Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34
1.
Telemed J E Health ; 30(2): 354-363, 2024 Feb.
Article En | MEDLINE | ID: mdl-37585548

Introduction: Electromyography (EMG) assessments have been conducted virtually more frequently in recent years, leading researchers to explore the barriers to EMG assessments in a telehealth setting and how to overcome them. Methods: A scoping review was conducted according to the methodology described by Arksey and O'Malley. A comprehensive search using controlled vocabulary and keywords for two concepts, EMG and telehealth, was conducted using Medline and EMBASE on February 7, 2022. Two independent reviewers screened titles, abstracts, and full-text articles. Two reviewers also extracted the data and described the findings in a descriptive analysis. Results: A total of 248 articles were screened during the abstract and title review, of which 64 full texts were screened for eligibility. Of these, 15 publications met the inclusion criteria. Most articles were published in 2018 or later (66.7%). The most frequently mentioned barrier to conducting a virtual EMG assessment was poor data and signal transmission (53.3%). Another frequently mentioned barrier was poor patient usability (33.3%). Solutions most frequently reported related to patient usability (33.3%). These included interactive instructions and video chat to monitor and provide the patient with technical support. Conclusion: The last 4 years have seen an increase in articles published on EMGs' use in telehealth to monitor or diagnose patients. Further research is required to determine if the proposed solutions have improved clinical outcomes for the patient.


Telemedicine , Humans , Electromyography , Telemedicine/methods
2.
JACC Basic Transl Sci ; 7(4): 384-403, 2022 Apr.
Article En | MEDLINE | ID: mdl-35540097

The authors show that increased poly(adenosine diphosphate-ribose) polymerase 1 (PARP1) and pyruvate kinase muscle isozyme 2 (PKM2) expression is a common feature of a decompensated right ventricle in patients with pulmonary arterial hypertension and animal models. The authors find in vitro that overactivated PARP1 promotes cardiomyocyte dysfunction by favoring PKM2 expression and nuclear function, glycolytic gene expression, activation of nuclear factor κB-dependent proinflammatory factors. Pharmacologic and genetic inhibition of PARP1 or enforced tetramerization of PKM2 attenuates maladaptive remodeling improving right ventricular (RV) function in multiple rodent models. Taken together, these data implicate the PARP1/PKM2 axis as a critical driver of maladaptive RV remodeling and a new promising target to directly sustain RV function in patients with pulmonary arterial hypertension.

3.
Thorax ; 77(3): 247-258, 2022 03.
Article En | MEDLINE | ID: mdl-34226205

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease characterised by exuberant tissue remodelling and associated with high unmet medical needs. Outcomes are even worse when IPF results in secondary pulmonary hypertension (PH). Importantly, exaggerated resistance to cell death, excessive proliferation and enhanced synthetic capacity are key endophenotypes of both fibroblasts and pulmonary artery smooth muscle cells, suggesting shared molecular pathways. Under persistent injury, sustained activation of the DNA damage response (DDR) is integral to the preservation of cells survival and their capacity to proliferate. Checkpoint kinases 1 and 2 (CHK1/2) are key components of the DDR. The objective of this study was to assess the role of CHK1/2 in the development and progression of IPF and IPF+PH. METHODS AND RESULTS: Increased expression of DNA damage markers and CHK1/2 were observed in lungs, remodelled pulmonary arteries and isolated fibroblasts from IPF patients and animal models. Blockade of CHK1/2 expression or activity-induced DNA damage overload and reverted the apoptosis-resistant and fibroproliferative phenotype of disease cells. Moreover, inhibition of CHK1/2 was sufficient to interfere with transforming growth factor beta 1-mediated fibroblast activation. Importantly, pharmacological inhibition of CHK1/2 using LY2606368 attenuated fibrosis and pulmonary vascular remodelling leading to improvement in respiratory mechanics and haemodynamic parameters in two animal models mimicking IPF and IPF+PH. CONCLUSION: This study identifies CHK1/2 as key regulators of lung fibrosis and provides a proof of principle for CHK1/2 inhibition as a potential novel therapeutic option for IPF and IPF+PH.


Hypertension, Pulmonary , Idiopathic Pulmonary Fibrosis , Animals , Fibroblasts/metabolism , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Myocytes, Smooth Muscle/metabolism
4.
Int J Mol Sci ; 22(6)2021 Mar 13.
Article En | MEDLINE | ID: mdl-33805714

Trifluoperazine (TFP), an antipsychotic drug approved by the Food and Drug Administration, has been show to exhibit anti-cancer effects. Pulmonary arterial hypertension (PAH) is a devastating disease characterized by a progressive obliteration of small pulmonary arteries (PAs) due to exaggerated proliferation and resistance to apoptosis of PA smooth muscle cells (PASMCs). However, the therapeutic potential of TFP for correcting the cancer-like phenotype of PAH-PASMCs and improving PAH in animal models remains unknown. PASMCs isolated from PAH patients were exposed to different concentrations of TFP before assessments of cell proliferation and apoptosis. The in vivo therapeutic potential of TFP was tested in two preclinical models with established PAH, namely the monocrotaline and sugen/hypoxia-induced rat models. Assessments of hemodynamics by right heart catheterization and histopathology were conducted. TFP showed strong anti-survival and anti-proliferative effects on cultured PAH-PASMCs. Exposure to TFP was associated with downregulation of AKT activity and nuclear translocation of forkhead box protein O3 (FOXO3). In both preclinical models, TFP significantly lowered the right ventricular systolic pressure and total pulmonary resistance and improved cardiac function. Consistently, TFP reduced the medial wall thickness of distal PAs. Overall, our data indicate that TFP could have beneficial effects in PAH and support the view that seeking new uses for old drugs may represent a fruitful approach.


Cardiovascular Agents/pharmacology , Gene Expression Regulation/drug effects , Hypertension, Pulmonary/drug therapy , Hypoxia/prevention & control , Myocytes, Smooth Muscle/drug effects , Trifluoperazine/pharmacology , Animals , Antipsychotic Agents/pharmacology , Cell Proliferation/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Repositioning , Female , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Hemodynamics/drug effects , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Hypoxia/chemically induced , Hypoxia/genetics , Hypoxia/physiopathology , Indoles/administration & dosage , Monocrotaline/administration & dosage , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pyrroles/administration & dosage , Rats , Rats, Sprague-Dawley , Survivin/genetics , Survivin/metabolism
5.
Int J Mol Sci ; 22(6)2021 Mar 15.
Article En | MEDLINE | ID: mdl-33803922

Pulmonary arterial hypertension (PAH) is a progressive disorder characterized by a sustained elevation of pulmonary artery (PA) pressure, right ventricular failure, and premature death. Enhanced proliferation and resistance to apoptosis (as seen in cancer cells) of PA smooth muscle cells (PASMCs) is a major pathological hallmark contributing to pulmonary vascular remodeling in PAH, for which current therapies have only limited effects. Emerging evidence points toward a critical role for Enhancer of Zeste Homolog 2 (EZH2) in cancer cell proliferation and survival. However, its role in PAH remains largely unknown. The aim of this study was to determine whether EZH2 represents a new factor critically involved in the abnormal phenotype of PAH-PASMCs. We found that EZH2 is overexpressed in human lung tissues and isolated PASMCs from PAH patients compared to controls as well as in two animal models mimicking the disease. Through loss- and gain-of-function approaches, we showed that EZH2 promotes PAH-PASMC proliferation and survival. By combining quantitative transcriptomic and proteomic approaches in PAH-PASMCs subjected or not to EZH2 knockdown, we found that inhibition of EZH2 downregulates many factors involved in cell-cycle progression, including E2F targets, and contributes to maintain energy production. Notably, we found that EZH2 promotes expression of several nuclear-encoded components of the mitochondrial translation machinery and tricarboxylic acid cycle genes. Overall, this study provides evidence that, by overexpressing EZH2, PAH-PASMCs remove the physiological breaks that normally restrain their proliferation and susceptibility to apoptosis and suggests that EZH2 or downstream factors may serve as therapeutic targets to combat pulmonary vascular remodeling.


Enhancer of Zeste Homolog 2 Protein/genetics , Proteome/genetics , Pulmonary Arterial Hypertension/genetics , Transcriptome/genetics , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Citric Acid Cycle/genetics , Epigenesis, Genetic/genetics , Female , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Lung/metabolism , Lung/pathology , Male , Middle Aged , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/growth & development , Pulmonary Artery/pathology , Rats
6.
J Clin Invest ; 131(6)2021 03 15.
Article En | MEDLINE | ID: mdl-33497359

Women with pulmonary arterial hypertension (PAH) exhibit better right ventricular (RV) function and survival than men; however, the underlying mechanisms are unknown. We hypothesized that 17ß-estradiol (E2), through estrogen receptor α (ER-α), attenuates PAH-induced RV failure (RVF) by upregulating the procontractile and prosurvival peptide apelin via a BMPR2-dependent mechanism. We found that ER-α and apelin expression were decreased in RV homogenates from patients with RVF and from rats with maladaptive (but not adaptive) RV remodeling. RV cardiomyocyte apelin abundance increased in vivo or in vitro after treatment with E2 or ER-α agonist. Studies employing ER-α-null or ER-ß-null mice, ER-α loss-of-function mutant rats, or siRNA demonstrated that ER-α is necessary for E2 to upregulate RV apelin. E2 and ER-α increased BMPR2 in pulmonary hypertension RVs and in isolated RV cardiomyocytes, associated with ER-α binding to the Bmpr2 promoter. BMPR2 is required for E2-mediated increases in apelin abundance, and both BMPR2 and apelin are necessary for E2 to exert RV-protective effects. E2 or ER-α agonist rescued monocrotaline pulmonary hypertension and restored RV apelin and BMPR2. We identified what we believe to be a novel cardioprotective E2/ER-α/BMPR2/apelin axis in the RV. Harnessing this axis may lead to novel RV-targeted therapies for PAH patients of either sex.


Apelin/metabolism , Bone Morphogenetic Protein Receptors, Type II/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/metabolism , Hypertension, Pulmonary/physiopathology , Ventricular Function, Right/physiology , Animals , Cardiotonic Agents/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Estrogen Receptor alpha/deficiency , Estrogen Receptor alpha/genetics , Female , Humans , Male , Mice , Mice, Knockout , Models, Cardiovascular , Myocytes, Cardiac/metabolism , Rats , Rats, Mutant Strains
7.
Am J Respir Crit Care Med ; 203(5): 614-627, 2021 03 01.
Article En | MEDLINE | ID: mdl-33021405

Rationale: Pulmonary arterial hypertension (PAH) is a life-threatening condition characterized by abnormally elevated pulmonary pressures and right ventricular failure. Excessive proliferation and resistance to apoptosis of pulmonary artery smooth muscle cells (PASMCs) is one of the most important drivers of vascular remodeling in PAH, for which available treatments have limited effectiveness.Objectives: To gain insights into the mechanisms leading to the development of the disease and identify new actionable targets.Methods: Protein expression profiling was conducted by two-dimensional liquid chromatography coupled to tandem mass spectrometry in isolated PASMCs from controls and patients with PAH. Multiple molecular, biochemical, and pharmacologic approaches were used to decipher the role of NUDT1 (nudrix hyrolase 1) in PAH.Measurements and Main Results: Increased expression of the detoxifying DNA enzyme NUDT1 was detected in cells and tissues from patients with PAH and animal models. In vitro, molecular or pharmacological inhibition of NUDT1 in PAH-PASMCs induced accumulation of oxidized nucleotides in the DNA, irresolvable DNA damage (comet assay), disruption of cellular bioenergetics (Seahorse), and cell death (terminal deoxynucleotidyl transferase dUTP nick end labeling assay). In two animal models with established PAH (i.e., monocrotaline and Sugen/hypoxia-treated rats), pharmacological inhibition of NUDT1 using (S)-Crizotinib significantly decreased pulmonary vascular remodeling and improved hemodynamics and cardiac function.Conclusions: Our results indicate that, by overexpressing NUDT1, PAH-PASMCs hijack persistent oxidative stress in preventing incorporation of oxidized nucleotides into DNA, thus allowing the cell to escape apoptosis and proliferate. Given that NUDT1 inhibitors are under clinical investigation for cancer, they may represent a new therapeutic option for PAH.


DNA Repair Enzymes/genetics , DNA/metabolism , Oxidative Stress/genetics , Phosphoric Monoester Hydrolases/genetics , Pulmonary Arterial Hypertension/genetics , Pulmonary Artery/metabolism , Vascular Remodeling/genetics , 8-Hydroxy-2'-Deoxyguanosine/metabolism , Adult , Aged , Animals , Apoptosis/genetics , Blotting, Western , Case-Control Studies , Cell Proliferation/genetics , Chromatography, Liquid , Comet Assay , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/metabolism , Disease Models, Animal , Female , Forkhead Box Protein M1/metabolism , Humans , In Vitro Techniques , Male , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Oxidation-Reduction , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Pulmonary Arterial Hypertension/metabolism , Pyrophosphatases/antagonists & inhibitors , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , RNA, Messenger/metabolism , Rats , Tandem Mass Spectrometry , Up-Regulation
8.
Circulation ; 142(15): 1464-1484, 2020 10 13.
Article En | MEDLINE | ID: mdl-32698630

BACKGROUND: Right ventricular (RV) function is the major determinant for both functional capacity and survival in patients with pulmonary arterial hypertension (PAH). Despite the recognized clinical importance of preserving RV function, the subcellular mechanisms that govern the transition from a compensated to a decompensated state remain poorly understood and as a consequence there are no clinically established treatments for RV failure and a paucity of clinically useful biomarkers. Accumulating evidence indicates that long noncoding RNAs are powerful regulators of cardiac development and disease. Nonetheless, their implication in adverse RV remodeling in PAH is unknown. METHODS: Expression of the long noncoding RNA H19 was assessed by quantitative PCR in plasma and RV from patients categorized as control RV, compensated RV or decompensated RV based on clinical history and cardiac index. The impact of H19 suppression using GapmeR was explored in 2 rat models mimicking RV failure, namely the monocrotaline and pulmonary artery banding. Echocardiographic, hemodynamic, histological, and biochemical analyses were conducted. In vitro gain- and loss-of-function experiments were performed in rat cardiomyocytes. RESULTS: We demonstrated that H19 is upregulated in decompensated RV from PAH patients and correlates with RV hypertrophy and fibrosis. Similar findings were observed in monocrotaline and pulmonary artery banding rats. We found that silencing H19 limits pathological RV hypertrophy, fibrosis and capillary rarefaction, thus preserving RV function in monocrotaline and pulmonary artery banding rats without affecting pulmonary vascular remodeling. This cardioprotective effect was accompanied by E2F transcription factor 1-mediated upregulation of enhancer of zeste homolog 2. In vitro, knockdown of H19 suppressed cardiomyocyte hypertrophy induced by phenylephrine, while its overexpression has the opposite effect. Finally, we demonstrated that circulating H19 levels in plasma discriminate PAH patients from controls, correlate with RV function and predict long-term survival in 2 independent idiopathic PAH cohorts. Moreover, H19 levels delineate subgroups of patients with differentiated prognosis when combined with the NT-proBNP (N-terminal pro-B-type natriuretic peptide) levels or the risk score proposed by both REVEAL (Registry to Evaluate Early and Long-Term PAH Disease Management) and the 2015 European Pulmonary Hypertension Guidelines. CONCLUSIONS: Our findings identify H19 as a new therapeutic target to impede the development of maladaptive RV remodeling and a promising biomarker of PAH severity and prognosis.


Heart Failure/metabolism , Pulmonary Arterial Hypertension/metabolism , RNA, Long Noncoding/metabolism , Vascular Remodeling , Ventricular Dysfunction, Right/metabolism , Animals , Biomarkers/metabolism , Heart Failure/mortality , Heart Failure/pathology , Humans , Natriuretic Peptide, Brain/metabolism , Peptide Fragments/metabolism , Pulmonary Arterial Hypertension/mortality , Pulmonary Arterial Hypertension/pathology , Rats , Ventricular Dysfunction, Right/mortality , Ventricular Dysfunction, Right/pathology
9.
PLoS One ; 15(6): e0234872, 2020.
Article En | MEDLINE | ID: mdl-32559203

The leading cause of death in Pulmonary Arterial Hypertension (PAH) is right ventricular (RV) failure. The tumor suppressor p53 has been associated with left ventricular hypertrophy (LVH) and remodeling but its role in RV hypertrophy (RVH) is unclear. The purpose of this study was to determine whether pharmacological activation of p53 by Quinacrine affects RV remodeling and function in the pulmonary artery banding (PAB) model of compensated RVH in mice. The effects of p53 activation on cellular functions were studied in isolated cardiomyocytes, cardiac fibroblasts and endothelial cells (ECs). The expression of p53 was examined both on human RV tissues from patients with compensated and decompensated RVH and in mouse RV tissues early and late after the PAB. As compared to control human RVs, there was no change in p53 expression in compensated RVH, while a marked upregulation was found in decompensated RVH. Similarly, in comparison to SHAM-operated mice, unaltered RV p53 expression 7 days after PAB, was markedly induced 21 days after the PAB. Quinacrine induced p53 accumulation did not further deteriorate RV function at day 7 after PAB. Quinacrine administration did not increase EC death, neither diminished EC number and capillary density in RV tissues. No major impact on the expression of markers of sarcomere organization, fatty acid and mitochondrial metabolism and respiration was noted in Quinacrine-treated PAB mice. p53 accumulation modulated the expression of Heme Oxygenase 1 (HO-1) and Glucose Transporter (Glut1) in mouse RVs and in adult cardiomyocytes. We conclude that early p53 activation in PAB-induced RVH does not cause substantial detrimental effects on right ventricular remodeling and function.


Hypertrophy, Right Ventricular/metabolism , Tumor Suppressor Protein p53/genetics , Adult , Aged , Animals , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fatty Acids/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Heme Oxygenase-1/metabolism , Humans , Hypertrophy, Right Ventricular/genetics , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mitochondria, Heart/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Quinacrine/pharmacology , Sarcomeres/metabolism , Tumor Suppressor Protein p53/metabolism
10.
Arterioscler Thromb Vasc Biol ; 40(3): 783-801, 2020 03.
Article En | MEDLINE | ID: mdl-31969012

OBJECTIVE: Pulmonary arterial hypertension (PAH) is a fatal disease characterized by the narrowing of pulmonary arteries (PAs). It is now established that this phenotype is associated with enhanced PA smooth muscle cells (PASMCs) proliferation and suppressed apoptosis. This phenotype is sustained in part by the activation of several DNA repair pathways allowing PASMCs to survive despite the unfavorable environmental conditions. PIM1 (Moloney murine leukemia provirus integration site) is an oncoprotein upregulated in PAH and involved in many prosurvival pathways, including DNA repair. The objective of this study was to demonstrate the implication of PIM1 in the DNA damage response and the beneficial effect of its inhibition by pharmacological inhibitors in human PAH-PASMCs and in rat PAH models. Approach and Results: We found in vitro that PIM1 inhibition by either SGI-1776, TP-3654, siRNA (silencer RNA) decreased the phosphorylation of its newly identified direct target KU70 (lupus Ku autoantigen protein p70) resulting in the inhibition of double-strand break repair (Comet Assay) by the nonhomologous end-joining as well as reduction of PAH-PASMCs proliferation (Ki67-positive cells) and resistance to apoptosis (Annexin V positive cells) of PAH-PASMCs. In vivo, SGI-1776 and TP-3654 given 3× a week, improved significantly pulmonary hemodynamics (right heart catheterization) and vascular remodeling (Elastica van Gieson) in monocrotaline and Fawn-Hooded rat models of PAH. CONCLUSIONS: We demonstrated that PIM1 phosphorylates KU70 and initiates DNA repair signaling in PAH-PASMCs and that PIM1 inhibitors represent a therapeutic option for patients with PAH.


DNA Damage , DNA End-Joining Repair , Hypertension, Pulmonary/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Antihypertensive Agents/pharmacology , Apoptosis , Cell Proliferation , Cells, Cultured , DNA End-Joining Repair/drug effects , Disease Models, Animal , Female , Histones/metabolism , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Ku Autoantigen/metabolism , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Phosphoproteins/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/genetics , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Rats, Sprague-Dawley , Vascular Remodeling
12.
Circ Res ; 125(4): 449-466, 2019 08 02.
Article En | MEDLINE | ID: mdl-31154939

RATIONALE: Pulmonary hypertension (PH) due to left heart disease (LHD), or group 2 PH, is the most prevalent form of PH worldwide. PH due to LHD is often associated with metabolic syndrome (MetS). In 12% to 13% of cases, patients with PH due to LHD display vascular remodeling of pulmonary arteries (PAs) associated with poor prognosis. Unfortunately, the underlying mechanisms remain unknown; PH-targeted therapies for this group are nonexistent, and the development of a new preclinical model is crucial. Among the numerous pathways dysregulated in MetS, inflammation plays also a critical role in both PH and vascular remodeling. OBJECTIVE: We hypothesized that MetS and inflammation may trigger the development of vascular remodeling in group 2 PH. METHODS AND RESULTS: Using supracoronary aortic banding, we induced diastolic dysfunction in rats. Then we induced MetS by a combination of high-fat diet and olanzapine treatment. We used metformin treatment and anti-IL-6 (interleukin-6) antibodies to inhibit the IL-6 pathway. Compared with sham conditions, only supracoronary aortic banding+MetS rats developed precapillary PH, as measured by both echocardiography and right/left heart catheterization. PH in supracoronary aortic banding+MetS was associated with macrophage accumulation and increased IL-6 production in lung. PH was also associated with STAT3 (signal transducer and activator of transcription 3) activation and increased proliferation of PA smooth muscle cells, which contributes to remodeling of distal PA. We reported macrophage accumulation, increased IL-6 levels, and STAT3 activation in the lung of group 2 PH patients. In vitro, IL-6 activates STAT3 and induces human PA smooth muscle cell proliferation. Metformin treatment decreased inflammation, IL-6 levels, STAT3 activation, and human PA smooth muscle cell proliferation. In vivo, in the supracoronary aortic banding+MetS animals, reducing IL-6, either by anti-IL-6 antibody or metformin treatment, reversed pulmonary vascular remodeling and improve PH due to LHD. CONCLUSIONS: We developed a new preclinical model of group 2 PH by combining MetS with LHD. We showed that MetS exacerbates group 2 PH. We provided evidence for the importance of the IL-6-STAT3 pathway in our experimental model of group 2 PH and human patients.


Disease Models, Animal , Hypertension, Pulmonary/pathology , Metabolic Syndrome/complications , Ventricular Dysfunction/complications , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/metabolism , Male , Metabolic Syndrome/etiology , Olanzapine/toxicity , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Rats , Rats, Wistar , Vascular Remodeling
13.
Am J Respir Crit Care Med ; 198(1): 90-103, 2018 07 01.
Article En | MEDLINE | ID: mdl-29394093

RATIONALE: Pulmonary arterial hypertension (PAH) is a vascular remodeling disease with a poor prognosis and limited therapeutic options. Although the mechanisms contributing to vascular remodeling in PAH are still unclear, several features, including hyperproliferation and resistance to apoptosis of pulmonary artery smooth muscle cells (PASMCs), have led to the emergence of the cancer-like concept. The molecular chaperone HSP90 (heat shock protein 90) is directly associated with malignant growth and proliferation under stress conditions. In addition to being highly expressed in the cytosol, HSP90 exists in a subcellular pool compartmentalized in the mitochondria (mtHSP90) of tumor cells, but not in normal cells, where it promotes cell survival. OBJECTIVES: We hypothesized that mtHSP90 in PAH-PASMCs represents a protective mechanism against stress, promoting their proliferation and resistance to apoptosis. METHODS: Expression and localization of HSP90 were analyzed by Western blot, immunofluorescence, and immunogold electron microscopy. In vitro, effects of mtHSP90 inhibition on mitochondrial DNA integrity, bioenergetics, cell proliferation and resistance to apoptosis were assessed. In vivo, the therapeutic potential of Gamitrinib, a mitochondria-targeted HSP90 inhibitor, was tested in fawn-hooded and monocrotaline rats. MEASUREMENTS AND MAIN RESULTS: We demonstrated that, in response to stress, HSP90 preferentially accumulates in PAH-PASMC mitochondria (dual immunostaining, immunoblot, and immunogold electron microscopy) to ensure cell survival by preserving mitochondrial DNA integrity and bioenergetic functions. Whereas cytosolic HSP90 inhibition displays a lack of absolute specificity for PAH-PASMCs, Gamitrinib decreased mitochondrial DNA content and repair capacity and bioenergetic functions, thus repressing PAH-PASMC proliferation (Ki67 labeling) and resistance to apoptosis (Annexin V assay) without affecting control cells. In vivo, Gamitrinib improves PAH in two experimental rat models (monocrotaline and fawn-hooded rat). CONCLUSIONS: Our data show for the first time that accumulation of mtHSP90 is a feature of PAH-PASMCs and a key regulator of mitochondrial homeostasis contributing to vascular remodeling in PAH.


Antihypertensive Agents/therapeutic use , HSP90 Heat-Shock Proteins/analysis , HSP90 Heat-Shock Proteins/metabolism , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Mitochondria/metabolism , Vascular Remodeling/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured/drug effects , Disease Models, Animal , Humans , Muscle, Smooth, Vascular/drug effects , Rats
14.
J Mol Med (Berl) ; 96(2): 223-235, 2018 02.
Article En | MEDLINE | ID: mdl-29290032

Pulmonary arterial hypertension (PAH) is a progressive vascular remodeling disease characterized by a persistent elevation of pulmonary artery pressure, leading to right heart failure and premature death. Exaggerated proliferation and resistance to apoptosis of pulmonary artery smooth muscle cells (PASMCs) is a key component of vascular remodeling. Despite major advances in the field, current therapies for PAH remain poorly effective in reversing the disease or significantly improving long-term survival. Because the transcription factor FOXM1 is necessary for PASMC proliferation during lung morphogenesis and its overexpression stimulates proliferation and evasion of apoptosis in cancer cells, we thus hypothesized that upregulation of FOXM1 in PAH-PASMCs promotes cell expansion and vascular remodeling. Our results showed that FOXM1 was markedly increased in distal pulmonary arteries and isolated PASMCs from PAH patients compared to controls as well as in two preclinical models. In vitro, we showed that miR-204 expression regulates FOXM1 levels and that inhibition of FOXM1 reduced cell proliferation and resistance to apoptosis through diminished DNA repair mechanisms and decreased expression of the pro-remodeling factor survivin. Accordingly, inhibition of FOXM1 with thiostrepton significantly improved established PAH in two rat models. Thus, we show for the first time that FOXM1 is implicated in PAH development and represents a new promising target. KEY MESSAGES: FOXM1 is overexpressed in human PAH-PASMCs and PAH animal models. FOXM1 promotes PAH-PASMC proliferation and resistance to apoptosis. Pharmacological inhibition of FOXM1 improves established PAH in the MCT and Su/Hx rat models. FOXM1 may be a novel therapeutic target in PAH.


Forkhead Box Protein M1/physiology , Hypertension, Pulmonary/metabolism , Myocytes, Smooth Muscle/physiology , Pulmonary Artery/physiology , Animals , Cell Line , Cell Proliferation , Forkhead Box Protein M1/antagonists & inhibitors , Humans , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/metabolism , Male , MicroRNAs/metabolism , Pulmonary Artery/cytology , Rats, Sprague-Dawley , Thiostrepton/therapeutic use , Vascular Remodeling
15.
Pulm Circ ; 8(1): 2045893217741429, 2018.
Article En | MEDLINE | ID: mdl-29064353

Dysregulated metabolism and rarefaction of the capillary network play a critical role in pulmonary arterial hypertension (PAH) etiology. They are associated with a decrease in perfusion of the lungs, skeletal muscles, and right ventricle (RV). Previous studies suggested that endothelin-1 (ET-1) modulates both metabolism and angiogenesis. We hypothesized that dual ETA/ETB receptors blockade improves PAH by improving cell metabolism and promoting angiogenesis. Five weeks after disease induction, Sugen/hypoxic rats presented severe PAH with pulmonary artery (PA) remodeling, RV hypertrophy and capillary rarefaction in the lungs, RV, and skeletal muscles (microCT angiogram, lectin perfusion, CD31 staining). Two-week treatment with dual ETA/ETB receptors antagonist macitentan (30 mg/kg/d) significantly improved pulmonary hemodynamics, PA vascular remodeling, and RV function and hypertrophy compared to vehicle-treated animals (all P = 0.05). Moreover, macitentan markedly increased lung, RV and quadriceps perfusion, and microvascular density (all P = 0.05). In vitro, these effects were associated with increases in oxidative phosphorylation (oxPhox) and markedly reduced cell proliferation of PAH-PA smooth muscle cells (PASMCs) treated with macitentan without affecting apoptosis. While macitentan did not affect oxPhox, proliferation, and apoptosis of PAH-PA endothelial cells (PAECs), it significantly improved their angiogenic capacity (tube formation assay). Exposure of control PASMC and PAEC to ET-1 fully mimicked the PAH cells phenotype, thus confirming that ET-1 is implicated in both metabolism and angiogenesis abnormalities in PAH. Dual ETA/ETB receptor blockade improved the metabolic changes involved in PAH-PASMCs' proliferation and the angiogenic capacity of PAH-PAEC leading to an increased capillary density in lungs, RV, and skeletal muscles.

16.
J Am Heart Assoc ; 6(10)2017 Oct 12.
Article En | MEDLINE | ID: mdl-29025748

BACKGROUND: Functional cerebrovascular regulatory mechanisms are important for maintaining constant cerebral blood flow and oxygen supply in heathy individuals and are altered in heart failure. We aim to examine whether pulmonary arterial hypertension (PAH) is associated with abnormal cerebrovascular regulation and lower cerebral oxygenation and their physiological and clinical consequences. METHODS AND RESULTS: Resting mean flow velocity in the middle cerebral artery mean flow velocity in the middle cerebral artery (MCAvmean); transcranial Doppler), cerebral pressure-flow relationship (assessed at rest and during squat-stand maneuvers; analyzed using transfer function analysis), cerebrovascular reactivity to CO2, and central chemoreflex were assessed in 11 patients with PAH and 11 matched healthy controls. Both groups also completed an incremental ramp exercise protocol until exhaustion, during which MCAvmean, mean arterial pressure, cardiac output (photoplethysmography), end-tidal partial pressure of CO2, and cerebral oxygenation (near-infrared spectroscopy) were measured. Patients were characterized by a significant decrease in resting MCAvmean (P<0.01) and higher transfer function gain at rest and during squat-stand maneuvers (both P<0.05). Cerebrovascular reactivity to CO2 was reduced (P=0.03), whereas central chemoreceptor sensitivity was increased in PAH (P<0.01), the latter correlating with increased resting ventilation (R2=0.47; P<0.05) and the exercise ventilation/CO2 production slope (V˙E/V˙CO2 slope; R2=0.62; P<0.05) during exercise for patients. Exercise-induced increases in MCAvmean were limited in PAH (P<0.05). Reduced MCAvmean contributed to impaired cerebral oxygen delivery and oxygenation (both P<0.05), the latter correlating with exercise capacity in patients with PAH (R2=0.52; P=0.01). CONCLUSIONS: These findings provide comprehensive evidence for physiologically and clinically relevant impairments in cerebral hemodynamic regulation and oxygenation in PAH.


Cerebrovascular Circulation , Exercise Tolerance , Hemodynamics , Hypertension, Pulmonary/physiopathology , Middle Cerebral Artery/physiopathology , Oxygen Consumption , Oxygen/blood , Adult , Biomarkers/blood , Blood Flow Velocity , Case-Control Studies , Chemoreceptor Cells/metabolism , Female , Homeostasis , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/diagnostic imaging , Male , Middle Aged , Middle Cerebral Artery/diagnostic imaging , Models, Cardiovascular , Photoplethysmography , Reflex , Ultrasonography, Doppler, Transcranial
17.
Sci Rep ; 7(1): 4546, 2017 07 03.
Article En | MEDLINE | ID: mdl-28674407

Pulmonary arterial hypertension (PAH) is a vascular remodeling disease with limited therapeutic options. Although exposed to stressful conditions, pulmonary artery (PA) smooth muscle cells (PASMCs) exhibit a "cancer-like" pro-proliferative and anti-apoptotic phenotype. HDAC6 is a cytoplasmic histone deacetylase regulating multiple pro-survival mechanisms and overexpressed in response to stress in cancer cells. Due to the similarities between cancer and PAH, we hypothesized that HDAC6 expression is increased in PAH-PASMCs to face stress allowing them to survive and proliferate, thus contributing to vascular remodeling in PAH. We found that HDAC6 is significantly up-regulated in lungs, distal PAs, and isolated PASMCs from PAH patients and animal models. Inhibition of HDAC6 reduced PAH-PASMC proliferation and resistance to apoptosis in vitro sparing control cells. Mechanistically, we demonstrated that HDAC6 maintains Ku70 in a hypoacetylated state, blocking the translocation of Bax to mitochondria and preventing apoptosis. In vivo, pharmacological inhibition of HDAC6 improved established PAH in two experimental models and can be safely given in combination with currently approved PAH therapies. Moreover, Hdac6 deficient mice were partially protected against chronic hypoxia-induced pulmonary hypertension. Our study shows for the first time that HDAC6 is implicated in PAH development and represents a new promising target to improve PAH.


Histone Deacetylase 6/genetics , Histone Deacetylase 6/metabolism , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Acetylation , Animals , Apoptosis/genetics , Cell Line , Cell Movement/genetics , Cell Proliferation/genetics , Disease Models, Animal , Gene Expression , Gene Expression Regulation , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Immunohistochemistry , Ku Autoantigen/metabolism , Mice , Mice, Knockout , Models, Biological , Myocytes, Smooth Muscle/metabolism , Rats , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
18.
Arterioscler Thromb Vasc Biol ; 37(8): 1513-1523, 2017 08.
Article En | MEDLINE | ID: mdl-28473439

OBJECTIVE: Pulmonary arterial hypertension (PAH) is a vascular disease not restricted to the lungs. Many signaling pathways described in PAH are also of importance in other vascular remodeling diseases, such as coronary artery disease (CAD). Intriguingly, CAD is 4× more prevalent in PAH compared with the global population, suggesting a link between these 2 diseases. Both PAH and CAD are associated with sustained inflammation and smooth muscle cell proliferation/apoptosis imbalance and we demonstrated in PAH that this phenotype is, in part, because of the miR-223/DNA damage/Poly[ADP-ribose] polymerase 1/miR-204 axis activation and subsequent bromodomain protein 4 (BRD4) overexpression. Interestingly, BRD4 is also a trigger for calcification and remodeling processes, both of which are important in CAD. Thus, we hypothesize that BRD4 activation in PAH influences the development of CAD. APPROACH AND RESULTS: PAH was associated with significant remodeling of the coronary arteries in both human and experimental models of the disease. As observed in PAH distal pulmonary arteries, coronary arteries of patients with PAH also exhibited increased DNA damage, inflammation, and BRD4 overexpression. In vitro, using human coronary artery smooth muscle cells from PAH, CAD and non-PAH-non-CAD patients, we showed that both PAH and CAD smooth muscle cells exhibited increased proliferation and suppressed apoptosis in a BRD4-dependent manner. In vivo, improvement of PAH by BRD4 inhibitor was associated with a reduction in coronary remodeling and interleukin-6 expression. CONCLUSIONS: Overall, this study demonstrates that increased BRD4 expression in coronary arteries of patient with PAH contributes to vascular remodeling and comorbidity development.


Coronary Artery Disease/metabolism , Coronary Vessels/metabolism , Epigenesis, Genetic , Hypertension, Pulmonary/metabolism , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Vascular Remodeling , Animals , Apoptosis , Case-Control Studies , Cell Cycle Proteins , Cell Proliferation , Cells, Cultured , Coronary Artery Disease/genetics , Coronary Artery Disease/pathology , Coronary Vessels/pathology , DNA Damage , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Interleukin-6/genetics , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Nuclear Proteins/genetics , Phenotype , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Transcription Factors/genetics , Vascular Remodeling/genetics
20.
Dev Dyn ; 246(1): 72-82, 2017 01.
Article En | MEDLINE | ID: mdl-27748998

BACKGROUND: Reciprocal epithelial-mesenchymal communications are critical throughout lung development, dictating branching morphogenesis and cell specification. Numerous signaling molecules are involved in these interactions, but the way epithelial-mesenchymal crosstalk is coordinated remains unclear. The ERK/MAPK pathway transduces several important signals in lung formation. Epithelial inactivation of both Mek genes, encoding ERK/MAPK kinases, causes lung agenesis and death. Conversely, Mek mutation in mesenchyme results in lung hypoplasia, trachea cartilage malformations, kyphosis, omphalocele, and death. Considering the negative impact of kyphosis and omphalocele on intrathoracic space and, consequently, on lung growth, the exact role of ERK/MAPK pathway in lung mesenchyme remains unresolved. RESULTS: To address the role of the ERK/MAPK pathway in lung mesenchyme in absence of kyphosis and omphalocele, we used the Tbx4Cre deleter mouse line, which acts specifically in lung mesenchyme. These Mek mutants did not develop kyphosis and omphalocele but they presented lung hypoplasia, tracheal defects, and neonatal death. Tracheal cartilage anomalies suggested a role for the ERK/MAPK pathway in the control of chondrocyte hypertrophy. Moreover, expression data indicated potential interactions between the ERK/MAPK and canonical Wnt pathways during lung formation. CONCLUSIONS: Lung development necessitates a functional ERK/MAPK pathway in the lung mesenchymal layer in order to coordinate efficient epithelial-mesenchymal interactions. Developmental Dynamics 246:72-82, 2017. © 2016 Wiley Periodicals, Inc.


Cell Communication , Lung/growth & development , MAP Kinase Signaling System/physiology , Mesoderm/metabolism , Organogenesis , Animals , Chondrocytes/pathology , Epithelium/embryology , Epithelium/physiology , Lung/embryology , MAP Kinase Signaling System/genetics , Mesoderm/embryology , Mesoderm/physiology , Mice , Mutation , Trachea/abnormalities , Wnt Signaling Pathway
...