemoattractant CXCL9.


Asunto(s)
Catepsinas/biosíntesis , Interleucina-12/farmacología , Interleucina-18/farmacología , Pulmón/efectos de los fármacos , Metaloproteinasa 12 de la Matriz/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Catepsinas/genética , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Quimioterapia Combinada , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Metaloproteinasa 12 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Receptores de Interleucina-12/metabolismo , Receptores de Interleucina-18/metabolismo , Proteínas Recombinantes/farmacología , Linfocitos T/metabolismo , Linfocitos T/patología , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...