Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters

Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 327(1): L102-L113, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38501173

ABSTRACT

We have reported previously that during hypoxia exposure, the expression of mature miR-17∼92 was first upregulated and then downregulated in pulmonary artery smooth muscle cells (PASMC) and in mouse lungs in vitro and in vivo. Here, we investigated the mechanisms regulating this biphasic expression of miR-17∼92 in PASMC in hypoxia. We measured the level of primary miR-17∼92 in PASMC during hypoxia exposure and found that short-term hypoxia exposure (3% O2, 6 h) induced the level of primary miR-17∼92, whereas long-term hypoxia exposure (3% O2, 24 h) decreased its level, suggesting a biphasic regulation of miR-17∼92 expression at the transcriptional level. We found that short-term hypoxia-induced upregulation of miR-17∼92 was hypoxia-inducible factor 1α (HIF1α) and E2F1 dependent. Two HIF1α binding sites on miR-17∼92 promoter were identified. We also found that long-term hypoxia-induced suppression of miR-17∼92 expression could be restored by silencing of p53. Mutation of the p53-binding sites in the miR-17∼92 promoter increased miR-17∼92 promoter activity in both normoxia and hypoxia. Our findings suggest that the biphasic transcriptional regulation of miR-17∼92 during hypoxia is controlled by HIF1/E2F1 and p53 in PASMC: during short-term hypoxia exposure, stabilization of HIF1 and induction of E2F1 induce the transcription of miR-17∼92, whereas during long-term hypoxia exposure, hyperphosphorylation of p53 suppresses the expression of miR-17∼92.NEW & NOTEWORTHY We showed that the biphasic transcriptional regulation of miR-17∼92 during hypoxia is controlled by two distinct mechanisms: during short-term hypoxia exposure, induction of HIF1 and E2F1 upregulates miR-17∼92. Longer hypoxia exposure induces hyperphosphorylation of p53 at ser15, which leads to its binding to miR-17∼92 promoter and inhibition of its expression. Our findings provide novel insights into the spatiotemporal regulation of miR-17∼92 that may play a role in the development of human lung diseases including pulmonary hypertension (PH).


Subject(s)
E2F1 Transcription Factor , Hypoxia-Inducible Factor 1, alpha Subunit , MicroRNAs , Pulmonary Artery , Tumor Suppressor Protein p53 , MicroRNAs/genetics , MicroRNAs/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Phosphorylation , Humans , Animals , E2F1 Transcription Factor/metabolism , E2F1 Transcription Factor/genetics , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Transcription, Genetic , Cell Hypoxia/genetics , Myocytes, Smooth Muscle/metabolism , Promoter Regions, Genetic/genetics , Mice , Hypoxia/metabolism , Hypoxia/genetics , Serine/metabolism , Gene Expression Regulation , Cells, Cultured
2.
J Pediatr ; 252: 131-140.e3, 2023 01.
Article in English | MEDLINE | ID: mdl-36027975

ABSTRACT

OBJECTIVE: To characterize distinct comorbidities, outcomes, and treatment patterns in children with Down syndrome and pulmonary hypertension in a large, multicenter pediatric pulmonary hypertension registry. STUDY DESIGN: We analyzed data from the Pediatric Pulmonary Hypertension Network (PPHNet) Registry, comparing demographic and clinical characteristics of children with Down syndrome and children without Down syndrome. We examined factors associated with pulmonary hypertension resolution and a composite outcome of pulmonary hypertension severity in the cohort with Down syndrome. RESULTS: Of 1475 pediatric patients with pulmonary hypertension, 158 (11%) had Down syndrome. The median age at diagnosis of pulmonary hypertension in patients with Down syndrome was 0.49 year (IQR, 0.21-1.77 years), similar to that in patients without Down syndrome. There was no difference in rates of cardiac catheterization and prescribed pulmonary hypertension medications in children with Down syndrome and those without Down syndrome. Comorbidities in Down syndrome included congenital heart disease (95%; repaired in 68%), sleep apnea (56%), prematurity (49%), recurrent respiratory exacerbations (35%), gastroesophageal reflux (38%), and aspiration (31%). Pulmonary hypertension resolved in 43% after 3 years, associated with a diagnosis of pulmonary hypertension at age <6 months (54% vs 29%; P = .002) and a pretricuspid shunt (65% vs 38%; P = .02). Five-year transplantation-free survival was 88% (95% CI, 80%-97%). Tracheostomy (hazard ratio [HR], 3.29; 95% CI, 1.61-6.69) and reflux medication use (HR, 2.08; 95% CI, 1.11-3.90) were independently associated with a composite outcome of severe pulmonary hypertension. CONCLUSIONS: Despite high rates of cardiac and respiratory comorbidities that influence the severity of pulmonary hypertension, children with Down syndrome-associated pulmonary hypertension generally have a survival rate similar to that of children with non-Down syndrome-associated pulmonary hypertension. Resolution of pulmonary hypertension is common but reduced in children with complicated respiratory comorbidities.


Subject(s)
Down Syndrome , Gastroesophageal Reflux , Heart Defects, Congenital , Hypertension, Pulmonary , Child , Humans , Infant , Hypertension, Pulmonary/epidemiology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/therapy , Retrospective Studies , Down Syndrome/complications , Heart Defects, Congenital/surgery , Registries , Gastroesophageal Reflux/complications
3.
Arterioscler Thromb Vasc Biol ; 39(6): 1191-1202, 2019 06.
Article in English | MEDLINE | ID: mdl-30943774

ABSTRACT

Objective- To determine whether pulmonary arterial hypertension is associated with endothelial cell (EC)-Cav-1 (caveolin-1) depletion, EC-derived extracellular vesicle cross talk with macrophages, and proliferation of Cav-1 depleted ECs via TGF-ß (transforming growth factor-ß) signaling. Approach and Results- Pulmonary vascular disease was induced in Sprague-Dawley rats by exposure to a single injection of VEGFRII (vascular endothelial growth factor receptor II) antagonist SU5416 (Su) followed by hypoxia (Hx) plus normoxia (4 weeks each-HxSu model) and in WT (wild type; Tie2.Cre-; Cav1 lox/lox) and EC- Cav1-/- (Tie2.Cre+; Cav1 fl/fl) mice (Hx: 4 weeks). We observed reduced lung Cav-1 expression in the HxSu rat model in association with increased Cav-1+ extracellular vesicle shedding into the circulation. Whereas WT mice exposed to hypoxia exhibited increased right ventricular systolic pressure and pulmonary microvascular thickening compared with the group maintained in normoxia, the remodeling was further increased in EC- Cav1-/- mice indicating EC Cav-1 expression protects against hypoxia-induced pulmonary hypertension. Depletion of EC Cav-1 was associated with reduced BMPRII (bone morphogenetic protein receptor II) expression, increased macrophage-dependent TGF-ß production, and activation of pSMAD2/3 signaling in the lung. In vitro, in the absence of Cav-1, eNOS (endothelial NO synthase) dysfunction was implicated in the mechanism of EC phenotype switching. Finally, reduced expression of EC Cav-1 in lung histological sections from human pulmonary arterial hypertension donors was associated with increased plasma concentration of Cav-1, extracellular vesicles, and TGF-ß, indicating Cav-1 may be a plasma biomarker of vascular injury and key determinant of TGF-ß-induced pulmonary vascular remodeling. Conclusions- EC Cav-1 depletion occurs, in part, via Cav-1+ extracellular vesicle shedding into the circulation, which contributes to increased TGF-ß signaling, EC proliferation, vascular remodeling, and pulmonary arterial hypertension.


Subject(s)
Caveolin 1/deficiency , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Pulmonary Arterial Hypertension/metabolism , Transforming Growth Factor beta/metabolism , Vascular Remodeling , Adolescent , Adult , Aged , Animals , Bone Morphogenetic Protein Receptors, Type II/metabolism , Case-Control Studies , Caveolin 1/genetics , Cell Proliferation , Disease Models, Animal , Endothelial Cells/pathology , Extracellular Vesicles/pathology , Female , Humans , Hypoxia/complications , Indoles , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/pathology , Pyrroles , Rats, Sprague-Dawley , Signal Transduction , Smad Proteins/metabolism , Young Adult
4.
J Pediatr ; 211: 63-71.e6, 2019 08.
Article in English | MEDLINE | ID: mdl-31176455

ABSTRACT

OBJECTIVE: To investigate racial and ethnic differences in pulmonary hypertension subtypes and survival differences in a pediatric population. STUDY DESIGN: This was a retrospective analysis of a cohort of patients with pulmonary hypertension (aged ≤18 years) enrolled in the Pediatric Pulmonary Hypertension Network registry between 2014 and 2018, comprising patients at eight Pediatric Centers throughout North America (n = 1417). RESULTS: Among children diagnosed after the neonatal period, pulmonary arterial hypertension was more prevalent among Asians (OR, 1.83; 95% CI, 1.21-2.79; P = .0045), lung disease-associated pulmonary hypertension among blacks (OR, 2.09; 95% CI, 1.48-2.95; P < .0001), idiopathic pulmonary arterial hypertension among whites (OR, 1.58; 95% CI, 1.06-2.41; P = .0289), and pulmonary veno-occlusive disease among Hispanics (OR, 6.11; 95% CI, 1.34-31.3; P = .0184). Among neonates, persistent pulmonary hypertension of the newborn (OR, 4.07; 95% CI, 1.54-10.0; P = .0029) and bronchopulmonary dysplasia (OR, 8.11; 95% CI, 3.28-19.8; P < .0001) were more prevalent among blacks, and congenital diaphragmatic hernia was more prevalent among whites (OR, 2.29; 95% CI, 1.25-4.18; P = .0070). An increased mortality risk was observed among blacks (HR, 1.99; 95% CI, 1.03-3.84; P = .0396), driven primarily by the heightened mortality risk among those with lung disease-associated pulmonary hypertension (HR, 2.84; 95% CI, 1.15-7.04; P = .0241). CONCLUSIONS: We found significant racial variability in the prevalence of pulmonary hypertension subtypes and survival outcomes among children with pulmonary hypertension. Given the substantial burden of this disease, further studies to validate phenotypic differences and to understand the underlying causes of survival disparities between racial and ethnic groups are warranted.


Subject(s)
Pediatrics/methods , Pulmonary Arterial Hypertension/ethnology , Registries , Adolescent , Black or African American , Child , Child, Preschool , Ethnicity , Female , Hispanic or Latino , Humans , Infant , Infant, Newborn , Male , North America/epidemiology , Prevalence , Pulmonary Arterial Hypertension/diagnosis , Pulmonary Arterial Hypertension/mortality , Racial Groups , Regression Analysis , Reproducibility of Results , Retrospective Studies , Survival Analysis , Treatment Outcome , White People
5.
Am J Respir Cell Mol Biol ; 58(2): 181-193, 2018 02.
Article in English | MEDLINE | ID: mdl-28915060

ABSTRACT

Pulmonary artery hypertension (PAH) is a rare and fatal disorder that involves extensive remodeling of the pulmonary arteries mediated by hyperproliferation of pulmonary artery smooth muscle cells (PASMCs). Aberrant platelet-derived growth factor (PDGF) activity can lead to hyperproliferation of PASMCs; however, little is known about the role of long noncoding RNA (lncRNA) in this process. Using RNA sequencing, we identified 725 lncRNAs in rat PASMCs, 95 of which were expressed differentially in response to PDGF-BB treatment. Depletion of four lncRNAs affected the proliferation of rat PASMCs as measured by 5-ethynyl-2'-deoxyuridine incorporation assay. Among these, one lncRNA, named LnRPT (lncRNA regulated by PDGF and transforming growth factor ß), was found to be the most potent in promoting the proliferation of PASMCs when knocked down. In contrast, proliferation of PASMCs was repressed when LnRPT was overexpressed. Mechanistically, LnRPT inhibited the expression of two genes involved in the Notch signaling pathway (notch3 and jag1) as well as the cell-cycle-regulating gene ccna2. In addition, downregulation of LnRPT induced by PDGF-BB was abrogated when phosphatidylinositol 3'-kinase activity was inhibited with pictilisib. Downregulation of LnRPT was also observed in the pulmonary arteries of rats with monocrotaline-induced PAH. This study provides novel insights into the effects of PDGF-BB on lncRNA expression in PASMCs, and identifies one lncRNA, LnRPT, that plays a role in PAH development as a regulator of PASMC proliferation by mediating the Notch signaling pathway and cell cycle.


Subject(s)
Hypertension, Pulmonary/pathology , Myocytes, Smooth Muscle/pathology , Proto-Oncogene Proteins c-sis/metabolism , Pulmonary Artery/pathology , RNA, Long Noncoding/genetics , Animals , Becaplermin , Cell Proliferation , Cells, Cultured , Cyclin A2/metabolism , Down-Regulation , Indazoles/pharmacology , Jagged-1 Protein/metabolism , Male , Monocrotaline/pharmacology , Myocytes, Smooth Muscle/cytology , Phosphoinositide-3 Kinase Inhibitors , Platelet-Derived Growth Factor/metabolism , Pulmonary Artery/cytology , RNA Interference , RNA, Long Noncoding/biosynthesis , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Receptor, Notch3/metabolism , Sulfonamides/pharmacology
6.
Am J Physiol Lung Cell Mol Physiol ; 315(2): L149-L161, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29644896

ABSTRACT

We have previously reported that miR-17~92 is critically involved in the pathogenesis of pulmonary hypertension (PH). We also identified two novel mR-17/20a direct targets, PDZ and LIM domain protein 5 (PDLIM5) and prolyl hydroxylase 2 (PHD2), and elucidated the signaling pathways by which PDLIM5 and PHD2 regulate functions of pulmonary artery smooth muscle cells (PASMCs). In addition, we have shown that plasminogen activator inhibitor-1 (PAI-1) is also downregulated in PASMCs that overexpress miR-17~92. However, it is unclear whether PAI-1 is a direct target of miR-17~92 and whether it plays a role in regulating the PASMC phenotype. In this study, we have identified PAI-1 as a novel target of miR-19a/b, two members of the miR-17~92 cluster. We found that the 3'-untranslated region (UTR) of PAI-1 contains a miR-19a/b binding site and that miR-19a/b can target this site to suppress PAI-1 protein expression. MiR-17/20a, two other members of miR-17~92, may also indirectly suppress PAI-1 expression through PDLIM5. PAI-1 is a negative regulator of miR-17~92-mediated PASMC proliferation. Silencing of PAI-1 induces Smad2/calponin signaling in PASMCs, suggesting that PAI-1 is a negative regulator of the PASMC contractile phenotype. We also found that PAI-1 is essential for the metabolic gene expression in PASMCs. Furthermore, although there is no significant change in PAI-1 levels in PASMCs isolated from idiopathic pulmonary arterial hypertension and associated pulmonary arterial hypertension patients, PAI-1 is downregulated in hypoxia/Sugen-induced hypertensive rat lungs. These results suggest that miR-17~92 regulates the PASMC contractile phenotype and proliferation coordinately and synergistically by direct and indirect targeting of PAI-1.


Subject(s)
MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Pulmonary Artery/metabolism , Signal Transduction , 3' Untranslated Regions , Animals , Cell Proliferation , Gene Expression Regulation , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Male , MicroRNAs/genetics , Muscle Contraction/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Plasminogen Activator Inhibitor 1/genetics , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley
7.
Physiol Rev ; 90(4): 1291-335, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20959617

ABSTRACT

During the development of the pulmonary vasculature in the fetus, many structural and functional changes occur to prepare the lung for the transition to air breathing. The development of the pulmonary circulation is genetically controlled by an array of mitogenic factors in a temporo-spatial order. With advancing gestation, pulmonary vessels acquire increased vasoreactivity. The fetal pulmonary vasculature is exposed to a low oxygen tension environment that promotes high intrinsic myogenic tone and high vasocontractility. At birth, a dramatic reduction in pulmonary arterial pressure and resistance occurs with an increase in oxygen tension and blood flow. The striking hemodynamic differences in the pulmonary circulation of the fetus and newborn are regulated by various factors and vasoactive agents. Among them, nitric oxide, endothelin-1, and prostaglandin I(2) are mainly derived from endothelial cells and exert their effects via cGMP, cAMP, and Rho kinase signaling pathways. Alterations in these signaling pathways may lead to vascular remodeling, high vasocontractility, and persistent pulmonary hypertension of the newborn.


Subject(s)
Fetus/physiology , Infant, Newborn/physiology , Lung/blood supply , Pulmonary Circulation/physiology , Gene Expression Regulation, Developmental/physiology , Humans , Lung/embryology , Lung/growth & development , Respiration
9.
Am J Respir Cell Mol Biol ; 54(4): 451-60, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26744837

ABSTRACT

In the pulmonary vasculature, the endothelial and smooth muscle cells are two key cell types that play a major role in the pathobiology of pulmonary vascular disease and pulmonary hypertension. The normal interactions between these two cell types are important for the homeostasis of the pulmonary circulation, and any aberrant interaction between them may lead to various disease states including pulmonary vascular remodeling and pulmonary hypertension. It is well recognized that the endothelial cell can regulate the function of the underlying smooth muscle cell by releasing various bioactive agents such as nitric oxide and endothelin-1. In addition to such paracrine regulation, other mechanisms exist by which there is cross-talk between these two cell types, including communication via the myoendothelial injunctions and information transfer via extracellular vesicles. Emerging evidence suggests that these nonparacrine mechanisms play an important role in the regulation of pulmonary vascular tone and the determination of cell phenotype and that they are critically involved in the pathobiology of pulmonary hypertension.


Subject(s)
Endothelium, Vascular/pathology , Hypertension, Pulmonary/pathology , Muscle, Smooth, Vascular/pathology , Humans , Signal Transduction
10.
Am J Respir Cell Mol Biol ; 55(6): 779-791, 2016 12.
Article in English | MEDLINE | ID: mdl-27438786

ABSTRACT

Insulin-like growth factor (IGF)-1 is a potent mitogen of vascular smooth muscle cells (SMCs), but its role in pulmonary vascular remodeling associated with pulmonary hypertension (PH) is not clear. In an earlier study, we implicated IGF-1 in the pathogenesis of hypoxia-induced PH in neonatal mice. In this study, we hypothesized that hypoxia-induced up-regulation of IGF-1 in vascular smooth muscle is directly responsible for pulmonary vascular remodeling and PH. We studied neonatal and adult mice with smooth muscle-specific deletion of IGF-1 and also used an inhibitor of IGF-1 receptor (IGF-1R), OSI-906, in neonatal mice. We found that, in neonatal mice, SMC-specific deletion of IGF-1 or IGF-1R inhibition with OSI-906 attenuated hypoxia-induced pulmonary vascular remodeling in small arteries, right ventricular hypertrophy, and right ventricular systolic pressure. Pulmonary arterial SMCs from IGF-1-deleted mice or after OSI-906 treatment exhibited reduced proliferative potential. However, in adult mice, smooth muscle-specific deletion of IGF-1 had no effect on hypoxia-induced PH. Our data suggest that vascular smooth muscle-derived IGF-1 plays a critical role in hypoxia-induced PH in neonatal mice but not in adult mice. We speculate that the IGF-1/IGF-1R axis is important in pathogenesis of PH in the developing lung and may be amenable to therapeutic manipulation in this age group.


Subject(s)
Hypertension, Pulmonary/complications , Hypertension, Pulmonary/metabolism , Hypoxia/complications , Hypoxia/metabolism , Insulin-Like Growth Factor I/metabolism , Muscle, Smooth/metabolism , Animals , Animals, Newborn , Blood Pressure/drug effects , Cell Proliferation/drug effects , Chronic Disease , Gene Deletion , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/pathology , Hypoxia/physiopathology , Imidazoles/pharmacology , Insulin-Like Growth Factor I/deficiency , Insulin-Like Growth Factor I/genetics , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth/drug effects , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Organ Specificity/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Pyrazines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 1/metabolism , Systole/drug effects , Up-Regulation/drug effects , Vascular Remodeling/drug effects
11.
Circulation ; 132(21): 2037-99, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26534956

ABSTRACT

Pulmonary hypertension is associated with diverse cardiac, pulmonary, and systemic diseases in neonates, infants, and older children and contributes to significant morbidity and mortality. However, current approaches to caring for pediatric patients with pulmonary hypertension have been limited by the lack of consensus guidelines from experts in the field. In a joint effort from the American Heart Association and American Thoracic Society, a panel of experienced clinicians and clinician-scientists was assembled to review the current literature and to make recommendations on the diagnosis, evaluation, and treatment of pediatric pulmonary hypertension. This publication presents the results of extensive literature reviews, discussions, and formal scoring of recommendations for the care of children with pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary/therapy , Cardiovascular Agents/therapeutic use , Child , Child, Preschool , Combined Modality Therapy , Diagnostic Imaging/methods , Disease Management , Extracorporeal Membrane Oxygenation , Genetic Counseling , Heart Defects, Congenital/complications , Heart Defects, Congenital/therapy , Hernias, Diaphragmatic, Congenital/complications , Hernias, Diaphragmatic, Congenital/therapy , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Infant , Infant, Newborn , Lung/embryology , Lung Transplantation , Nitric Oxide/administration & dosage , Nitric Oxide/therapeutic use , Oxygen Inhalation Therapy , Persistent Fetal Circulation Syndrome/diagnosis , Persistent Fetal Circulation Syndrome/therapy , Postoperative Complications/therapy , Respiration, Artificial/adverse effects , Respiration, Artificial/methods , Ventilator-Induced Lung Injury/prevention & control
12.
Am J Respir Crit Care Med ; 191(6): 678-92, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25647182

ABSTRACT

RATIONALE: Recent studies suggest that microRNAs (miRNAs) play important roles in regulation of pulmonary artery smooth muscle cell (PASMC) phenotype and are implicated in pulmonary arterial hypertension (PAH). However, the underlying molecular mechanisms remain elusive. OBJECTIVES: This study aims to understand the mechanisms regulating PASMC proliferation and differentiation by microRNA-17∼92 (miR-17∼92) and to elucidate its implication in PAH. METHODS: We generated smooth muscle cell (SMC)-specific miR-17∼92 and PDZ and LIM domain 5 (PDLIM5) knockout mice and overexpressed miR-17∼92 and PDLIM5 by injection of miR-17∼92 mimics or PDLIM5-V5-His plasmids and measured their responses to hypoxia. We used miR-17∼92 mimics, inhibitors, overexpression vectors, small interfering RNAs against PDLIM5, Smad, and transforming growth factor (TGF)-ß to determine the role of miR-17∼92 and its downstream targets in PASMC proliferation and differentiation. MEASUREMENTS AND MAIN RESULTS: We found that human PASMC (HPASMC) from patients with PAH expressed decreased levels of the miR-17∼92 cluster, TGF-ß, and SMC markers. Overexpression of miR-17∼92 increased and restored the expression of TGF-ß3, Smad3, and SMC markers in HPASMC of normal subjects and patients with idiopathic PAH, respectively. Knockdown of Smad3 but not Smad2 prevented miR-17∼92-induced expression of SMC markers. SMC-specific knockout of miR-17∼92 attenuated hypoxia-induced pulmonary hypertension (PH) in mice, whereas reconstitution of miR-17∼92 restored hypoxia-induced PH in these mice. We also found that PDLIM5 is a direct target of miR-17/20a, and hypertensive HPASMC and mouse PASMC expressed elevated PDLIM5 levels. Suppression of PDLIM5 increased expression of SMC markers and enhanced TGF-ß/Smad2/3 activity in vitro and enhanced hypoxia-induced PH in vivo, whereas overexpression of PDLIM5 attenuated hypoxia-induced PH. CONCLUSIONS: We provided the first evidence that miR-17∼92 inhibits PDLIM5 to induce the TGF-ß3/SMAD3 pathway, contributing to the pathogenesis of PAH.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Hypertension, Pulmonary/etiology , MicroRNAs/physiology , Microfilament Proteins/physiology , Muscle, Smooth, Vascular/cytology , Animals , Mass Spectrometry , Mice , Mice, Knockout , Transforming Growth Factors/physiology
14.
Am J Respir Cell Mol Biol ; 52(2): 139-51, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25192340

ABSTRACT

Pulmonary arterial hypertension (PAH) is a devastating disease without effective treatment. Despite decades of research and the development of novel treatments, PAH remains a fatal disease, suggesting an urgent need for better understanding of the pathogenesis of PAH. Recent studies suggest that microRNAs (miRNAs) are dysregulated in patients with PAH and in experimental pulmonary hypertension. Furthermore, normalization of a few miRNAs is reported to inhibit experimental pulmonary hypertension. We have reviewed the current knowledge about miRNA biogenesis, miRNA expression pattern, and their roles in regulation of pulmonary artery smooth muscle cells, endothelial cells, and fibroblasts. We have also identified emerging trends in our understanding of the role of miRNAs in the pathogenesis of PAH and propose future studies that might lead to novel therapeutic strategies for the treatment of PAH.


Subject(s)
Endothelial Cells/metabolism , Hypertension, Pulmonary/metabolism , MicroRNAs/genetics , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , Animals , Fibroblasts/metabolism , Humans , Hypertension, Pulmonary/genetics , MicroRNAs/metabolism
15.
Am J Physiol Cell Physiol ; 308(9): C720-8, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25696812

ABSTRACT

The fast response to stimuli and subsequent activation of the nuclear factor of activated T cells (NFAT) signaling pathway play an essential role in human T cell functions. MicroRNAs (miRNAs) are increasingly implicated in regulation of numerous biological and pathological processes. In this study we demonstrate a novel function of miRNA-9 (miR-9) in regulation of the NFAT signaling pathway. Upon PMA-ionomycin stimulation, miR-9 was markedly increased, consistent with NFAT activation. Overexpression of miR-9 significantly enhanced NFAT activity and accelerated NFAT dephosphorylation and its nuclear translocation in response to PMA-ionomycin. Karyopherin-ß1 (KPNB1, a nucleocytoplasmic transporter) and dual-specificity tyrosine phosphorylation-regulated kinase 1B (DYRK1B) were identified as direct targets of miR-9. Functionally, miR-9 promoted IL-2 production in stimulated human lymphocyte Jurkat T cells. Collectively, our data reveal a novel role for miR-9 in regulation of the NFAT pathway by targeting KPNB1 and DYRK1B.


Subject(s)
Lymphocyte Activation , MicroRNAs/metabolism , NFATC Transcription Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , T-Lymphocytes/enzymology , Transcriptional Activation , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , HEK293 Cells , HeLa Cells , Humans , Interleukin-2/metabolism , Ionomycin/pharmacology , Jurkat Cells , Lymphocyte Activation/drug effects , MicroRNAs/genetics , NFATC Transcription Factors/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Signal Transduction , T-Lymphocytes/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transcriptional Activation/drug effects , Transfection , beta Karyopherins/genetics , Dyrk Kinases
16.
Am J Respir Crit Care Med ; 190(9): 1032-43, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25180446

ABSTRACT

RATIONALE: Sphingosine kinases (SphKs) 1 and 2 regulate the synthesis of the bioactive sphingolipid sphingosine-1-phosphate (S1P), an important lipid mediator that promotes cell proliferation, migration, and angiogenesis. OBJECTIVES: We aimed to examine whether SphKs and their product, S1P, play a role in the development of pulmonary arterial hypertension (PAH). METHODS: SphK1(-/-), SphK2(-/-), and S1P lyase heterozygous (Sgpl1(+/-)) mice, a pharmacologic SphK inhibitor (SKI2), and a S1P receptor 2 (S1PR2) antagonist (JTE013) were used in rodent models of hypoxia-mediated pulmonary hypertension (HPH). S1P levels in lung tissues from patients with PAH and pulmonary arteries (PAs) from rodent models of HPH were measured. MEASUREMENTS AND MAIN RESULTS: mRNA and protein levels of SphK1, but not SphK2, were significantly increased in the lungs and isolated PA smooth muscle cells (PASMCs) from patients with PAH, and in lungs of experimental rodent models of HPH. S1P levels were increased in lungs of patients with PAH and PAs from rodent models of HPH. Unlike SphK2(-/-) mice, SphK1(-/-) mice were protected against HPH, whereas Sgpl1(+/-) mice were more susceptible to HPH. Pharmacologic SphK1 and S1PR2 inhibition prevented the development of HPH in rodent models of HPH. Overexpression of SphK1 and stimulation with S1P potentially via ligation of S1PR2 promoted PASMC proliferation in vitro, whereas SphK1 deficiency inhibited PASMC proliferation. CONCLUSIONS: The SphK1/S1P axis is a novel pathway in PAH that promotes PASMC proliferation, a major contributor to pulmonary vascular remodeling. Our results suggest that this pathway is a potential therapeutic target in PAH.


Subject(s)
Hypertension, Pulmonary/etiology , Lysophospholipids/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Sphingosine/analogs & derivatives , Animals , Humans , Male , Mice , Rats , Rats, Sprague-Dawley , Signal Transduction , Sphingosine/physiology , Tissue Culture Techniques
17.
J Biol Chem ; 288(35): 25414-25427, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23853098

ABSTRACT

Abnormal proliferation and phenotypic modulation of pulmonary artery smooth muscle cells (PASMC) contributes to the pathogenesis of numerous cardiovascular disorders, including pulmonary arterial hypertension (PAH). The nuclear factor of activated T cells (NFAT) signaling pathway is linked to PASMC proliferation and PAH. MicroRNAs (miRNAs) are small non-coding RNAs that function in diverse biological processes. To systemically identify the specific miRNAs that regulate the NFAT pathway, a human primary miRNA library was applied for cell-based high throughput screening with the NFAT luciferase reporter system. Eight miRNAs were found to modulate NFAT activity efficiently. Of them, miR-124 robustly inhibited NFAT reporter activity and decreased both the dephosphorylation and the nuclear translocation of NFAT. miR-124 also inhibited NFAT-dependent transcription of IL-2 in Jurkat T cells. miR-124 exerted its effects by targeting multiple genes, including a known component of the NFAT pathway, NFATc1, and two new regulators of NFAT signaling, CAMTA1 (calmodulin-binding transcription activator 1) and PTBP1 (polypyrimidine tract-binding protein 1). Physiologically, miR-124 was down-regulated by hypoxia in human PASMC, consistent with the activation of NFAT during this process. Down-regulation of miR-124 was also observed in 3-week hypoxia-treated mouse lungs. Furthermore, the overexpression of miR-124 not only inhibited human PASMC proliferation but also maintained its differentiated phenotype by repressing the NFAT pathway. Taken together, our data provide the first evidence that miR-124 acts as an inhibitor of the NFAT pathway. Down-regulation of miR-124 in hypoxia-treated PASMC and its antiproliferative and prodifferentiation effects imply a potential value for miR-124 in the treatment of PAH.


Subject(s)
Cell Proliferation , Hypertension, Pulmonary/metabolism , MicroRNAs/biosynthesis , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NFATC Transcription Factors/metabolism , Pulmonary Artery/metabolism , Transcriptional Activation , Animals , Cell Hypoxia/genetics , Down-Regulation/genetics , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Jurkat Cells , Mice , MicroRNAs/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NFATC Transcription Factors/genetics , Pulmonary Artery/pathology , Signal Transduction/genetics
18.
Am J Physiol Lung Cell Mol Physiol ; 307(7): L537-44, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25128522

ABSTRACT

Pulmonary hypertension (PH) is a chronic disease characterized by a progressive increase in vasomotor tone, narrowing of the vasculature with structural remodeling, and increase in pulmonary vascular resistance. Current treatment strategies include nitric oxide therapy and methods to increase cGMP-mediated vasodilatation. cGMP-dependent protein kinases (PKG) are known mediators of nitric oxide- and cGMP-induced vasodilatation. Deletion of PKG-1 in mice has been shown to induce PH, however, the exact mechanisms by which loss of PKG-1 function leads to PH is not known. In a mouse model with a selective mutation in the NH2-terminus leucine zipper protein interaction domain of PKG-1α [leucine zipper mutant (LZM)], we found a progressive increase in right ventricular systolic pressure and right heart hypertrophy compared with wild-type (WT) mice and increased RhoA-GTPase activity in the lungs. When exposed to chronic hypoxia, LZM mice developed modestly enhanced right ventricular remodeling compared with WT mice. Tadalafil, a phosphodiesterase-5 inhibitor that increases cGMP levels, significantly attenuated hypoxia-induced cardiopulmonary remodeling in WT mice but had no effect in LZM mice. We conclude that a functional leucine zipper domain in PKG-1α is essential for maintenance of a low pulmonary vascular tone in normoxia and for cGMP-mediated beneficial effects of phosphodiesterase-5 inhibition in hypoxic cardiopulmonary remodeling.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type I/genetics , Hypertension, Pulmonary/enzymology , Pulmonary Artery/physiopathology , Animals , Carbolines/pharmacology , Carbolines/therapeutic use , Cell Hypoxia , Cells, Cultured , Drug Evaluation, Preclinical , Female , Gene Expression , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/enzymology , Lung/blood supply , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Mutation , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Circulation/drug effects , Tadalafil , Vasodilation , Vasodilator Agents/pharmacology , Vasodilator Agents/therapeutic use , Ventricular Remodeling/drug effects , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
19.
J Cell Sci ; 125(Pt 24): 6117-26, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23038777

ABSTRACT

Soluble guanylyl cyclase (sGC) is the principal receptor for nitric oxide (NO) and crucial for the control of various physiological functions. The ß1 subunit of sGC is obligatory for the biological stability and activity of the sGC heterodimer. MicroRNAs (miRNAs) are important regulators of gene expression and exert great influences on diverse biological activities. The aim of the present study was to determine whether or not the expression of sGCß1 is specifically regulated by miRNAs. We report that miR-34c-5p directly targets sGCß1 under hypoxia. Bioinformatics analysis of the sGCß1 3'-untranslated region (3'-UTR) revealed a putative binding site for miR-34b-5p and miR-34c-5p, but only miR-34c-5p inhibited luciferase activity through interaction with sGCß1 3'-UTR in HEK293T cells. Site-directed mutagenesis of the putative miR-34c-5p binding site abolished the negative regulation of luciferase expression. Overexpression of miR-34c-5p repressed the expression of sGCß1 in stable cell lines, which was reversed by miR-34c-5p-specific sponge. Inoculation of mouse lung tissues in vitro with lentivirus bearing miR-34c-5p significantly decreased both the expression of sGCß1 and NO-stimulated sGC activity, which was also rescued by miR-34c-5p-specific sponge. Furthermore, we identified the putative Sp1-binding site in the promoter region of miR-34c-5p. Luciferase reporter constructs revealed that Sp1 directly binds to the wild-type promoter of miR-34c-5p, which was confirmed by chromatin immunoprecipitation. In summary, these findings reveal that miR-34c-5p directly regulates sGCß1 expression, and they identify the key transcription factor Sp1 that governs miR-34c-5p expression during hypoxia.


Subject(s)
Cell Hypoxia/physiology , Guanylate Cyclase/metabolism , MicroRNAs/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Down-Regulation , Female , Guanylate Cyclase/genetics , HEK293 Cells , Humans , Mice , MicroRNAs/genetics , Muscle, Smooth, Vascular/cytology , Protein Subunits , Random Allocation , Receptors, Cytoplasmic and Nuclear/genetics , Soluble Guanylyl Cyclase
20.
Am J Pathol ; 183(4): 1169-1182, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23933064

ABSTRACT

Bronchopulmonary dysplasia of the premature newborn is characterized by lung injury, resulting in alveolar simplification and reduced pulmonary function. Exposure of neonatal mice to hyperoxia enhanced sphingosine-1-phosphate (S1P) levels in lung tissues; however, the role of increased S1P in the pathobiological characteristics of bronchopulmonary dysplasia has not been investigated. We hypothesized that an altered S1P signaling axis, in part, is responsible for neonatal lung injury leading to bronchopulmonary dysplasia. To validate this hypothesis, newborn wild-type, sphingosine kinase1(-/-) (Sphk1(-/-)), sphingosine kinase 2(-/-) (Sphk2(-/-)), and S1P lyase(+/-) (Sgpl1(+/-)) mice were exposed to hyperoxia (75%) from postnatal day 1 to 7. Sphk1(-/-), but not Sphk2(-/-) or Sgpl1(+/-), mice offered protection against hyperoxia-induced lung injury, with improved alveolarization and alveolar integrity compared with wild type. Furthermore, SphK1 deficiency attenuated hyperoxia-induced accumulation of IL-6 in bronchoalveolar lavage fluids and NADPH oxidase (NOX) 2 and NOX4 protein expression in lung tissue. In vitro experiments using human lung microvascular endothelial cells showed that exogenous S1P stimulated intracellular reactive oxygen species (ROS) generation, whereas SphK1 siRNA, or inhibitor against SphK1, attenuated hyperoxia-induced S1P generation. Knockdown of NOX2 and NOX4, using specific siRNA, reduced both basal and S1P-induced ROS formation. These results suggest an important role for SphK1-mediated S1P signaling-regulated ROS in the development of hyperoxia-induced lung injury in a murine neonatal model of bronchopulmonary dysplasia.


Subject(s)
Bronchopulmonary Dysplasia/enzymology , Bronchopulmonary Dysplasia/prevention & control , Hyperoxia/complications , Lysophospholipids/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Sphingosine/analogs & derivatives , Aldehyde-Lyases/deficiency , Aldehyde-Lyases/metabolism , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Humans , Hyperoxia/enzymology , Hyperoxia/pathology , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidase 4 , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pneumonia/complications , Pneumonia/pathology , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/pathology , Reactive Oxygen Species/metabolism , Signal Transduction , Sphingosine/metabolism , rac1 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL