Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Eur Respir J ; 57(3)2021 03.
Article in English | MEDLINE | ID: mdl-32972983

ABSTRACT

Pulmonary arterial hypertension (PAH) is a destructive disease of the pulmonary vasculature often leading to right heart failure and death. Current therapeutic intervention strategies only slow disease progression. The role of aberrant hypoxia-inducible factor (HIF)2α stability and function in the initiation and development of pulmonary hypertension (PH) has been an area of intense interest for nearly two decades.Here we determine the effect of a novel HIF2α inhibitor (PT2567) on PH disease initiation and progression, using two pre-clinical models of PH. Haemodynamic measurements were performed, followed by collection of heart, lung and blood for pathological, gene expression and biochemical analysis. Blood outgrowth endothelial cells from idiopathic PAH patients were used to determine the impact of HIF2α-inhibition on endothelial function.Global inhibition of HIF2a reduced pulmonary vascular haemodynamics and pulmonary vascular remodelling in both su5416/hypoxia prevention and intervention models. PT2567 intervention reduced the expression of PH-associated target genes in both lung and cardiac tissues and restored plasma nitrite concentration. Treatment of monocrotaline-exposed rodents with PT2567 reduced the impact on cardiovascular haemodynamics and promoted a survival advantage. In vitro, loss of HIF2α signalling in human pulmonary arterial endothelial cells suppresses target genes associated with inflammation, and PT2567 reduced the hyperproliferative phenotype and overactive arginase activity in blood outgrowth endothelial cells from idiopathic PAH patients. These data suggest that targeting HIF2α hetero-dimerisation with an orally bioavailable compound could offer a new therapeutic approach for PAH. Future studies are required to determine the role of HIF in the heterogeneous PAH population.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Animals , Cells, Cultured , Endothelial Cells , Humans , Hypertension, Pulmonary/drug therapy , Pulmonary Artery
2.
Pulm Circ ; 10(3): 2045894020935783, 2020.
Article in English | MEDLINE | ID: mdl-32733669

ABSTRACT

Pulmonary arterial hypertension is a fatal disorder of the lung circulation in which accumulation of vascular cells progressively obliterates the pulmonary arterioles. This results in sustained elevation in pulmonary artery pressure leading eventually to right heart failure. Approximately, 80% of familial and 20% of sporadic idiopathic pulmonary arterial hypertension cases are caused by mutations in the bone morphogenetic protein receptor type 2 (BMPR2). Nonsense mutations in BMPR2 are amongst the most common mutations found, where the insertion of a premature termination codon causes mRNA degradation via activation of the nonsense-mediated decay pathway leading to a state of haploinsufficiency. Ataluren (PTC124), a compound that permits ribosomal read-through of premature stop codons, has been previously reported to increase BMPR2 protein expression in cells derived from pulmonary arterial hypertension patients harbouring nonsense mutations. In this study, we characterised the effects of PTC124 on a range of nonsense BMPR2 mutations, focusing on the R584X mutation both in vitro and in vivo. Treatment with PTC124 partially restored BMPR2 protein expression in blood outgrowth endothelial cells isolated from a patient harbouring the R584X mutation. Furthermore, a downstream bone morphogenetic protein signalling target, Id1, was rescued by PTC124 treatment. Mutant cells also exhibited increased lipopolysaccharide-induced permeability, which was reversed by PTC124 treatment. Increased proliferation and apoptosis in R584X blood outgrowth endothelial cells were also significantly reduced by PTC124. Moreover, oral PTC124 increased lung BMPR2 protein expression in mice harbouring the R584X mutation (Bmpr2 +/R584X ). Our findings provide support for future experimental medicine studies of PTC124 in pulmonary arterial hypertension patients with specific nonsense BMPR2 mutations.

3.
Am J Respir Cell Mol Biol ; 63(2): 160-171, 2020 08.
Article in English | MEDLINE | ID: mdl-32255665

ABSTRACT

Mutations in the gene encoding BMPR2 (bone morphogenetic protein type 2 receptor) are the major cause of heritable pulmonary arterial hypertension (PAH). Point mutations in the BMPR2 ligand-binding domain involving cysteine residues (such as C118W) are causative of PAH and predicted to cause protein misfolding. Using heterologous overexpression systems, we showed previously that these mutations lead to retention of BMPR2 in the endoplasmic reticulum but are partially rescued by chemical chaperones. Here, we sought to determine whether the chemical chaperone 4-phenylbutyrate (4PBA) restores BMPR2 signaling in primary cells and in a knockin mouse harboring a C118W mutation. First, we confirmed dysfunctional BMP signaling in dermal fibroblasts isolated from a family with PAH segregating the BMPR2 C118W mutation. After BMP4 treatment, the induction of downstream signaling targets (Smad1/5, ID1 [inhibitor of DNA binding 1], and ID2) was significantly reduced in C118W mutant cells. Treatment with 4PBA significantly rescued Smad1/5, ID1, and ID2 expression. Pulmonary artery smooth muscle cells isolated from the lungs of heterozygous mice harboring the Bmpr2 C118W mutation exhibited significantly increased proliferation. In the presence of 4PBA, hyperproliferation was dramatically reduced. Furthermore, in vivo, 4PBA treatment of Bmpr2 C118W mice partially rescued Bmpr2 expression, restored downstream signaling, and improved vascular remodeling. These findings demonstrate in primary cells and in a knockin mouse that the repurposed small-molecule chemical chaperone 4PBA might be a promising precision medicine approach to treat PAH in patients with specific subtypes of BMPR2 mutation involving cysteine substitutions in the ligand-binding domain.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/genetics , Cysteine/genetics , Mutation/genetics , Organophosphorus Compounds/pharmacology , Pulmonary Arterial Hypertension/drug therapy , Signal Transduction/drug effects , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Humans , Mice , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Pulmonary Arterial Hypertension/genetics , Pulmonary Artery/drug effects , Signal Transduction/genetics , Vascular Remodeling/drug effects , Vascular Remodeling/genetics
4.
PLoS One ; 15(2): e0229409, 2020.
Article in English | MEDLINE | ID: mdl-32109943

ABSTRACT

The microRNA miR-1 is an important regulator of muscle phenotype including cardiac muscle. Down-regulation of miR-1 has been shown to occur in left ventricular hypertrophy but its contribution to right ventricular hypertrophy in pulmonary arterial hypertension are not known. Previous studies have suggested that miR-1 may suppress transforming growth factor-beta (TGF-ß) signalling, an important pro-hypertrophic pathway but only indirect mechanisms of regulation have been identified. We identified the TGF-ß type 1 receptor (TGF-ßR1) as a putative miR-1 target. We therefore hypothesized that miR-1 and TGF-ßR1 expression would be inversely correlated in hypertrophying right ventricle of rats with pulmonary arterial hypertension and that miR-1 would inhibit TGF-ß signalling by targeting TGF-ßR1 expression. Quantification of miR-1 and TGF-ßR1 in rats treated with monocrotaline to induce pulmonary arterial hypertension showed appropriate changes in miR-1 and TGF-ßR1 expression in the hypertrophying right ventricle. A miR-1-mimic reduced enhanced green fluorescent protein expression from a reporter vector containing the TGF-ßR1 3'- untranslated region and knocked down endogenous TGF-ßR1. Lastly, miR-1 reduced TGF-ß activation of a (mothers against decapentaplegic homolog) SMAD2/3-dependent reporter. Taken together, these data suggest that miR-1 targets TGF-ßR1 and reduces TGF-ß signalling, so a reduction in miR-1 expression may increase TGF-ß signalling and contribute to cardiac hypertrophy.


Subject(s)
Cardiomegaly/pathology , Gene Expression Regulation , Hypertrophy, Right Ventricular/pathology , MicroRNAs/genetics , Pulmonary Arterial Hypertension/complications , Receptor, Transforming Growth Factor-beta Type I/metabolism , Animals , Cardiomegaly/etiology , Cardiomegaly/metabolism , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type I/genetics
5.
Proc Natl Acad Sci U S A ; 116(26): 13122-13130, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31152133

ABSTRACT

Iron deficiency augments hypoxic pulmonary arterial pressure in healthy individuals and exacerbates pulmonary arterial hypertension (PAH) in patients, even without anemia. Conversely, iron supplementation has been shown to be beneficial in both settings. The mechanisms underlying the effects of iron availability are not known, due to lack of understanding of how cells of the pulmonary vasculature respond to changes in iron levels. The iron export protein ferroportin (FPN) and its antagonist peptide hepcidin control systemic iron levels by regulating release from the gut and spleen, the sites of absorption and recycling, respectively. We found FPN to be present in pulmonary arterial smooth muscle cells (PASMCs) and regulated by hepcidin cell autonomously. To interrogate the importance of this regulation, we generated mice with smooth muscle-specific knock in of the hepcidin-resistant isoform fpn C326Y. While retaining normal systemic iron levels, this model developed PAH and right heart failure as a consequence of intracellular iron deficiency and increased expression of the vasoconstrictor endothelin-1 (ET-1) within PASMCs. PAH was prevented and reversed by i.v. iron and by the ET receptor antagonist BQ-123. The regulation of ET-1 by iron was also demonstrated in healthy humans exposed to hypoxia and in PASMCs from PAH patients with mutations in bone morphogenetic protein receptor type II. Such mutations were further associated with dysregulation of the HAMP/FPN axis in PASMCs. This study presents evidence that intracellular iron deficiency specifically within PASMCs alters pulmonary vascular function. It offers a mechanistic underpinning for the known effects of iron availability in humans.


Subject(s)
Iron Deficiencies , Myocytes, Smooth Muscle/pathology , Pulmonary Arterial Hypertension/etiology , Pulmonary Artery/pathology , Administration, Intravenous , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Disease Models, Animal , Endothelin A Receptor Antagonists/administration & dosage , Endothelin-1/metabolism , Gene Knock-In Techniques , Hepcidins/metabolism , Humans , Iron/administration & dosage , Male , Mice , Myocytes, Smooth Muscle/metabolism , Pulmonary Arterial Hypertension/pathology , Pulmonary Arterial Hypertension/prevention & control , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , Receptor, Endothelin A/metabolism , Up-Regulation
6.
Br J Pharmacol ; 176(9): 1206-1221, 2019 05.
Article in English | MEDLINE | ID: mdl-30710493

ABSTRACT

BACKGROUND AND PURPOSE: Apelin is an endogenous vasodilatory and inotropic peptide that is down-regulated in human pulmonary arterial hypertension, although the density of the apelin receptor is not significantly attenuated. We hypothesised that a G protein-biased apelin analogue MM07, which is more stable than the endogenous apelin peptide, may be beneficial in this condition with the advantage of reduced ß-arrestin-mediated receptor internalisation with chronic use. EXPERIMENTAL APPROACH: Male Sprague-Dawley rats received either monocrotaline to induce pulmonary arterial hypertension or saline and then daily i.p. injections of either MM07 or saline for 21 days. The extent of disease was assessed by right ventricular catheterisation, cardiac MRI, and histological analysis of the pulmonary vasculature. The effect of MM07 on signalling, proliferation, and apoptosis of human pulmonary artery endothelial cells was investigated. KEY RESULTS: MM07 significantly reduced the elevation of right ventricular systolic pressure and hypertrophy induced by monocrotaline. Monocrotaline-induced changes in cardiac structure and function, including right ventricular end-systolic and end-diastolic volumes, ejection fraction, and left ventricular end-diastolic volume, were attenuated by MM07. MM07 also significantly reduced monocrotaline-induced muscularisation of small pulmonary blood vessels. MM07 stimulated endothelial NOS phosphorylation and expression, promoted proliferation, and attenuated apoptosis of human pulmonary arterial endothelial cells in vitro. CONCLUSION AND IMPLICATIONS: Our findings suggest that chronic treatment with MM07 is beneficial in this animal model of pulmonary arterial hypertension by addressing disease aetiology. These data support the development of G protein-biased apelin receptor agonists with improved pharmacokinetic profiles for use in human disease.


Subject(s)
Apelin Receptors/agonists , Disease Models, Animal , Monocrotaline/pharmacology , Pulmonary Arterial Hypertension/drug therapy , Animals , Apelin Receptors/metabolism , Male , Pulmonary Arterial Hypertension/metabolism , Rats , Rats, Sprague-Dawley
7.
Thorax ; 74(2): 164-176, 2019 02.
Article in English | MEDLINE | ID: mdl-30554141

ABSTRACT

INTRODUCTION: Skeletal muscle dysfunction is a clinically important complication of pulmonary arterial hypertension (PAH). Growth/differentiation factor 15 (GDF-15), a prognostic marker in PAH, has been associated with muscle loss in other conditions. We aimed to define the associations of GDF-15 and muscle wasting in PAH, to assess its utility as a biomarker of muscle loss and to investigate its downstream signalling pathway as a therapeutic target. METHODS: GDF-15 levels and measures of muscle size and strength were analysed in the monocrotaline (MCT) rat, Sugen/hypoxia mouse and in 30 patients with PAH. In C2C12 myotubes the downstream targets of GDF-15 were identified. The pathway elucidated was then antagonised in vivo. RESULTS: Circulating GDF-15 levels correlated with tibialis anterior (TA) muscle fibre diameter in the MCT rat (Pearson r=-0.61, p=0.003). In patients with PAH, plasma GDF-15 levels of <564 pg/L predicted those with preserved muscle strength with a sensitivity and specificity of ≥80%. In vitro GDF-15 stimulated an increase in phosphorylation of TGFß-activated kinase 1 (TAK1). Antagonising TAK1, with 5(Z)-7-oxozeaenol, in vitro and in vivo led to an increase in fibre diameter and a reduction in mRNA expression of atrogin-1 in both C2C12 cells and in the TA of animals who continued to grow. Circulating GDF-15 levels were also reduced in those animals which responded to treatment. CONCLUSIONS: Circulating GDF-15 is a biomarker of muscle loss in PAH that is responsive to treatment. TAK1 inhibition shows promise as a method by which muscle atrophy may be directly prevented in PAH. TRIAL REGISTRATION NUMBER: NCT01847716; Results.


Subject(s)
Growth Differentiation Factor 15/metabolism , Hypertension, Pulmonary/complications , MAP Kinase Kinase Kinases/metabolism , Muscular Atrophy/etiology , Transforming Growth Factor beta/metabolism , Adult , Animals , Biomarkers/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hypertension, Pulmonary/metabolism , Immunohistochemistry , Male , Mice , Middle Aged , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction
8.
Pulm Circ ; 8(4): 2045894018801642, 2018.
Article in English | MEDLINE | ID: mdl-30160594

ABSTRACT

Increasing evidence suggests that patients with pulmonary arterial hypertension (PAH) demonstrate abnormalities in the bone marrow (BM) and hematopoietic progenitor cells. In addition, PAH is associated with myeloproliferative diseases. We have previously demonstrated that low-dose lipopolysaccharide (LPS) is a potent stimulus for the development of PAH in the context of a genetic PAH mouse model of BMPR2 dysfunction. We hypothesized that the hematopoietic progenitor cells might be driving disease in this model. To test this hypothesis, we performed adoptive transfer of BM between wild-type (Ctrl) and heterozygous Bmpr2 null (Mut) mice. Sixteen weeks after BM reconstitution, mice were exposed to low-dose chronic LPS (0.5 mg/kg three times a week for six weeks). Mice underwent right heart catheterization and tissues were removed for histology. After chronic LPS dosing, Ctrl mice in receipt of Mut BM developed PAH, whereas Mut mice receiving Ctrl BM were protected from PAH. BM histology demonstrated an increase in megakaryocytes and there was an increase in circulating platelets in Ctrl mice receiving Mut BM. These findings demonstrate that the hematopoietic stem cell compartment is involved in the susceptibility to PAH in the Mut mouse. The results raise the possibility that hematopoietic stem cell transplantation might be a potential treatment strategy in genetic forms of PAH.

9.
FEBS Open Bio ; 8(3): 339-348, 2018 03.
Article in English | MEDLINE | ID: mdl-29511611

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterised by remodelling of the pulmonary vasculature leading to right ventricular hypertrophy. Here, we show that miR-322-5p (the rodent orthologue of miR-424-5p) expression is decreased in the right ventricle of monocrotaline-treated rats, a model of PAH, whereas a putative target insulin-like growth factor 1 (IGF-1) is increased. IGF-1 mRNA was enriched 16-fold in RNA immunoprecipitated with Ago2, indicating binding to miR-322-5p. In cell transfection experiments, miR-322-5p suppressed the activity of a luciferase reporter containing a section of the IGF-1 3' untranslated region (UTR) as well as IGF-1 mRNA and protein levels. Taken together, these data suggest that miR-322 targets IGF-1, a process downregulated in PAH-related RV hypertrophy.

10.
Circulation ; 135(12): 1160-1173, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-28137936

ABSTRACT

BACKGROUND: Elabela/toddler (ELA) is a critical cardiac developmental peptide that acts through the G-protein-coupled apelin receptor, despite lack of sequence similarity to the established ligand apelin. Our aim was to investigate the receptor pharmacology, expression pattern, and in vivo function of ELA peptides in the adult cardiovascular system, to seek evidence for alteration in pulmonary arterial hypertension (PAH) in which apelin signaling is downregulated, and to demonstrate attenuation of PAH severity with exogenous administration of ELA in a rat model. METHODS: In silico docking analysis, competition binding experiments, and downstream assays were used to characterize ELA receptor binding in human heart and signaling in cells expressing the apelin receptor. ELA expression in human cardiovascular tissues and plasma was determined using real-time quantitative polymerase chain reaction, dual-labeling immunofluorescent staining, and immunoassays. Acute cardiac effects of ELA-32 and [Pyr1]apelin-13 were assessed by MRI and cardiac catheterization in anesthetized rats. Cardiopulmonary human and rat tissues from PAH patients and monocrotaline- and Sugen/hypoxia-exposed rats were used to show changes in ELA expression in PAH. The effect of ELA treatment on cardiopulmonary remodeling in PAH was investigated in the monocrotaline rat model. RESULTS: ELA competed for binding of apelin in human heart with overlap for the 2 peptides indicated by in silico modeling. ELA activated G-protein- and ß-arrestin-dependent pathways. We detected ELA expression in human vascular endothelium and plasma. Comparable to apelin, ELA increased cardiac contractility, ejection fraction, and cardiac output and elicited vasodilatation in rat in vivo. ELA expression was reduced in cardiopulmonary tissues from PAH patients and PAH rat models, respectively. ELA treatment significantly attenuated elevation of right ventricular systolic pressure and right ventricular hypertrophy and pulmonary vascular remodeling in monocrotaline-exposed rats. CONCLUSIONS: These results show that ELA is an endogenous agonist of the human apelin receptor, exhibits a cardiovascular profile comparable to apelin, and is downregulated in human disease and rodent PAH models, and exogenous peptide can reduce the severity of cardiopulmonary remodeling and function in PAH in rats. This study provides additional proof of principle that an apelin receptor agonist may be of therapeutic use in PAH in humans.


Subject(s)
Hypertension, Pulmonary/drug therapy , Peptide Hormones/therapeutic use , Amino Acid Sequence , Animals , Apelin , Binding Sites , Catheterization , Disease Models, Animal , Down-Regulation/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Humans , Hypertension, Pulmonary/physiopathology , Intercellular Signaling Peptides and Proteins/agonists , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/therapeutic use , Male , Molecular Dynamics Simulation , Peptide Hormones/chemistry , Peptide Hormones/metabolism , Peptide Hormones/pharmacology , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley
11.
Nat Commun ; 8: 14079, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28084316

ABSTRACT

Heterozygous germ-line mutations in the bone morphogenetic protein type-II receptor (BMPR-II) gene underlie heritable pulmonary arterial hypertension (HPAH). Although inflammation promotes PAH, the mechanisms by which inflammation and BMPR-II dysfunction conspire to cause disease remain unknown. Here we identify that tumour necrosis factor-α (TNFα) selectively reduces BMPR-II transcription and mediates post-translational BMPR-II cleavage via the sheddases, ADAM10 and ADAM17 in pulmonary artery smooth muscle cells (PASMCs). TNFα-mediated suppression of BMPR-II subverts BMP signalling, leading to BMP6-mediated PASMC proliferation via preferential activation of an ALK2/ACTR-IIA signalling axis. Furthermore, TNFα, via SRC family kinases, increases pro-proliferative NOTCH2 signalling in HPAH PASMCs with reduced BMPR-II expression. We confirm this signalling switch in rodent models of PAH and demonstrate that anti-TNFα immunotherapy reverses disease progression, restoring normal BMP/NOTCH signalling. Collectively, these findings identify mechanisms by which BMP and TNFα signalling contribute to disease, and suggest a tractable approach for therapeutic intervention in PAH.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/metabolism , Familial Primary Pulmonary Hypertension/metabolism , Receptor, Notch2/metabolism , Receptor, Notch3/metabolism , Tumor Necrosis Factor-alpha/metabolism , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Animals , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Protein Receptors, Type II/genetics , Familial Primary Pulmonary Hypertension/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Rats , Receptor, Notch2/genetics , Receptor, Notch3/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/genetics
12.
J Immunol ; 197(8): 3302-3314, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27647829

ABSTRACT

Bone morphogenetic protein (BMP)9 is a circulating growth factor that is part of the TGF-ß superfamily and is an essential regulator of vascular endothelial homeostasis. Previous studies have suggested a role for BMP9 signaling in leukocyte recruitment to the endothelium, but the directionality of this effect and underlying mechanisms have not been elucidated. In this study, we report that BMP9 upregulates TLR4 expression in human endothelial cells and that BMP9 pretreatment synergistically increases human neutrophil recruitment to LPS-stimulated human endothelial monolayers in an in vitro flow adhesion assay. BMP9 alone did not induce neutrophil recruitment to the endothelium. We also show that E-selectin and VCAM-1, but not ICAM-1, are upregulated in response to BMP9 in LPS-stimulated human endothelial cells. Small interfering RNA knockdown of activin receptor-like kinase 1 inhibited the BMP9-induced expression of TLR4 and VCAM-1 and inhibited BMP9-induced human neutrophil recruitment to LPS-stimulated human endothelial cells. BMP9 treatment also increased leukocyte recruitment within the pulmonary circulation in a mouse acute endotoxemia model. These results demonstrate that although BMP9 alone does not influence leukocyte recruitment, it primes the vascular endothelium to mount a more intense response when challenged with LPS through an increase in TLR4, E-selectin, and VCAM-1 and ultimately through enhanced leukocyte recruitment.


Subject(s)
Endothelium, Vascular/cytology , Growth Differentiation Factors/metabolism , Leukocytes/cytology , Leukocytes/drug effects , Lipopolysaccharides/pharmacology , Animals , Cells, Cultured , Endothelium, Vascular/drug effects , Growth Differentiation Factor 2 , Humans , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL
13.
Proc Natl Acad Sci U S A ; 113(31): 8801-6, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27432976

ABSTRACT

Hypoxic pulmonary vasoconstriction is correlated with pulmonary vascular remodeling. The hypoxia-inducible transcription factors (HIFs) HIF-1α and HIF-2α are known to contribute to the process of hypoxic pulmonary vascular remodeling; however, the specific role of pulmonary endothelial HIF expression in this process, and in the physiological process of vasoconstriction in response to hypoxia, remains unclear. Here we show that pulmonary endothelial HIF-2α is a critical regulator of hypoxia-induced pulmonary arterial hypertension. The rise in right ventricular systolic pressure (RVSP) normally observed following chronic hypoxic exposure was absent in mice with pulmonary endothelial HIF-2α deletion. The RVSP of mice lacking HIF-2α in pulmonary endothelium after exposure to hypoxia was not significantly different from normoxic WT mice and much lower than the RVSP values seen in WT littermate controls and mice with pulmonary endothelial deletion of HIF-1α exposed to hypoxia. Endothelial HIF-2α deletion also protected mice from hypoxia remodeling. Pulmonary endothelial deletion of arginase-1, a downstream target of HIF-2α, likewise attenuated many of the pathophysiological symptoms associated with hypoxic pulmonary hypertension. We propose a mechanism whereby chronic hypoxia enhances HIF-2α stability, which causes increased arginase expression and dysregulates normal vascular NO homeostasis. These data offer new insight into the role of pulmonary endothelial HIF-2α in regulating the pulmonary vascular response to hypoxia.


Subject(s)
Arginase/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Endothelium, Vascular/metabolism , Hypertension, Pulmonary/metabolism , Animals , Arginase/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Hypoxia , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Hypertension, Pulmonary/genetics , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide/metabolism , Ventricular Function, Right/genetics , Ventricular Function, Right/physiology , Ventricular Pressure/genetics , Ventricular Pressure/physiology
14.
J Cell Biol ; 211(1): 53-61, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26459598

ABSTRACT

Caveolae are strikingly abundant in endothelial cells, yet the physiological functions of caveolae in endothelium and other tissues remain incompletely understood. Previous studies suggest a mechanoprotective role, but whether this is relevant under the mechanical forces experienced by endothelial cells in vivo is unclear. In this study we have sought to determine whether endothelial caveolae disassemble under increased hemodynamic forces, and whether caveolae help prevent acute rupture of the plasma membrane under these conditions. Experiments in cultured cells established biochemical assays for disassembly of caveolar protein complexes, and assays for acute loss of plasma membrane integrity. In vivo, we demonstrate that caveolae in endothelial cells of the lung and cardiac muscle disassemble in response to acute increases in cardiac output. Electron microscopy and two-photon imaging reveal that the plasma membrane of microvascular endothelial cells in caveolin 1(-/-) mice is much more susceptible to acute rupture when cardiac output is increased. These data imply that mechanoprotection through disassembly of caveolae is important for endothelial function in vivo.


Subject(s)
Cardiac Output , Caveolae/physiology , Endothelial Cells/physiology , Animals , Biomechanical Phenomena , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Membrane/physiology , Cells, Cultured , Endocytosis , Mice, Inbred C57BL , Mice, Knockout
15.
Am J Respir Crit Care Med ; 192(11): 1355-65, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26308618

ABSTRACT

RATIONALE: Schistosomiasis is a major cause of pulmonary arterial hypertension (PAH). Mutations in the bone morphogenetic protein type-II receptor (BMPR-II) are the commonest genetic cause of PAH. OBJECTIVES: To determine whether Bmpr2(+/-) mice are more susceptible to schistosomiasis-induced pulmonary vascular remodeling. METHODS: Wild-type (WT) and Bmpr2(+/-) mice were infected percutaneously with Schistosoma mansoni. At 17 weeks postinfection, right ventricular systolic pressure and liver and lung egg counts were measured. Serum, lung and liver cytokine, pulmonary vascular remodeling, and liver histology were assessed. MEASUREMENTS AND MAIN RESULTS: By 17 weeks postinfection, there was a significant increase in pulmonary vascular remodeling in infected mice. This was greater in Bmpr2(+/-) mice and was associated with an increase in egg deposition and cytokine expression, which induced pulmonary arterial smooth muscle cell proliferation, in the lungs of these mice. Interestingly, Bmpr2(+/-) mice demonstrated dilatation of the hepatic central vein at baseline and postinfection, compared with WT. Bmpr2(+/-) mice also showed significant dilatation of the liver sinusoids and an increase in inflammatory cells surrounding the central hepatic vein, compared with WT. This is consistent with an increase in the transhepatic passage of eggs. CONCLUSIONS: This study has shown that levels of BMPR-II expression modify the pulmonary vascular response to chronic schistosomiasis. The likely mechanism involves the increased passage of eggs to the lungs, caused by altered diameter of the hepatic veins and sinusoids in Bmpr2(+/-) mice. Genetically determined differences in the remodeling of hepatic vessels may represent a new risk factor for PAH associated with schistosomiasis.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II , Hypertension, Pulmonary/physiopathology , Liver/parasitology , Pulmonary Artery/physiopathology , Schistosomiasis/physiopathology , Vascular Remodeling/genetics , Animals , Cell Proliferation , Disease Models, Animal , Female , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/parasitology , Mice , Pulmonary Artery/parasitology , Schistosoma mansoni , Schistosomiasis/genetics , Signal Transduction , Vascular Remodeling/physiology
16.
Am J Respir Crit Care Med ; 192(7): 859-72, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26073741

ABSTRACT

RATIONALE: Mutations in bone morphogenetic protein receptor type II (BMPR-II) underlie most cases of heritable pulmonary arterial hypertension (PAH). However, disease penetrance is only 20-30%, suggesting a requirement for additional triggers. Inflammation is emerging as a key disease-related factor in PAH, but to date there is no clear mechanism linking BMPR-II deficiency and inflammation. OBJECTIVES: To establish a direct link between BMPR-II deficiency, a consequentially heightened inflammatory response, and development of PAH. METHODS: We used pulmonary artery smooth muscle cells from Bmpr2(+/-) mice and patients with BMPR2 mutations and compared them with wild-type controls. For the in vivo model, we used mice heterozygous for a null allele in Bmpr2 (Bmpr2(+/-)) and wild-type littermates. MEASUREMENTS AND MAIN RESULTS: Acute exposure to LPS increased lung and circulating IL-6 and KC (IL-8 analog) levels in Bmpr2(+/-) mice to a greater extent than in wild-type controls. Similarly, pulmonary artery smooth muscle cells from Bmpr2(+/-) mice and patients with BMPR2 mutations produced higher levels of IL-6 and KC/IL-8 after lipopolysaccharide stimulation compared with controls. BMPR-II deficiency in mouse and human pulmonary artery smooth muscle cells was associated with increased phospho-STAT3 and loss of extracellular superoxide dismutase. Chronic lipopolysaccharide administration caused pulmonary hypertension in Bmpr2(+/-) mice but not in wild-type littermates. Coadministration of tempol, a superoxide dismutase mimetic, ameliorated the exaggerated inflammatory response and prevented development of PAH. CONCLUSIONS: This study demonstrates that BMPR-II deficiency promotes an exaggerated inflammatory response in vitro and in vivo, which can instigate development of pulmonary hypertension.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/deficiency , Cytokines/biosynthesis , Hypertension, Pulmonary/physiopathology , Animals , Antioxidants/therapeutic use , Bone Morphogenetic Protein Receptors, Type II/metabolism , Cyclic N-Oxides/therapeutic use , Fenoterol , Genetic Predisposition to Disease , Humans , Hypertension, Pulmonary/genetics , Immunohistochemistry , Mice, Inbred Strains , Spin Labels , Superoxide Dismutase/physiology
18.
Am J Respir Crit Care Med ; 184(4): 467-73, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21659614

ABSTRACT

RATIONALE: Schistosomiasis is the most common worldwide cause of pulmonary arterial hypertension. The anti-schistosome drug praziquantel has been shown to reverse the liver fibrosis associated with Schistosoma mansoni in mice. OBJECTIVES: We sought to determine whether praziquantel reverses established pulmonary vascular remodeling and pulmonary hypertension in a mouse model of S. mansoni. METHODS: Mice were infected percutaneously with S. mansoni. At 17 weeks after infection mice were either killed or received two doses of praziquantel or vehicle by oral gavage. Treated mice were studied at 25 weeks after infection. MEASUREMENTS AND MAIN RESULTS: Vehicle-treated mice demonstrated significant increases in right ventricular systolic pressures (RVSP) and right ventricular hypertrophy (RVH) at 25 weeks, accompanied by pulmonary vascular remodeling. The degree of vascular remodeling correlated with proximity to granulomas. The elevation of RVSP and RVH at 25 weeks was dependent on the presence of eggs in the lung. Praziquantel eliminated the production of eggs in feces and led to clearance of eggs from the lung and to a lesser extent from liver. Praziquantel prevented the rise in RVSP and RVH seen in vehicle-treated mice and reversed established pulmonary vascular remodeling. Praziquantel significantly reduced lung mRNA expression of IL-13, IL-8, and IL-4, but did not reduce serum cytokine levels. CONCLUSIONS: The development of pulmonary hypertension associated with S. mansoni infection can be prevented by praziquantel, and established vascular remodeling can be reversed. The mechanism involves clearance of lung eggs and reduced local expression of lung cytokines.


Subject(s)
Anthelmintics/pharmacology , Blood Vessels/drug effects , Hypertension, Pulmonary/physiopathology , Lung/blood supply , Praziquantel/pharmacology , Schistosomiasis mansoni/complications , Schistosomiasis mansoni/physiopathology , Animals , Blood Pressure , Cytokines/metabolism , Down-Regulation , Female , Granuloma/parasitology , Granuloma/pathology , Heart Ventricles/physiopathology , Hypertension, Pulmonary/parasitology , Hypertrophy, Right Ventricular/parasitology , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/prevention & control , Inflammation Mediators/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Interleukin-8/metabolism , Liver/parasitology , Lung/metabolism , Lung/parasitology , Mice , Mice, Inbred C57BL , Ovum/drug effects , RNA, Messenger/metabolism , Schistosoma mansoni/drug effects , Schistosomiasis mansoni/pathology
19.
Expert Rev Anti Infect Ther ; 8(12): 1467-73, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21133670

ABSTRACT

In this article we focus on the pathogenesis and clinical characteristics of schistosomiasis infection on the lung vasculature. Overall, the basic biology and understanding of Schistosoma immune responses and their effect on the cardiopulmonary system is limited in both animal and human models, which hinders clinical care and drug development. The inflammatory response to the eggs in the lung appears to contribute to the remodeling of the pulmonary vessels. Portal hypertension caused by parasitemia also appears to contribute to the development of pathophysiologic alterations of the pulmonary vascular bed. Antischistosomal therapy, praziquantel, used for pulmonary hypertension secondary to schistosomiasis usually has no effect, but it is given to prevent further progression of disease. Currently, there are no clinical trials for the treatment of pulmonary vascular disease secondary to schistosomiasis. Specialty drugs such as phosphodiesterase type 5 or tyrosine kinase inhibitors exhibit some interesting activity, yet are prohibitively expensive, lack safety and efficacy studies in schistosomiasis endemic populations, and tend to be limited by safety, efficacy, route of administration and compliance problems.


Subject(s)
Lung/blood supply , Schistosomiasis/complications , Schistosomiasis/drug therapy , Animals , Familial Primary Pulmonary Hypertension , Female , Humans , Hypertension, Portal/complications , Hypertension, Portal/drug therapy , Hypertension, Portal/parasitology , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/parasitology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Inflammation , Lung/pathology , Male , Praziquantel/therapeutic use , Schistosoma/drug effects , Schistosoma/growth & development , Schistosomiasis/epidemiology
20.
Am J Respir Crit Care Med ; 181(3): 279-88, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19965814

ABSTRACT

RATIONALE: Schistosomiasis is considered to be the most common worldwide cause of pulmonary hypertension. At present there is no well-characterized animal model to study the pathobiology of this important condition. OBJECTIVES: To develop a mouse model of schistosomiasis, characterize the extent of pulmonary vascular remodeling, and determine the potential role of inflammatory cytokines. METHODS: Mice (C57/Bl6) were infected transcutaneously with a high dose (approximately 75-100 cercariae) or a low dose (approximately 30 cercariae) of Schistosoma mansoni, and the development of lung and liver pathology was studied in the subacute (high-dose) and chronic (low-dose) settings. MEASUREMENTS AND MAIN RESULTS: In the subacute setting, mice showed few eggs in the lungs and no evidence of pulmonary vascular remodeling. In contrast, chronically infected animals had a much greater lung egg burden and developed marked pulmonary vascular remodeling accompanied by perivascular inflammation from 12 weeks onwards. In addition, we observed the presence of plexiform-like lesions in these mice. Lung egg burden correlated with both liver egg burden and right ventricular (RV) index in the chronic group, although significant RV hypertrophy was lacking. Plasma Th1 and Th2 cytokines increased with time in the chronic group and correlated with the degree of pulmonary vascular remodeling. CONCLUSIONS: This study provides evidence for extensive pulmonary vascular remodeling, despite the absence of RV hypertrophy, in a mouse model of schistosomiasis, including the formation of plexiform-like lesions. Inflammatory cytokines and lung egg burden may contribute to vascular lesion formation.


Subject(s)
Cytokines/metabolism , Hypertension, Pulmonary/pathology , Lung/parasitology , Pulmonary Artery/pathology , Schistosoma mansoni/isolation & purification , Schistosomiasis/pathology , Vascular Resistance/physiology , Animals , Disease Models, Animal , Female , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Immunohistochemistry , Lung/pathology , Lung/physiopathology , Mice , Mice, Inbred C57BL , Photomicrography , Schistosomiasis/complications , Schistosomiasis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...