Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Respir Cell Mol Biol ; 49(4): 619-26, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23713977

ABSTRACT

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.


Subject(s)
Endothelial Cells/metabolism , Hemoglobins/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Catalase/genetics , Catalase/metabolism , Cell Line , Cell Membrane Permeability , Hemoglobins/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myeloid Differentiation Factor 88/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidation-Reduction , Permeability , RNA, Messenger/genetics , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Up-Regulation
2.
Am J Physiol Lung Cell Mol Physiol ; 303(4): L312-26, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22728465

ABSTRACT

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.


Subject(s)
Hemoglobins/adverse effects , Inflammation/pathology , Lung Diseases/chemically induced , Vascular Diseases/chemically induced , Animals , Blood Pressure/drug effects , Blotting, Western , Cardiac Output/drug effects , Cyclic N-Oxides/pharmacology , Hemodynamics/drug effects , Hemoglobins/administration & dosage , Hemoglobins/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Inflammation/complications , Infusion Pumps , Intercellular Adhesion Molecule-1/metabolism , Kidney/drug effects , Kidney/pathology , Kidney/physiopathology , Lipid Peroxidation/drug effects , Lung/drug effects , Lung/pathology , Lung/physiopathology , Lung Diseases/blood , Lung Diseases/pathology , Lung Diseases/urine , Male , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , Spin Labels , Vascular Diseases/blood , Vascular Diseases/pathology , Vascular Diseases/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...