Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Respir Cell Mol Biol ; 51(3): 413-25, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24684441

ABSTRACT

NF-κB and IL-6, a NF-κB downstream mediator, play a central role in the inflammatory response of tissues. We aimed to determine the role of the classical NF-κB pathway in severe pulmonary arterial hypertension (PAH) induced by SU5416 and chronic hypoxia (SuHx) in rats. Tissue samples from patients with idiopathic PAH (iPAH) and control subjects were investigated. SuHx rats were treated from Days 1 to 3, 1 to 21, and 29 to 42 with the NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) and/or from Days 1 to 21 with anti-IL-6 antibody. Nuclear staining for NF-κB, an indicator of the activation of the classical NF-κB pathway, was detected in pulmonary arterial lesions of patients with iPAH and SuHx rats. NF-κB inhibition with PDTC prevented and reduced pulmonary arterial obliteration without reducing muscularization. However, the elevated lung levels of IL-6 were not reduced in PDTC-treated SuHx animals. PDTC treatment prevented or reduced apoptosis of pulmonary artery wall cells and pulmonary arterial obliteration. IL-6 inhibition had only a partial effect on apoptosis and obliteration. Pulmonary arterial media wall thickness was not affected by any of these treatments. Preventive and therapeutic PDTC treatment promoted immune regulation by increasing the number of perivascular CD4(+) T cells, in particular regulatory T cells (early treatment), and by reducing the number of perivascular CD8(+) T lymphocytes and CD45RA(+) B lymphocytes. Therapeutic PDTC treatment further preserved right ventricular function in SuHx animals. Inhibition of NF-κB may represent a therapeutic option for pulmonary arterial obliteration via reduced vessel wall cell apoptosis and improved regulation of the immune system.


Subject(s)
Hypertension, Pulmonary/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Animals , Apoptosis , CD4-Positive T-Lymphocytes/metabolism , Familial Primary Pulmonary Hypertension , Humans , Hypertension, Pulmonary/pathology , Hypoxia/metabolism , Indoles/chemistry , Inflammation , Interleukin-6/metabolism , Leukocyte Common Antigens/metabolism , Lung/pathology , Pulmonary Artery/pathology , Pyrroles/chemistry , Pyrrolidines/chemistry , Rats , Signal Transduction , Thiocarbamates/chemistry , Time Factors
2.
PLoS One ; 9(2): e89810, 2014.
Article in English | MEDLINE | ID: mdl-24587052

ABSTRACT

Successful curative treatment of severe pulmonary arterial hypertension with luminal obliteration will require a thorough understanding of the mechanism underlying the development and progression of pulmonary vascular lesions. But the cells that obliterate the pulmonary arterial lumen in severe pulmonary arterial hypertension are incompletely characterized. The goal of our study was to evaluate whether inhibition of CXC chemokine receptor 4 will prevent the accumulation of c-kit⁺ cells and severe pulmonary arterial hypertension. We detected c-kit⁺⁻ cells expressing endothelial (von Willebrand Factor) or smooth muscle cell/myofibroblast (α-smooth muscle actin) markers in pulmonary arterial lesions of SU5416/chronic hypoxia rats. We found increased expression of CXC chemokine ligand 12 in the lung tissue of SU5416/chronic hypoxia rats. In our prevention study, AMD3100, an inhibitor of the CXC chemokine ligand 12 receptor, CXC chemokine receptor 4, only moderately decreased pulmonary arterial obliteration and pulmonary hypertension in SU5416/chronic hypoxia animals. AMD3100 treatment reduced the number of proliferating c-kit⁺ α-smooth muscle actin⁺ cells and pulmonary arterial muscularization and did not affect c-kit⁺ von Willebrand Factor⁺ cell numbers. Both c-kit⁺ cell types expressed CXC chemokine receptor 4. In conclusion, our data demonstrate that in the SU5416/chronic hypoxia model of severe pulmonary hypertension, the CXC chemokine receptor 4-expressing c-kit⁺ α-smooth muscle actin⁺ cells contribute to pulmonary arterial muscularization. In contrast, vascular lumen obliteration by c-kit⁺ von Willebrand Factor⁺ cells is largely independent of CXC chemokine receptor 4.


Subject(s)
Arterial Occlusive Diseases/complications , Arterial Occlusive Diseases/drug therapy , Heterocyclic Compounds/pharmacology , Hypertension, Pulmonary/prevention & control , Proto-Oncogene Proteins c-kit/metabolism , Receptors, CXCR4/antagonists & inhibitors , Actins/metabolism , Analysis of Variance , Animals , Benzylamines , Cyclams , Fluorescent Antibody Technique , Hypertension, Pulmonary/etiology , Immunohistochemistry , In Situ Hybridization , Indoles , Microscopy, Confocal , Pyrroles , Rats , Receptors, CXCR4/metabolism , von Willebrand Factor/metabolism
3.
Chest ; 142(3): 663-672, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22383663

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) are small noncoding RNAs that silence target gene expression posttranscriptionally, and their impact on gene expression has been reported in various diseases. It has been reported that the expression of the hypoxia-inducible factor-1α (HIF-1α) is reduced and that of p53 is increased in lungs from patients with COPD. However, the role of miRNAs associated with these genes in lungs from patients with COPD is unknown. METHODS: Lung tissue samples from 55 patients were included in this study. Total RNA, miRNA, and protein were extracted from lung tissues and used for reverse transcriptase polymerase chain reaction and Western blot analysis. Cell culture experiments were performed using cultured human pulmonary microvascular endothelial cells (HPMVECs). RESULTS: miR-34a and miR-199a-5p expressions were increased, and the phosphorylation of AKT was decreased in the lung tissue samples of patients with COPD. The miR-199a-5p expression was correlated with HIF-1α protein expression in the lungs of patients with COPD. Transfection of HPMVECs with the miR-199a-5p precursor gene decreased HIF-1α protein expression, and transfection with the miR-34a precursor gene increased miR-199a-5p expression. CONCLUSIONS: These data suggest that miR-34a and miR-199a-5p contribute to the pathogenesis of COPD, and these miRNAs may also affect the HIF-1α-dependent lung structure maintenance program.


Subject(s)
Gene Expression Regulation/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/metabolism , MicroRNAs/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Severity of Illness Index , Adult , Aged , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Lung/pathology , Male , Middle Aged , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/pathology , Transfection , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/metabolism
4.
PLoS One ; 7(1): e30678, 2012.
Article in English | MEDLINE | ID: mdl-22276220

ABSTRACT

BACKGROUND: Copper is an important regulator of hypoxia inducible factor 1 alpha (HIF-1α) dependent vascular endothelial growth factor (VEGF) expression, and is also required for the activity of lysyl oxidase (LOX) to effect matrix protein cross-linking. Cell detachment from the extracellular matrix can induce apoptosis (anoikis) via inactivation of focal adhesion kinase (FAK). METHODOLOGY: To examine the molecular mechanisms whereby copper depletion causes the destruction of the normal alveolar architecture via anoikis, Male Sprague-Dawley rats were fed a copper deficient diet for 6 weeks while being treated with the copper chelator, tetrathiomolybdate. Other groups of rats were treated with the inhibitor of auto-phosphorylation of FAK, 1,2,4,5-benzenetetraamine tetrahydrochloride (1,2,4,5-BT) or FAK small interfering RNA (siRNA). PRINCIPAL FINDINGS: Copper depletion caused emphysematous changes, decreased HIF-1α activity, and downregulated VEGF expression in the rat lungs. Cleaved caspase-3, caspase-8 and Bcl-2 interacting mediator of cell death (Bim) expression was increased, and the phosphorylation of FAK was decreased in copper depleted rat lungs. Administration of 1,2,4,5-BT and FAK siRNA caused emphysematous lung destruction associated with increased expression of cleaved capase-3, caspase-8 and Bim. CONCLUSIONS: These data indicate that copper-dependent mechanisms contribute to the pathogenesis of emphysema, which may be associated with decreased HIF-1α and FAK activity in the lung.


Subject(s)
Copper/deficiency , Emphysema/enzymology , Emphysema/etiology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Aniline Compounds/toxicity , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Blotting, Western , Caspase 3/genetics , Caspase 3/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Emphysema/genetics , Focal Adhesion Protein-Tyrosine Kinases/genetics , Immunohistochemistry , Immunoprecipitation , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering , Rats , Real-Time Polymerase Chain Reaction
5.
Am J Physiol Lung Cell Mol Physiol ; 302(4): L363-9, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-21964406

ABSTRACT

Severe forms of pulmonary arterial hypertension (PAH) are characterized by various degrees of remodeling of the pulmonary arterial vessels, which increases the pulmonary vascular resistance and right ventricular afterload, thus contributing to the development of right ventricle dysfunction and failure. Recent years have seen advances in the understanding of the pathobiology of PAH; however, many important questions remain unanswered. Elucidating the pathobiology of PAH continues to be critical to design new effective therapeutic strategies, and appropriate animal models of PAH are necessary to achieve the task. Although the monocrotaline rat model of PAH has contributed to a better understanding of vascular remodeling in pulmonary hypertension, we question the validity of this model as a preclinically relevant model of severe plexogenic PAH. Here we review pertinent publications that either have been forgotten or ignored, and we reexamine the monocrotaline model in the context of human forms of PAH.


Subject(s)
Hypertension, Pulmonary/chemically induced , Acute Lung Injury/chemically induced , Acute Lung Injury/physiopathology , Animals , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/physiopathology , Disease Models, Animal , Humans , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Lung/blood supply , Lung/pathology , Monocrotaline , Myocarditis/chemically induced , Myocarditis/pathology , Myocarditis/physiopathology
6.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L753-61, 2011 May.
Article in English | MEDLINE | ID: mdl-21335523

ABSTRACT

Chronic hypoxia induces pulmonary arterial remodeling, resulting in pulmonary hypertension and right ventricular hypertrophy. Hypoxia has been implicated as a physiological stimulus for p53 induction and hypoxia-inducible factor-1α (HIF-1α). However, the subcellular interactions between hypoxic exposure and expression of p53 and HIF-1α remain unclear. To examine the role of p53 and HIF-1α expression on hypoxia-induced pulmonary arterial remodeling, wild-type (WT) and p53 knockout (p53KO) mice were exposed to either normoxia or hypoxia for 8 wk. Following chronic hypoxia, both genotypes demonstrated elevated right ventricular pressures, right ventricular hypertrophy as measured by the ratio of the right ventricle to the left ventricle plus septum weights, and vascular remodeling. However, the right ventricular systolic pressures, the ratio of the right ventricle to the left ventricle plus septum weights, and the medial wall thickness of small vessels were significantly greater in the p53KO mice than in the WT mice. The p53KO mice had lower levels of p21 and miR34a expression, and higher levels of HIF-1α, VEGF, and PDGF expression than WT mice following chronic hypoxic exposure. This was associated with a higher proliferating cell nuclear antigen expression of pulmonary artery in p53KO mice. We conclude that p53 plays a critical role in the mitigation of hypoxia-induced small pulmonary arterial remodeling. By interacting with p21 and HIF-1α, p53 may suppress hypoxic pulmonary arterial remodeling and pulmonary arterial smooth muscle cell proliferation under hypoxia.


Subject(s)
Genes, p53/physiology , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Animals , Hypertension, Pulmonary/etiology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/complications , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Knockout , MicroRNAs/biosynthesis , p21-Activated Kinases/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL