Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Am J Respir Crit Care Med ; 203(12): 1533-1545, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33523764

ABSTRACT

Rationale: Data on the molecular mechanisms that regulate platelet-pulmonary endothelial adhesion under conditions of hypoxia are lacking, but may have important therapeutic implications. Objectives: To identify a hypoxia-sensitive, modifiable mediator of platelet-pulmonary artery endothelial cell adhesion and thrombotic remodeling. Methods: Network medicine was used to profile protein-protein interactions in hypoxia-treated human pulmonary artery endothelial cells. Data from liquid chromatography-mass spectrometry and microscale thermophoresis informed the development of a novel antibody (Ab) to inhibit platelet-endothelial adhesion, which was tested in cells from patients with chronic thromboembolic pulmonary hypertension (CTEPH) and three animal models in vivo. Measurements and Main Results: The protein NEDD9 was identified in the hypoxia thrombosome network in silico. Compared with normoxia, hypoxia (0.2% O2) for 24 hours increased HIF-1α (hypoxia-inducible factor-1α)-dependent NEDD9 upregulation in vitro. Increased NEDD9 was localized to the plasma-membrane surface of cells from control donors and patients with CTEPH. In endarterectomy specimens, NEDD9 colocalized with the platelet surface adhesion molecule P-selectin. Our custom-made anti-NEDD9 Ab targeted the NEDD9-P-selectin interaction and inhibited the adhesion of activated platelets to pulmonary artery endothelial cells from control donors in vitro and from patients with CTEPH ex vivo. Compared with control mice, platelet-pulmonary endothelial aggregates and pulmonary hypertension induced by ADP were decreased in NEDD9-/- mice or wild-type mice treated with the anti-NEDD9 Ab, which also decreased chronic pulmonary thromboembolic remodeling in vivo. Conclusions: The NEDD9-P-selectin protein-protein interaction is a modifiable target with which to inhibit platelet-pulmonary endothelial adhesion and thromboembolic vascular remodeling, with potential therapeutic implications for patients with disorders of increased hypoxia signaling pathways, including CTEPH.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Adhesion/physiology , Hypoxia/physiopathology , Pulmonary Circulation/physiology , Pulmonary Embolism/physiopathology , Signal Transduction/physiology , Animals , Blood Platelets/physiology , Cells, Cultured/physiology , Endothelial Cells/physiology , Female , Humans , Male , Mice , Middle Aged , Models, Animal
2.
Chest ; 163(3): 678-686, 2023 03.
Article in English | MEDLINE | ID: mdl-36243062

ABSTRACT

BACKGROUND: Elevated mean pulmonary artery pressure (mPAP) is common in patients with hypertrophic cardiomyopathy (HCM) and heart failure symptoms. However, dynamic left ventricular (LV) outflow tract obstruction may confound interpretation of pulmonary hypertension (PH) pathophysiologic features in HCM when relying on resting invasive hemodynamic data alone. RESEARCH QUESTION: Do structural changes to the lung vasculature clarify PH pathophysiologic features in patients with HCM with progressive heart failure? STUDY DESIGN AND METHODS: Clinical data and ultrarare lung autopsy specimens were acquired retrospectively from the National Institutes of Health (1975-1992). Patients were included based on the availability of lung tissue and recorded mPAP. Discarded tissue from rejected lung donors served as control specimens. Histomorphology was performed on pulmonary arterioles and veins. Comparisons were calculated using the Student t test and Mann-Whitney U test; Pearson correlation was used to assess association between morphometric measurements and HCM cardiac and hemodynamic measurements. RESULTS: The HCM cohort (n = 7; mean ± SD age, 43 ± 18 years; 71% men) showed maximum mean ± SD LV wall thickness of 25 ± 2.8 mm, mean ± SD outflow tract gradient of 90 ± 30 mm Hg, median mPAP of 25 mm Hg (interquartile range [IQR], 6 mm Hg), median pulmonary artery wedge pressure (PAWP) of 16 mm Hg (IQR, 4 mm Hg), and median pulmonary vascular resistance of 1.8 Wood units (WU; IQR, 2.4 WU). Compared with control samples (n = 5), patients with HCM showed greater indexed pulmonary arterial hypertrophy (20.7 ± 7.2% vs 49.7 ± 12%; P < .001) and arterial wall fibrosis (11.5 ± 3.4 mm vs 21.0 ± 4.7 mm; P < .0001), which correlated with mPAP (r = 0.84; P = .018), PAWP (r = 0.74; P = .05), and LV outflow tract gradient (r = 0.78; P = .035). Compared with control samples, pulmonary vein thickness was increased by 2.9-fold (P = .008) in the HCM group, which correlated with mPAP (r = 0.81; P = .03) and LV outflow tract gradient (r = 0.83; P = .02). INTERPRETATION: To the best of our knowledge, these data demonstrate for the first time that in patients with obstructive HCM, heart failure is associated with pathogenic pulmonary vascular remodeling even when mPAP is elevated only mildly. These observations clarify PH pathophysiologic features in HCM, with future implications for clinical strategies that mitigate outflow tract obstruction.


Subject(s)
Cardiomyopathy, Hypertrophic , Heart Failure , Hypertension, Pulmonary , Male , Humans , Adult , Middle Aged , Female , Hypertension, Pulmonary/complications , Retrospective Studies , Vascular Remodeling , Cardiomyopathy, Hypertrophic/complications , Heart Failure/complications
3.
Pulm Circ ; 12(2): e12071, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35599981

ABSTRACT

The pathobiology of in situ pulmonary thrombosis in acute respiratory distress syndrome (ARDS) due to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is incompletely characterized. In human pulmonary artery endothelial cells (HPAECs), hypoxia increases neural precursor cell expressed, developmentally downregulated 9 (NEDD9) and induces expression of a prothrombotic NEDD9 peptide (N9P) on the extracellular plasma membrane surface. We hypothesized that the SARS-CoV-2-ARDS pathophenotype involves increased pulmonary endothelial N9P. Paraffin-embedded autopsy lung specimens were acquired from patients with SARS-CoV-2-​​​​​​ARDS (n = 13), ARDS from other causes (n = 10), and organ donor controls (n = 5). Immunofluorescence characterized the expression of N9P, fibrin, and transcription factor 12 (TCF12), a putative binding target of SARS-CoV-2 and known transcriptional regulator of NEDD9. We performed RNA-sequencing on normal HPAECs treated with normoxia or hypoxia (0.2% O2) for 24 h. Immunoprecipitation-liquid chromatography-mass spectrometry (IP-LC-MS) profiled protein-protein interactions involving N9P relevant to thrombus stabilization. Hypoxia increased TCF12 messenger RNA significantly compared to normoxia in HPAECs in vitro (+1.19-fold, p = 0.001; false discovery rate = 0.005), and pulmonary endothelial TCF12 expression was increased threefold in SARS-CoV-2-ARDS versus donor control lungs (p < 0.001). Compared to donor controls, pulmonary endothelial N9P-fibrin colocalization was increased in situ in non-SARS-CoV-2-ARDS and SARS-CoV-2-ARDS decedents (3.7 ± 1.2 vs. 10.3 ± 3.2 and 21.8 ± 4.0 arb. units, p < 0.001). However, total pulmonary endothelial N9P was increased significantly only in SARS-CoV-2-ARDS versus donor controls (15 ± 4.2 vs. 6.3 ± 0.9 arb. units, p < 0.001). In HPAEC plasma membrane isolates, IP-LC-MS identified a novel protein-protein interaction between NEDD9 and the ß3-subunit of the αvß3-integrin, which regulates fibrin anchoring to endothelial cells. In conclusion, lethal SARS-CoV-2-ARDS is associated with increased pulmonary endothelial N9P expression and N9P-fibrin colocalization in situ. Further investigation is needed to determine the pathogenetic and potential therapeutic relevance of N9P to the thrombotic pathophenotype of SARS-CoV-2-ARDS.

4.
Am J Pathol ; 176(3): 1148-56, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20093498

ABSTRACT

Sarcoidosis is a chronic disease of unknown etiology characterized by the formation of non-necrotizing epithelioid granulomas in various organs, especially in the lungs. The lack of an adequate animal model reflecting the pathogenesis of the human disease is one of the major impediments in studying sarcoidosis. In this report, we describe ApoE-/- mice on a cholate-containing high-fat diet that exhibit granulomatous lung inflammation similar to human sarcoidosis. Histological analysis revealed well-defined and non-necrotizing granulomas in about 40% of mice with the highest number of granulomas after 16 weeks on a cholate-containing high-fat diet. Granulomas contained CD4+ and CD8+ T cells, and the majority of the cells in granulomas showed immunoreactivity for the macrophage marker Mac-3. Cells with morphological features of epithelioid cells expressed angiotensin-converting enzyme, osteopontin, and cathepsin K, all characteristics of epithelioid and giant cells in granulomas of human sarcoidosis. Giant cells and nonspecific inclusions such as Schaumann's bodies and crystalline deposits were also detected in some lungs. Granulomatous inflammation resulted in progressive pulmonary fibrosis. Removal of cholate from the diet prevented the formation of lung granulomas. The observed similarities between the analyzed mouse lung granulomas and granulomas of human sarcoidosis, as well as the chronic disease character leading to fibrosis, suggest that this mouse model might be a useful tool to study sarcoidosis.


Subject(s)
Apolipoproteins E/deficiency , Cholates/pharmacology , Diet , Dietary Fats/pharmacology , Sarcoidosis, Pulmonary/pathology , Adult , Animals , Apolipoproteins E/metabolism , Cathepsin K/metabolism , Cholates/administration & dosage , Dietary Fats/administration & dosage , Female , Granuloma/enzymology , Granuloma/pathology , Humans , Lung/enzymology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology , Sarcoidosis, Pulmonary/complications , Sarcoidosis, Pulmonary/enzymology
5.
Respir Res ; 12: 13, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21251246

ABSTRACT

BACKGROUND: Remodeling of lung tissues during the process of granuloma formation requires significant restructuring of the extra-cellular matrix and cathepsins K, L and S are among the strongest extra-cellular matrix degrading enzymes. Cathepsin K is highly expressed in various pathological granulomatous infiltrates and all three enzymes in their active form are detected in bronchoalveolar lavage fluids from patients with sarcoidosis. Granulomatous inflammation is driven by T-cell response and cathepsins S and L are actively involved in the regulation of antigen presentation and T-cell selection. Here, we show that the disruption of the activities of cathepsins K, L, or S affects the development of lung granulomas in a mouse model of sarcoidosis. METHODS: Apolipoprotein E-deficient mice lacking cathepsin K or L were fed Paigen diet for 16 weeks and lungs were analyzed and compared with their cathepsin-expressing littermates. The role of cathepsin S in the development of granulomas was evaluated using mice treated for 8 weeks with a potent and selective cathepsin S inhibitor. RESULTS: When compared to wild-type litters, more cathepsin K-deficient mice had lung granulomas, but individually affected mice developed smaller granulomas that were present in lower numbers. The absence of cathepsin K increased the number of multinucleated giant cells and the collagen content in granulomas. Cathepsin L deficiency resulted in decreased size and number of lung granulomas. Apoe-/- mice treated with a selective cathepsin S inhibitor did not develop lung granulomas and only individual epithelioid cells were observed. CONCLUSIONS: Cathepsin K deficiency affected mostly the occurrence and composition of lung granulomas, whereas cathepsin L deficiency significantly reduced their number and cathepsin S inhibition prevented the formation of granulomas.


Subject(s)
Cathepsin K/deficiency , Cathepsin L/deficiency , Cathepsins/antagonists & inhibitors , Granuloma, Respiratory Tract/prevention & control , Lung/drug effects , Protease Inhibitors/pharmacology , Sarcoidosis, Pulmonary/drug therapy , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , CD4-Positive T-Lymphocytes/immunology , Cathepsin K/genetics , Cathepsin L/genetics , Cathepsins/metabolism , Collagen/metabolism , Disease Models, Animal , Granuloma, Respiratory Tract/enzymology , Granuloma, Respiratory Tract/genetics , Granuloma, Respiratory Tract/immunology , Granuloma, Respiratory Tract/pathology , Hypertrophy , Lung/enzymology , Lung/immunology , Lung/pathology , Mice , Mice, Knockout , Sarcoidosis, Pulmonary/enzymology , Sarcoidosis, Pulmonary/genetics , Sarcoidosis, Pulmonary/immunology , Sarcoidosis, Pulmonary/pathology , Thymus Gland/enzymology , Thymus Gland/pathology , Time Factors
6.
Circ Res ; 104(4): 455-65, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19122179

ABSTRACT

The molecular events linking lipid accumulation in atherosclerotic plaques to complications such as aneurysm formation and plaque disruption are poorly understood. BALB/c-Apoe(-/-) mice bearing a null mutation in the Npc1 gene display prominent medial erosion and atherothrombosis, whereas their macrophages accumulate free cholesterol in late endosomes and show increased cathepsin K (Ctsk) expression. We now show increased cathepsin K immunostaining and increased cysteinyl proteinase activity using near infrared fluorescence imaging over proximal aortas of Apoe(-/-), Npc1(-/-) mice. In mechanistic studies, cholesterol loading of macrophage plasma membranes (cyclodextrin-cholesterol) or endosomal system (AcLDL+U18666A or Npc1 null mutation) activated Toll-like receptor (TLR) signaling, leading to sustained phosphorylation of p38 mitogen-activated protein kinase and induction of p38 targets, including Ctsk, S100a8, Mmp8, and Mmp14. Studies in macrophages from knockout mice showed major roles for TLR4, following plasma membrane cholesterol loading, and for TLR3, after late endosomal loading. TLR signaling via p38 led to phosphorylation and activation of the transcription factor Microphthalmia transcription factor, acting at E-box elements in the Ctsk promoter. These studies suggest that free cholesterol enrichment of either plasma or endosomal membranes in macrophages leads to activation of signaling via various TLRs, prolonged p38 mitogen-activated protein kinase activation, and induction of Mmps, Ctsk, and S100a8, potentially contributing to plaque complications.


Subject(s)
Cathepsins/biosynthesis , Cell Membrane/metabolism , Cholesterol/metabolism , Endosomes/metabolism , Macrophages/metabolism , Signal Transduction , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/genetics , Animals , Aorta/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Calgranulin A , Cathepsin K , Cell Membrane/enzymology , Cell Membrane/immunology , Cells, Cultured , E-Box Elements , Endosomes/enzymology , Endosomes/immunology , Enzyme Induction , Humans , Intracellular Signaling Peptides and Proteins , Macrophages/enzymology , Macrophages/immunology , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 8/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Microphthalmia-Associated Transcription Factor/metabolism , Niemann-Pick C1 Protein , Phosphorylation , Promoter Regions, Genetic , Proteins/genetics , Proteins/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , S100 Proteins/metabolism , Time Factors , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/genetics , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , p38 Mitogen-Activated Protein Kinases/deficiency , p38 Mitogen-Activated Protein Kinases/genetics , rab GTP-Binding Proteins/metabolism
7.
Arterioscler Thromb Vasc Biol ; 30(6): 1166-73, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20203298

ABSTRACT

OBJECTIVE: To determine the role of multinucleated giant cells (MGCs) in cardiovascular diseases. METHODS AND RESULTS: MGCs are a hallmark of giant cell arteritis. They are also described in atherosclerotic plaques from aortic aneurysms and carotid and coronary arteries. Herein, we demonstrate that the cholate-containing Paigen diet yields many MGCs in atherosclerotic plaques of apolipoprotein E-/- mice. These mice revealed a 4-fold increase in MGC numbers when compared with mice on a Western or Paigen diet without cholate. Most of the MGCs stained intensively for cathepsin K and were located at fibrous caps and close to damaged elastic laminae, with associated medial smooth muscle cell depletion. During in vitro experiments, MGCs demonstrated a 6-fold increase in elastolytic activity when compared with macrophages and facilitated transmigration of smooth muscle cells through a collagen-elastin matrix. An elastin-derived hexapeptide (Val-Gly-Val-Ala-Pro-Gly [VGVAPG]) significantly increased the rate of macrophage fusion, providing a possible mechanism of in vivo MGC formation. Comparable to the mouse model, human specimens from carotid arteries and aortic aneurysms contained cathepsin K-positive MGCs. CONCLUSIONS: Apolipoprotein E-/- mice fed a Paigen diet provide a model to analyze the tissue-destructive role of MGCs in vascular diseases.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/pathology , Giant Cells, Foreign-Body/pathology , Macrophages, Peritoneal/pathology , Animals , Antigens, Differentiation/metabolism , Aortic Aneurysm/metabolism , Aortic Aneurysm/pathology , Apolipoproteins E/genetics , Atherosclerosis/chemically induced , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Cathepsin K/deficiency , Cathepsin K/genetics , Cathepsin K/metabolism , Cell Fusion , Cell Movement , Cells, Cultured , Cholates , Collagen/metabolism , Dietary Fats , Disease Models, Animal , Elastin/metabolism , Endotoxins/blood , Giant Cells, Foreign-Body/drug effects , Giant Cells, Foreign-Body/immunology , Giant Cells, Foreign-Body/metabolism , Humans , Immunohistochemistry , Interleukin-4/blood , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Oligopeptides/pharmacology , Time Factors , Toll-Like Receptor 4/metabolism
8.
Nat Commun ; 12(1): 873, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33558530

ABSTRACT

Progress in precision medicine is limited by insufficient knowledge of transcriptomic or proteomic features in involved tissues that define pathobiological differences between patients. Here, myectomy tissue from patients with obstructive hypertrophic cardiomyopathy and heart failure is analyzed using RNA-Seq, and the results are used to develop individualized protein-protein interaction networks. From this approach, hypertrophic cardiomyopathy is distinguished from dilated cardiomyopathy based on the protein-protein interaction network pattern. Within the hypertrophic cardiomyopathy cohort, the patient-specific networks are variable in complexity, and enriched for 30 endophenotypes. The cardiac Janus kinase 2-Signal Transducer and Activator of Transcription 3-collagen 4A2 (JAK2-STAT3-COL4A2) expression profile informed by the networks was able to discriminate two hypertrophic cardiomyopathy patients with extreme fibrosis phenotypes. Patient-specific network features also associate with other important hypertrophic cardiomyopathy clinical phenotypes. These proof-of-concept findings introduce personalized protein-protein interaction networks (reticulotypes) for characterizing patient-specific pathobiology, thereby offering a direct strategy for advancing precision medicine.


Subject(s)
Cardiomyopathy, Hypertrophic/pathology , Gene Regulatory Networks , Precision Medicine , Case-Control Studies , Cohort Studies , Endophenotypes , Fibrosis , Heart Failure/genetics , Humans , Phenotype , Protein Interaction Maps/genetics , Signal Transduction , Transcriptome/genetics
9.
J Cardiovasc Pharmacol ; 56(1): 98-105, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20410833

ABSTRACT

Recent studies provided evidence for a significant role of cathepsin S during extracellular remodeling in atherosclerosis. In this study, we investigated the effect of a specific cathepsin S inhibitor on atherosclerotic plaque progression in the brachiocephalic artery. Male and female Apoe-/- mice on a cholate-containing high-fat diet containing or lacking a specific cathepsin S inhibitor were evaluated for the remodeling of atherosclerotic lesions. The in vivo efficacy of the cathepsin S inhibitor was demonstrated by the inhibition of invariant chain processing in spleen. After 8 weeks of diet, brachiocephalic arteries were analyzed for plaque size, collagen, macrophage, and smooth muscle cell content, for elastic lamina breaks, and the number of buried fibrous caps. The size of atherosclerotic plaques in inhibitor-treated mice was reduced by 36% in male and 68% in female mice, and they showed significantly smaller numbers in elastin lamina breaks (60% less in males; 75% less in females), plaque macrophages (47% less in males; 40% less in females), and buried fibrous caps (50% less in males; 86% less in females). In conclusion, the inhibition of cathepsin S showed a strong atheroprotective activity, demonstrating the potential benefits of a small molecule anti-cathepsin therapy.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Cathepsins/antagonists & inhibitors , Animals , Antigens, Differentiation, B-Lymphocyte/drug effects , Antigens, Differentiation, B-Lymphocyte/metabolism , Atherosclerosis/genetics , Brachiocephalic Trunk/pathology , Cathepsins/metabolism , Diet, Atherogenic , Female , Histocompatibility Antigens Class II/drug effects , Histocompatibility Antigens Class II/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Sex Factors , Spleen/drug effects , Spleen/pathology
10.
J Heart Lung Transplant ; 39(4): 289-299, 2020 04.
Article in English | MEDLINE | ID: mdl-31952977

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension (PAH) is a highly morbid disease characterized by elevated pulmonary vascular resistance (PVR) and pathogenic right ventricular remodeling. Endothelial expression of the prometastatic protein NEDD9 is increased in fibrotic PAH arterioles, and NEDD9 inhibition decreases PVR in experimental PAH. We hypothesized that circulating NEDD9 is increased in PAH and informs the clinical profile of patients. METHODS: Clinical data and plasma samples were analyzed retrospectively for 242 patients from 5 referral centers (2010-2017): PAH (n = 139; female 82%, 58 [48-67] years), non-PAH pulmonary hypertension (PH) (n = 54; female 56%, 63.4 ± 12.2 years), and dyspnea non-PH controls (n = 36; female 75%, 54.2 ± 14.0 years). RESULTS: Compared with controls, NEDD9 was increased in PAH by 1.82-fold (p < 0.0001). Elevated NEDD9 correlated with PVR in idiopathic PAH (ρ = 0.42, p < 0.0001, n = 54), connective tissue disease (CTD)-PAH (ρ = 0.53, p < 0.0001, n = 53), and congenital heart disease-PAH (ρ = 0.68, p < 0.0001, n = 10). In CTD-PAH, NEDD9 correlated with 6-minute walk distance (ρ = -0.35, p = 0.028, n = 39). In contrast to the PAH biomarker N-terminal pro-brain natriuretic peptide (n = 38), NEDD9 correlated inversely with exercise pulmonary artery wedge pressure and more strongly with right ventricular ejection fraction (ρ = -0.41, p = 0.006, n = 45) in a mixed population. The adjusted hazard ratio for lung transplant-free survival was 1.12 (95% confidence interval [CI], 1.02-1.22, p = 0.01) and 1.75 (95% CI, 1.12-2.73, p = 0.01) per 1 ng/ml and 5 ng/ml increase in plasma NEDD9, respectively, by Cox proportional hazard model. CONCLUSIONS: In PAH, plasma NEDD9 is increased and associates with key prognostic variables. Prospective studies that include hard end points are warranted to validate NEDD9 as a novel PAH biomarker.


Subject(s)
Adaptor Proteins, Signal Transducing/blood , Pulmonary Arterial Hypertension/blood , Pulmonary Wedge Pressure/physiology , Vascular Resistance/physiology , Ventricular Function, Right/physiology , Aged , Biomarkers/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Pulmonary Arterial Hypertension/physiopathology , Retrospective Studies , Signal Transduction
11.
PLoS One ; 14(2): e0211909, 2019.
Article in English | MEDLINE | ID: mdl-30811450

ABSTRACT

Transcriptomic analysis of pulmonary microvascular endothelial cells from experimental models offers insight into pulmonary arterial hypertension (PAH) pathobiology. However, culturing may alter the molecular profile of endothelial cells prior to analysis, limiting the translational relevance of results. Here we present a novel and validated method for isolating RNA from pulmonary microvascular endothelial cells (PMVECs) ex vivo that does not require cell culturing. Initially, presumed rat PMVECs were isolated from rat peripheral lung tissue using tissue dissociation and enzymatic digestion, and cells were cultured until confluence to assess endothelial marker expression. Anti-CD31, anti-von Willebrand Factor, and anti-α-smooth muscle actin immunocytochemistry/immunofluorescence signal was detected in presumed rat PMVECs, but also in non-endothelial cell type controls. By contrast, flow cytometry using an anti-CD31 antibody and isolectin 1-B4 (from Griffonia simplicifolia) was highly specific for rat PMVECs. We next developed a strategy in which the addition of an immunomagnetic selection step for CD31+ cells permitted culture-free isolation of rat PMVECs ex vivo for RNA isolation and transcriptomic analysis using fluorescence-activated cell sorting. Heterogeneity in the validity and reproducibility of results using commercial antibodies against endothelial surface markers corresponded to a substantial burden on laboratory time, labor, and scientific budget. We demonstrate a novel protocol for the culture-free isolation and transcriptomic analysis of rat PMVECs with translational relevance to PAH. In doing so, we highlight wide variability in the quality of commonly used biological reagents, which emphasizes the importance of investigator-initiated validation of commercial biomaterials.


Subject(s)
Biocompatible Materials/pharmacology , Cell Separation , Endothelial Cells/metabolism , Hypertension, Pulmonary/metabolism , Lung/metabolism , Microvessels/metabolism , Animals , Antigens, Differentiation/biosynthesis , Endothelial Cells/pathology , Gene Expression Regulation/drug effects , Humans , Hypertension, Pulmonary/pathology , Lung/blood supply , Lung/pathology , Microvessels/pathology , Rats
13.
Sci Transl Med ; 10(445)2018 06 13.
Article in English | MEDLINE | ID: mdl-29899023

ABSTRACT

Germline mutations involving small mothers against decapentaplegic-transforming growth factor-ß (SMAD-TGF-ß) signaling are an important but rare cause of pulmonary arterial hypertension (PAH), which is a disease characterized, in part, by vascular fibrosis and hyperaldosteronism (ALDO). We developed and analyzed a fibrosis protein-protein network (fibrosome) in silico, which predicted that the SMAD3 target neural precursor cell expressed developmentally down-regulated 9 (NEDD9) is a critical ALDO-regulated node underpinning pathogenic vascular fibrosis. Bioinformatics and microscale thermophoresis demonstrated that oxidation of Cys18 in the SMAD3 docking region of NEDD9 impairs SMAD3-NEDD9 protein-protein interactions in vitro. This effect was reproduced by ALDO-induced oxidant stress in cultured human pulmonary artery endothelial cells (HPAECs), resulting in impaired NEDD9 proteolytic degradation, increased NEDD9 complex formation with Nk2 homeobox 5 (NKX2-5), and increased NKX2-5 binding to COL3A1 Up-regulation of NEDD9-dependent collagen III expression corresponded to changes in cell stiffness measured by atomic force microscopy. HPAEC-derived exosomal signaling targeted NEDD9 to increase collagen I/III expression in human pulmonary artery smooth muscle cells, identifying a second endothelial mechanism regulating vascular fibrosis. ALDO-NEDD9 signaling was not affected by treatment with a TGF-ß ligand trap and, thus, was not contingent on TGF-ß signaling. Colocalization of NEDD9 with collagen III in HPAECs was observed in fibrotic pulmonary arterioles from PAH patients. Furthermore, NEDD9 ablation or inhibition prevented fibrotic vascular remodeling and pulmonary hypertension in animal models of PAH in vivo. These data identify a critical TGF-ß-independent posttranslational modification that impairs SMAD3-NEDD9 binding in HPAECs to modulate vascular fibrosis and promote PAH.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Collagen Type III/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Lung/metabolism , Lung/pathology , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Collagen Type III/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Lung/physiopathology , Male , Phosphoproteins/genetics , Protein Binding , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Smad3 Protein/genetics , Smad3 Protein/metabolism , Systems Biology/methods
14.
JACC Basic Transl Sci ; 2(4): 398-414, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28920098

ABSTRACT

Expression of the interleukin-1 receptor type I (IL-1RI) co-receptor Toll-like and interleukin-1 receptor regulator (TILRR) is significantly increased in blood monocytes following myocardial infarction and in the atherosclerotic plaque, whereas levels in healthy tissue are low. TILRR association with IL-1RI at these sites causes aberrant activation of inflammatory genes, which underlie progression of cardiovascular disease. The authors show that genetic deletion of TILRR or antibody blocking of TILRR function reduces development of atherosclerotic plaques. Lesions exhibit decreased levels of monocytes, with increases in collagen and smooth muscle cells, characteristic features of stable plaques. The results suggest that TILRR may constitute a rational target for site- and signal-specific inhibition of vascular disease.

16.
Atherosclerosis ; 200(1): 58-68, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18291403

ABSTRACT

Cathepsin K, a potent extracellular matrix degrading cysteine protease, has been linked to the pathogenesis of osteoporosis, arthritis, cardiovascular and respiratory diseases. Here, we report the effects of cathepsin K deficiency (ctsK-/-) on atherosclerotic plaque formation in brachiocephalic arteries in an aggressive atherosclerosis model using apoE-deficient mice on cholate-containing high fat diet (HFD). On this diet, apoE-/- mice displayed severe lesions with buried fibrous caps after 8 weeks, whereas the apoE-/-ctsK-/- mice revealed a significantly decreased number of buried fibrous caps accompanied by increased collagen content in plaque areas and fibrous cap thickness. After 16 weeks of HFD, ctsK-/- mice had smaller plaque areas and maintained the structure of the tunica media in terms of their smooth muscle cell content and elastic lamina integrity. Overall macrophage content in the tunica media was lower in ctsK-/- mice but higher in the plaque area after 8 weeks of HFD. Decreased apoptosis rates in atherosclerotic plaques in brachiocephalic arteries of cathepsin K-deficient indicated a lower level of inflammation. In conclusion, cathepsin K deficiency appears to increase lesion stability in brachiocephalic arteries by maintaining the integrity of the tunica media and by decreasing plaque vulnerability to rupture.


Subject(s)
Arteries/physiopathology , Atherosclerosis/physiopathology , Cathepsins/deficiency , Cathepsins/physiology , Animals , Apolipoproteins E/deficiency , Arteries/pathology , Cathepsin K , Diet, Atherogenic , Forelimb/blood supply , Mice , Mice, Knockout , Tunica Intima/pathology , Tunica Media/pathology
SELECTION OF CITATIONS
SEARCH DETAIL