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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Respir Cell Mol Biol ; 49(4): 619-26, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23713977

RESUMEN

The release of hemoglobin (Hb) with hemolysis causes vascular dysfunction. New evidence implicates Hb-induced NF-κB and hypoxia inducible factor (HIF) activation, which may be under the control of a Toll-like receptor (TLR)-signaling pathway. Nearly all TLR-signaling pathways activate the myeloid differentiation primary response gene-88 (MyD88) that regulates NF-κB. We hypothesized that the differing transition states of Hb influence endothelial cell permeability via NF-κB activation and HIF regulation through a MyD88-dependent pathway. In cultured human dermal microvascular endothelial cells (HMECs-1), we examined the effects of Hb in the ferrous (HbFe(2+)), ferric (HbFe(3+)), and ferryl (HbFe(4+)) transition states on NF-κB and HIF activity, HIF-1α and HIF-2α mRNA up-regulation, and monolayer permeability, in the presence or absence of TLR4, MyD88, NF-κB, or HIF inhibition, as well as superoxide dismutase (SOD) and catalase. Our data showed that cell-free Hb, in each transition state, induced NF-κB and HIF activity, up-regulated HIF-1α and HIF-2α mRNA, and increased HMEC-1 permeability. The blockade of either MyD88 or NF-κB, but not TLR4, attenuated Hb-induced HIF activity, the up-regulation HIF-1 and HIF-2α mRNA, and HMEC-1 permeability. The inhibition of HIF activity exerted less of an effect on Hb-induced monolayer permeability. Moreover, SOD and catalase attenuated NF-κB, HIF activity, and monolayer permeability. Our results demonstrate that Hb-induced NF-κB and HIF are regulated by two mechanisms, either MyD88 activation or Hb transition state-induced ROS formation, that influence HMEC-1 permeability.


Asunto(s)
Células Endoteliales/metabolismo , Hemoglobinas/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Catalasa/genética , Catalasa/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular , Hemoglobinas/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor 88 de Diferenciación Mieloide/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Oxidación-Reducción , Permeabilidad , ARN Mensajero/genética , Transducción de Señal , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Regulación hacia Arriba
2.
Am J Physiol Lung Cell Mol Physiol ; 303(4): L312-26, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22728465

RESUMEN

Cell-free hemoglobin (Hb) exposure may be a pathogenic mediator in the development of pulmonary arterial hypertension (PAH), and when combined with chronic hypoxia the potential for exacerbation of PAH and vascular remodeling is likely more pronounced. We hypothesized that Hb may contribute to hypoxia-driven PAH collectively as a prooxidant, inflammatory, and nitric oxide (NO) scavenger. Using programmable micropump technology, we exposed male Sprague-Dawley rats housed under room air or hypoxia to 12 or 30 mg per day Hb for 3, 5, and 7 wk. Blood pressure, cardiac output, right ventricular hypertrophy, and indexes of pulmonary vascular remodeling were evaluated. Additionally, markers of oxidative stress, NO bioavailability and inflammation were determined. Hb increased pulmonary arterial (PA) pressure, pulmonary vessel wall stiffening, and right heart hypertrophy with temporal and dose dependence in both room air and hypoxic cohorts. Hb induced a modest increase in plasma oxidative stress markers (malondialdehyde and 4-hydroxynonenal), no change in NO bioavailability, and increased lung ICAM protein expression. Treatment with the antioxidant Tempol attenuated Hb-induced pulmonary arterial wall thickening, but not PA pressures or ICAM expression. Chronic exposure to low plasma Hb concentrations (range = 3-10 µM) lasting up to 7 wk in rodents induces pulmonary vascular disease via inflammation and to a lesser extent by Hb-mediated oxidation. Tempol demonstrated a modest effect on the attenuation of Hb-induced pulmonary vascular disease. NO bioavailability was found to be of minimal importance in this model.


Asunto(s)
Hemoglobinas/efectos adversos , Inflamación/patología , Enfermedades Pulmonares/inducido químicamente , Enfermedades Vasculares/inducido químicamente , Animales , Presión Sanguínea/efectos de los fármacos , Western Blotting , Gasto Cardíaco/efectos de los fármacos , Óxidos N-Cíclicos/farmacología , Hemodinámica/efectos de los fármacos , Hemoglobinas/administración & dosificación , Hemoglobinas/farmacología , Humanos , Peróxido de Hidrógeno/farmacología , Inflamación/complicaciones , Bombas de Infusión , Molécula 1 de Adhesión Intercelular/metabolismo , Riñón/efectos de los fármacos , Riñón/patología , Riñón/fisiopatología , Peroxidación de Lípido/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Enfermedades Pulmonares/sangre , Enfermedades Pulmonares/patología , Enfermedades Pulmonares/orina , Masculino , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Ratas , Ratas Sprague-Dawley , Marcadores de Spin , Enfermedades Vasculares/sangre , Enfermedades Vasculares/patología , Enfermedades Vasculares/orina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA