Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Am J Respir Crit Care Med ; 204(12): 1433-1451, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34550870

ABSTRACT

Rationale: Mechanical signaling through cell-matrix interactions plays a major role in progressive vascular remodeling in pulmonary arterial hypertension (PAH). MMP-8 (matrix metalloproteinase-8) is an interstitial collagenase involved in regulating inflammation and fibrosis of the lung and systemic vasculature, but its role in PAH pathogenesis remains unexplored. Objectives: To evaluate MMP-8 as a modulator of pathogenic mechanical signaling in PAH. Methods: MMP-8 levels were measured in plasma from patients with pulmonary hypertension (PH) and controls by ELISA. MMP-8 vascular expression was examined in lung tissue from patients with PAH and rodent models of PH. MMP-8-/- and MMP-8+/+ mice were exposed to normobaric hypoxia or normoxia for 4-8 weeks. PH severity was evaluated by right ventricular systolic pressure, echocardiography, pulmonary artery morphometry, and immunostaining. Proliferation, migration, matrix component expression, and mechanical signaling were assessed in MMP-8-/- and MMP-8+/+ pulmonary artery smooth muscle cells (PASMCs). Measurements and Main Results: MMP-8 expression was significantly increased in plasma and pulmonary arteries of patients with PH compared with controls and induced in the pulmonary vasculature in rodent PH models. Hypoxia-exposed MMP-8-/- mice had significant mortality, increased right ventricular systolic pressure, severe right ventricular dysfunction, and exaggerated vascular remodeling compared with MMP-8+/+ mice. MMP-8-/- PASMCs demonstrated exaggerated proliferation and migration mediated by altered matrix protein expression, elevated integrin-ß3 levels, and induction of FAK (focal adhesion kinase) and downstream YAP (Yes-associated protein)/TAZ (transcriptional coactivator with PDZ-binding motif) activity. Conclusions: MMP-8 is a novel protective factor upregulated in the pulmonary vasculature during PAH pathogenesis. MMP-8 opposes pathologic mechanobiological feedback by altering matrix composition and disrupting integrin-ß3/FAK and YAP/TAZ-dependent mechanical signaling in PASMCs.


Subject(s)
Matrix Metalloproteinase 8/metabolism , Pulmonary Arterial Hypertension/metabolism , Pulmonary Artery/metabolism , Adult , Aged , Animals , Biomarkers/metabolism , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Matrix Metalloproteinase 8/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Pulmonary Arterial Hypertension/pathology , Pulmonary Arterial Hypertension/prevention & control , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Up-Regulation , Vascular Remodeling
2.
JCI Insight ; 1(8)2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27347562

ABSTRACT

Pulmonary arterial (PA) stiffness is associated with increased mortality in patients with pulmonary hypertension (PH); however, the role of PA stiffening in the pathogenesis of PH remains elusive. Here, we show that distal vascular matrix stiffening is an early mechanobiological regulator of experimental PH. We identify cyclooxygenase-2 (COX-2) suppression and corresponding reduction in prostaglandin production as pivotal regulators of stiffness-dependent vascular cell activation. Atomic force microscopy microindentation demonstrated early PA stiffening in experimental PH and human lung tissue. Pulmonary artery smooth muscle cells (PASMC) grown on substrates with the stiffness of remodeled PAs showed increased proliferation, decreased apoptosis, exaggerated contraction, enhanced matrix deposition, and reduced COX-2-derived prostanoid production compared with cells grown on substrates approximating normal PA stiffness. Treatment with a prostaglandin I2 analog abrogated monocrotaline-induced PA stiffening and attenuated stiffness-dependent increases in proliferation, matrix deposition, and contraction in PASMC. Our results suggest a pivotal role for early PA stiffening in PH and demonstrate the therapeutic potential of interrupting mechanobiological feedback amplification of vascular remodeling in experimental PH.

3.
Cardiovasc Toxicol ; 16(2): 163-71, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25900702

ABSTRACT

Chronic cardiovascular disease is associated with air pollution exposure in epidemiology and toxicology studies. Inhaled toxicants can induce changes in serum bioactivity that impact endothelial inflammatory gene expression in vitro and impair vasorelaxation ex vivo, which are common precursors to atherosclerosis. Comparisons between single pollutants and common combustion mixtures, in terms of driving such serum inflammatory and vasoactive effects, have not been characterized. Healthy C57BL/6 mice were exposed to a single 6-h period of contrasting pollutant atmospheres: road dust, mixed vehicle emissions (MVE; a combination of gasoline and diesel engine emissions) particulate matter, mixed vehicle emissions gases, road dust plus ozone, road dust plus MVE, and hardwood smoke. Serum obtained from mice 24 h after these exposures was used as a stimulus to assess inflammatory potential in two assays: incubated with primary murine cerebrovascular endothelial cells for 4 h to measure inflammatory gene expression or applied to naïve aortic rings in an ex vivo myographic preparation. Road dust and wood smoke exposures were most potent at inducing inflammatory gene expression, while MVE atmospheres and wood smoke were most potent at impairing vasorelaxation to acetylcholine. Responses are consistent with recent reports on MVE toxicity, but reveal novel serum bioactivity related to wood smoke and road dust. These studies suggest that the compositional changes in serum and resultant bioactivity following inhalation exposure to pollutants may be highly dependent on the composition of mixtures.


Subject(s)
Air Pollutants/toxicity , Inflammation Mediators/blood , Inhalation Exposure , Particulate Matter/toxicity , Serum/metabolism , Vehicle Emissions/toxicity , Air Pollutants/adverse effects , Animals , Aorta/drug effects , Aorta/physiology , Dose-Response Relationship, Drug , Inhalation Exposure/adverse effects , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Particulate Matter/administration & dosage , Vasodilator Agents/pharmacology
4.
Am J Respir Cell Mol Biol ; 49(6): 1093-101, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23885794

ABSTRACT

Systemic sclerosis (SSc) is a systemic autoimmune disease that causes inflammation, vasculopathy, and fibrosis of the skin and internal organs. One of the most severe complications of SSc involves the development of pulmonary fibrosis. Endothelial cell injury precedes the development of fibrosis, and is believed to be an initiating event. Therefore, we aimed to characterize the role of endothelial cells in the progression of pulmonary fibrosis, using a well-established bleomycin (BLM) model of pulmonary fibrosis. Endothelial cells were isolated by cell sorting, and the analysis of gene expression was performed with quantitative RT-PCR. Endothelial injury was induced between the first and second week, as shown by the elevated expression of the vascular injury markers matrix metallopeptidase-12 and von Willebrand factor. After injury, endothelial activation was indicated by the up-regulation of selectins, CCL chemokines, and inflammatory mediators, including complement anaphylatoxin receptors (C3aR and C5aR), oncostatin M, and leukemia inhibitory factor. The endothelial cell overexpression of fibrotic mediators, including connective tissue growth factor, plasminogen activator inhibitor-1, osteopontin, fibronectin, and fibroblast specific protein-1, was observed in the second and fourth weeks. This study suggests that endothelial cells actively contribute to the disease process via multiple mechanisms, including the recruitment of inflammatory cells and the establishment of a profibrotic environment during the development of BLM-induced pulmonary fibrosis.


Subject(s)
Bleomycin/toxicity , Endothelial Cells/drug effects , Lung Injury/etiology , Macrophages/drug effects , Pulmonary Fibrosis/etiology , Animals , Chemokines/genetics , Chemokines/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Inflammation Mediators/metabolism , Lung Injury/metabolism , Lung Injury/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Scleroderma, Systemic/etiology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Selectins/genetics , Selectins/metabolism , Up-Regulation
5.
Am J Pathol ; 182(6): 2391-406, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23583651

ABSTRACT

Pulmonary arterial hypertension (PAH) is a chronic and progressive disease characterized by pulmonary vasculopathy with elevation of pulmonary artery pressure, often culminating in right ventricular failure. GATA-6, a member of the GATA family of zinc-finger transcription factors, is highly expressed in quiescent vasculature and is frequently lost during vascular injury. We hypothesized that endothelial GATA-6 may play a critical role in the molecular mechanisms underlying endothelial cell (EC) dysfunction in PAH. Here we report that GATA-6 is markedly reduced in pulmonary ECs lining both occluded and nonoccluded vessels in patients with idiopathic and systemic sclerosis-associated PAH. GATA-6 transcripts are also rapidly decreased in rodent PAH models. Endothelial GATA-6 is a direct transcriptional regulator of genes controlling vascular tone [endothelin-1, endothelin-1 receptor type A, and endothelial nitric oxide synthase (eNOS)], pro-inflammatory genes, CX3CL1 (fractalkine), 5-lipoxygenease-activating protein, and markers of vascular remodeling, including PAI-1 and RhoB. Mice with the genetic deletion of GATA-6 in ECs (Gata6-KO) spontaneously develop elevated pulmonary artery pressure and increased vessel muscularization, and these features are further exacerbated in response to hypoxia. Furthermore, innate immune cells including macrophages (CD11b(+)/F4/80(+)), granulocytes (Ly6G(+)/CD45(+)), and dendritic cells (CD11b(+)/CD11c(+)) are significantly increased in normoxic Gata6-KO mice. Together, our findings suggest a critical role of endothelial GATA-6 deficiency in development and disease progression in PAH.


Subject(s)
Endothelium, Vascular/metabolism , GATA6 Transcription Factor/deficiency , Hypertension, Pulmonary/metabolism , Adaptation, Physiological/physiology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/biosynthesis , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Case-Control Studies , Chronic Disease , Disease Progression , Down-Regulation/physiology , Endothelial Cells/physiology , Familial Primary Pulmonary Hypertension , GATA6 Transcription Factor/metabolism , GATA6 Transcription Factor/physiology , Gene Expression Regulation/physiology , Humans , Hypertension, Pulmonary/etiology , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/complications , Lung/blood supply , Male , Mice , Mice, Knockout , Pneumonia/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Scleroderma, Systemic/complications
6.
PLoS One ; 8(2): e56123, 2013.
Article in English | MEDLINE | ID: mdl-23441162

ABSTRACT

Endothelin 1 (ET-1) is a key regulator of vascular homeostasis. We have recently reported that the presence of Human antigen class I, HLA-B35, contributes to human dermal microvascular endothelial cell (HDMEC) dysfunction by upregulating ET-1 and proinflammatory genes. Likewise, a Toll-like receptor 3 (TLR3) ligand, Poly(I:C), was shown to induce ET-1 expression in HDMECs. The goal of this study was to determine the molecular mechanism of ET-1 induction by these two agonists. Because HLA-B35 expression correlated with induction of Binding Immunoglobulin Protein (BiP/GRP78) and several heat shock proteins, we first focused on ER stress and unfolded protein response (UPR) as possible mediators of this response. ER stress inducer, Thapsigargin (TG), HLA-B35, and Poly(I:C) induced ET-1 expression with similar potency in HDMECs. TG and HLA-B35 activated the PERK/eIF2α/ATF4 branch of the UPR and modestly increased the spliced variant of XBP1, but did not affect the ATF6 pathway. Poly(I:C) also activated eIF2α/ATF4 in a protein kinase R (PKR)-dependent manner. Depletion of ATF4 decreased basal expression levels of ET-1 mRNA and protein, and completely prevented upregulation of ET-1 by all three agonists. Additional experiments have demonstrated that the JNK and NF-κB pathways are also required for ET-1 upregulation by these agonists. Formation of the ATF4/c-JUN complex, but not the ATF4/NF-κB complex was increased in the agonist treated cells. The functional role of c-JUN in responses to HLA-B35 and Poly(I:C) was further confirmed in ET-1 promoter assays. This study identified ATF4 as a novel activator of the ET-1 gene. The ER stress/UPR and TLR3 pathways converge on eIF2α/ATF4 during activation of endothelial cells.


Subject(s)
Activating Transcription Factor 4/genetics , Endothelial Cells/metabolism , Endothelin-1/genetics , HLA-B35 Antigen/metabolism , RNA, Double-Stranded/metabolism , Activating Transcription Factor 4/metabolism , Adult , Aged , Animals , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Endothelin-1/metabolism , Enzyme Activation , Female , Gene Expression Regulation/drug effects , HLA-B35 Antigen/genetics , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Microvessels , Middle Aged , NF-kappa B/metabolism , Poly I-C/administration & dosage , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Unfolded Protein Response/drug effects , Young Adult , eIF-2 Kinase/metabolism
7.
J Cell Physiol ; 228(8): 1774-83, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23359533

ABSTRACT

Systemic sclerosis (SSc) is a complex disease characterized by vascular alterations, activation of the immune system and tissue fibrosis. Previous studies have implicated activation of the interferon pathways in the pathogenesis of SSc. The goal of this study was to determine whether interferon type I and/or type II could play a pathogenic role in SSc vasculopathy. Human dermal microvascular endothelial cells (HDMVECs) and fibroblasts were obtained from foreskins of healthy newborns. The RT Profiler PCR Array System was utilized to screen for EndoMT genes. Treatment with IFN-α or IFN-γ downregulated Fli1 and VE-cadherin. In contrast, IFN-α and IFN-γ exerted opposite effects on the expression of α-SMA, CTGF, ET-1, and TGFß2, with IFN-α downregulating and IFN-γ upregulating this set of genes. Blockade of TGFß signaling normalized IFN-γ-mediated changes in Fli1, VE-cadherin, CTGF, and ET-1 levels, whereas upregulation of α-SMA and TGFß2 was not affected. Bosentan treatment was more effective than TGFß blockade in reversing the actions of IFN-γ, including downregulation of α-SMA and TGFß2, suggesting that activation of the ET-1 pathway plays a main role in the IFN-γ responses in HDMECs. IFN-γ induced expression of selected genes related to endothelial-to-mesenchymal transition (EndoMT), including Snail1, FN1, PAI1, TWIST1, STAT3, RGS2, and components of the WNT pathway. The effect of IFN-γ on EndoMT was mediated via TGFß2 and ET-1 signaling pathways. This study demonstrates distinct effects of IFN-α and IFN-γ on the biology of vascular endothelial cells. IFN-γ may contribute to abnormal vascular remodeling and fibrogenesis in SSc, partially via induction of EndoMT.


Subject(s)
Endothelial Cells/metabolism , Endothelin-1/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Interferon-gamma/physiology , Transforming Growth Factor beta2/metabolism , Cells, Cultured , Endothelin-1/genetics , Fibrosis , Humans , Scleroderma, Systemic/etiology , Scleroderma, Systemic/pathology , Scleroderma, Systemic/physiopathology , Transforming Growth Factor beta2/genetics , Up-Regulation/drug effects
8.
J Cell Physiol ; 226(12): 3340-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21344387

ABSTRACT

TGF-ß is the primary inducer of extracellular matrix proteins in scleroderma (systemic sclerosis, SSc). Previous studies indicate that in a subset of SSc fibroblasts TGF-ß signaling is activated via elevated levels of activin receptor-like kinase (ALK) 1 and phosphorylated Smad1 (pSmad1). The goal of this study was to determine the role of endoglin/ALK1 in TGF-ß/Smad1 signaling in SSc fibroblasts. In SSc fibroblasts, increased levels of endoglin correlated with high levels of pSmad1, collagen, and connective tissue growth factor (CCN2). Endoglin depletion via siRNA in SSc fibroblasts inhibited pSmad1 but did not affect pSmad2/3. Following endoglin depletion mRNA and protein levels of collagen and CCN2 were significantly decreased in SSc fibroblasts but remained unchanged in normal fibroblasts. ALK1 was expressed at similar levels in SSc and normal fibroblasts. Depletion of ALK1 resulted in inhibition of pSmad1 and a moderate but significant reduction of mRNA and protein levels of collagen and CCN2 in SSc fibroblasts. Furthermore, constitutively high levels of endoglin were found in complexes with ALK1 in SSc fibroblasts. Overexpression of constitutively active ALK1 (caALK1) in normal and SSc fibroblasts led to a moderate increase of collagen and CCN2. However, caALK1 potently induced endothelin 1 (ET-1) mRNA and protein levels in SSc fibroblasts. Additional experiments demonstrated that endoglin and ALK1 mediate TGF-ß induction of ET-1 in SSc and normal fibroblasts. In conclusion, this study has revealed an important profibrotic role of endoglin in SSc fibroblasts. The endoglin/ALK1/Smad1 pathway could be a therapeutic target in patients with SSc if appropriately blocked.


Subject(s)
Antigens, CD/metabolism , Fibroblasts/metabolism , Receptors, Cell Surface/metabolism , Scleroderma, Diffuse/metabolism , Signal Transduction , Skin/metabolism , Smad1 Protein/metabolism , Transforming Growth Factor beta/metabolism , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Antigens, CD/genetics , Collagen/genetics , Collagen/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Endoglin , Endothelin-1/genetics , Endothelin-1/metabolism , Enzyme Activation , Fibroblasts/pathology , Fibrosis , HEK293 Cells , Humans , Mutation , Phenotype , Phosphorylation , RNA Interference , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Scleroderma, Diffuse/genetics , Scleroderma, Diffuse/pathology , Skin/pathology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transfection , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL