Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 40
1.
Circ Res ; 133(11): 944-958, 2023 11 10.
Article En | MEDLINE | ID: mdl-37869877

BACKGROUND: ß1AR (beta-1 adrenergic receptor) and ß2AR (beta-2 adrenergic receptor)-mediated cyclic adenosine monophosphate signaling has distinct effects on cardiac function and heart failure progression. However, the mechanism regulating spatial localization and functional compartmentation of cardiac ß-ARs remains elusive. Emerging evidence suggests that microtubule-dependent trafficking of mRNP (messenger ribonucleoprotein) and localized protein translation modulates protein compartmentation in cardiomyocytes. We hypothesized that ß-AR compartmentation in cardiomyocytes is accomplished by selective trafficking of its mRNAs and localized translation. METHODS: The localization pattern of ß-AR mRNA was investigated using single molecule fluorescence in situ hybridization and subcellular nanobiopsy in rat cardiomyocytes. The role of microtubule on ß-AR mRNA localization was studied using vinblastine, and its effect on receptor localization and function was evaluated with immunofluorescent and high-throughput Förster resonance energy transfer microscopy. An mRNA protein co-detection assay identified plausible ß-AR translation sites in cardiomyocytes. The mechanism by which ß-AR mRNA is redistributed post-heart failure was elucidated by single molecule fluorescence in situ hybridization, nanobiopsy, and high-throughput Förster resonance energy transfer microscopy on 16 weeks post-myocardial infarction and detubulated cardiomyocytes. RESULTS: ß1AR and ß2AR mRNAs show differential localization in cardiomyocytes, with ß1AR found in the perinuclear region and ß2AR showing diffuse distribution throughout the cell. Disruption of microtubules induces a shift of ß2AR transcripts toward the perinuclear region. The close proximity between ß2AR transcripts and translated proteins suggests that the translation process occurs in specialized, precisely defined cellular compartments. Redistribution of ß2AR transcripts is microtubule-dependent, as microtubule depolymerization markedly reduces the number of functional receptors on the membrane. In failing hearts, both ß1AR and ß2AR mRNAs are redistributed toward the cell periphery, similar to what is seen in cardiomyocytes undergoing drug-induced detubulation. This suggests that t-tubule remodeling contributes to ß-AR mRNA redistribution and impaired ß2AR function in failing hearts. CONCLUSIONS: Asymmetrical microtubule-dependent trafficking dictates differential ß1AR and ß2AR localization in healthy cardiomyocyte microtubules, underlying the distinctive compartmentation of the 2 ß-ARs on the plasma membrane. The localization pattern is altered post-myocardial infarction, resulting from transverse tubule remodeling, leading to distorted ß2AR-mediated cyclic adenosine monophosphate signaling.


Heart Failure , Myocardial Infarction , Rats , Animals , In Situ Hybridization, Fluorescence , Heart Failure/metabolism , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Cyclic AMP/metabolism , Receptors, Adrenergic, beta-1/metabolism , Microtubules/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology
2.
Am J Respir Cell Mol Biol ; 68(1): 103-115, 2023 01.
Article En | MEDLINE | ID: mdl-36264759

Mitochondrial fission and a metabolic switch from oxidative phosphorylation to glycolysis are key features of vascular pathology in pulmonary arterial hypertension (PAH) and are associated with exuberant endothelial proliferation and apoptosis. The underlying mechanisms are poorly understood. We describe the contribution of two intracellular chloride channel proteins, CLIC1 and CLIC4, both highly expressed in PAH and cancer, to mitochondrial dysfunction and energy metabolism in PAH endothelium. Pathological overexpression of CLIC proteins induces mitochondrial fragmentation, inhibits mitochondrial cristae formation, and induces metabolic shift toward glycolysis in human pulmonary artery endothelial cells, consistent with changes observed in patient-derived cells. Interactions of CLIC proteins with structural components of the inner mitochondrial membrane offer mechanistic insights. Endothelial CLIC4 excision and mitofusin 2 supplementation have protective effects in human PAH cells and preclinical PAH. This study is the first to demonstrate the key role of endothelial intracellular chloride channels in the regulation of mitochondrial structure, biogenesis, and metabolic reprogramming in expression of the PAH phenotype.


Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Humans , Pulmonary Arterial Hypertension/metabolism , Hypertension, Pulmonary/pathology , Endothelial Cells/metabolism , Familial Primary Pulmonary Hypertension/metabolism , Pulmonary Artery/pathology , Endothelium/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism
3.
Commun Biol ; 5(1): 1192, 2022 11 07.
Article En | MEDLINE | ID: mdl-36344664

Pulmonary arterial hypertension (PAH) is an unmet clinical need. The lack of models of human disease is a key obstacle to drug development. We present a biomimetic model of pulmonary arterial endothelial-smooth muscle cell interactions in PAH, combining natural and induced bone morphogenetic protein receptor 2 (BMPR2) dysfunction with hypoxia to induce smooth muscle activation and proliferation, which is responsive to drug treatment. BMPR2- and oxygenation-specific changes in endothelial and smooth muscle gene expression, consistent with observations made in genomic and biochemical studies of PAH, enable insights into underlying disease pathways and mechanisms of drug response. The model captures key changes in the pulmonary endothelial phenotype that are essential for the induction of SMC remodelling, including a BMPR2-SOX17-prostacyclin signalling axis and offers an easily accessible approach for researchers to study pulmonary vascular remodelling and advance drug development in PAH.


Hypertension, Pulmonary , Pulmonary Arterial Hypertension , SOXF Transcription Factors , Humans , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Epoprostenol/genetics , Epoprostenol/metabolism , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Pulmonary Arterial Hypertension/genetics , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
4.
Int J Biochem Cell Biol ; 134: 105977, 2021 05.
Article En | MEDLINE | ID: mdl-33839307

Pulmonary arterial hypertension is a rare but deadly disease with a complex pathogenesis. Recent evidence demonstrates that Krüppel-like factors, a diverse family of transcription factors, are involved in several key disease processes such as the phenotypic transition of endothelial cells and smooth muscle cells. Importantly, manipulation of certain Krüppel-like factors enables protection or attenuation against pulmonary arterial hypertension in both animal models and preliminary human studies. In this review, we discuss how Krüppel-like factors, in particular Krüppel-like factors 2, 4 and 5 contribute to the pathological phenomena seen in pulmonary arterial hypertension and how associated signaling and microRNA pathways may be suitable targets for new therapies.


Endothelial Cells/pathology , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/genetics , Myocytes, Smooth Muscle/pathology , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/pathology , Animals , Endothelial Cells/metabolism , Humans , Myocytes, Smooth Muscle/metabolism , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/metabolism , Pulmonary Artery/metabolism , Signal Transduction
5.
Mol Ther Nucleic Acids ; 23: 142-153, 2021 Mar 05.
Article En | MEDLINE | ID: mdl-33335799

Circulating levels of endothelial miR-150 are reduced in pulmonary arterial hypertension (PAH) and act as an independent predictor of patient survival, but links between endothelial miR-150 and vascular dysfunction are not well understood. We studied the effects of endothelial miR-150 supplementation and inhibition in PAH mice and cells from patients with idiopathic PAH. The role of selected mediators of miR-150 identified by RNA sequencing was evaluated in vitro and in vivo. Endothelium-targeted miR-150 delivery prevented the disease in Sugen/hypoxia mice, while endothelial knockdown of miR-150 had adverse effects. miR-150 target genes revealed significant associations with PAH pathways, including proliferation, inflammation, and phospholipid signaling, with PTEN-like mitochondrial phosphatase (PTPMT1) most markedly altered. PTPMT1 reduced inflammation and apoptosis and improved mitochondrial function in human pulmonary endothelial cells and blood-derived endothelial colony-forming cells from idiopathic PAH. Beneficial effects of miR-150 in vitro and in vivo were linked with PTPMT1-dependent biosynthesis of mitochondrial phospholipid cardiolipin and reduced expression of pro-apoptotic, pro-inflammatory, and pro-fibrotic genes, including c-MYB, NOTCH3, transforming growth factor ß (TGF-ß), and Col1a1. In conclusion, we are the first to show that miR-150 supplementation attenuates pulmonary endothelial damage induced by vascular stresses and may be considered as a potential therapeutic strategy in PAH.

6.
Hypertension ; 77(2): 605-616, 2021 02.
Article En | MEDLINE | ID: mdl-33356404

Pulmonary hypertension is a complex disorder characterized by pulmonary vascular remodeling and right ventricular hypertrophy, leading to right heart failure. The mechanisms underlying this process are not well understood. We hypothesize that the structural remodeling occurring in the cardiomyocytes of the right ventricle affects the cytosolic Ca2+ handling leading to arrhythmias. After 12 days of monocrotaline-induced pulmonary hypertension in rats, epicardial mapping showed electrical remodeling in both ventricles. In myocytes isolated from the hypertensive rats, a combination of high-speed camera and confocal line-scan documented a prolongation of Ca2+ transients along with a higher local Ca2+-release activity. These Ca2+ transients were less synchronous than in controls, likely due to disorganized transverse-axial tubular system. In fact, following pulmonary hypertension, hypertrophied right ventricular myocytes showed significantly reduced number of transverse tubules and increased number of axial tubules; however, Stimulation Emission Depletion microscopy demonstrated that the colocalization of L-type Ca2+ channels and RyR2 (ryanodine receptor 2) remained unchanged. Finally, Stimulation Emission Depletion microscopy and super-resolution scanning patch-clamp analysis uncovered a decrease in the density of active L-type Ca2+ channels in right ventricular myocytes with an elevated open probability of the T-tubule anchored channels. This may represent a general mechanism of how nanoscale structural changes at the early stage of pulmonary hypertension impact on the development of the end stage failing phenotype in the right ventricle.


Calcium Signaling/physiology , Calcium/metabolism , Hypertension, Pulmonary/metabolism , Myocytes, Cardiac/metabolism , Animals , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Male , Monocrotaline , Rats , Rats, Sprague-Dawley , Vascular Remodeling/physiology
7.
Cell Death Dis ; 11(9): 729, 2020 09 09.
Article En | MEDLINE | ID: mdl-32908129

Anti-double stranded DNA antibodies (anti-dsDNA) are a hallmark of SLE but their role in disease pathogenesis is not fully resolved. Anti-dsDNA in serum are highly heterogeneous therefore in this study, we aimed to dissect the functional specificities of anti-dsDNA using a panel of human monoclonal antibodies (humAbs) generated from patients with active lupus nephritis. A total of 46 ANA reactive humAbs were isolated and divided into four broad classes based on their reactivity to histones, DNA and Crithidia. Functional analysis indicated that one subclass of antibodies bound strongly to decondensed DNA areas in neutrophil extracellular traps (NETs) and protected NETs from nuclease digestion, similar to the sera from active SLE patients. In addition, these anti-dsDNA antibodies could stimulate type I interferon responses in mononuclear phagocytic cells, or NF-kB activity in endothelial cells, by uptake of NETs-anti-NETs immune complexes and subsequently trigging inflammatory responses in an Fc-gamma receptor (Fcg-R)-dependant manner. Together our data suggest that only a subset of anti-dsDNA antibodies is capable to amplify inflammatory responses by deposit in the nephritic kidney in vivo, protecting NETs digestion as well as uptake of NETs immune complexes into Fcg-R-expressing cells in vitro.


Autoantibodies/metabolism , Inflammation/genetics , Lupus Erythematosus, Systemic/complications , Animals , Cell Death , Disease Models, Animal , Humans , Lupus Erythematosus, Systemic/pathology , Mice , Transfection
9.
Nat Commun ; 11(1): 1185, 2020 03 04.
Article En | MEDLINE | ID: mdl-32132543

Pulmonary arterial hypertension (PAH) is a severe disorder of lung vasculature that causes right heart failure. Homoeostatic effects of flow-activated transcription factor Krüppel-like factor 2 (KLF2) are compromised in PAH. Here, we show that KLF2-induced exosomal microRNAs, miR-181a-5p and miR-324-5p act together to attenuate pulmonary vascular remodelling and that their actions are mediated by Notch4 and ETS1 and other key regulators of vascular homoeostasis. Expressions of KLF2, miR-181a-5p and miR-324-5p are reduced, while levels of their target genes are elevated in pre-clinical PAH, idiopathic PAH and heritable PAH with missense p.H288Y KLF2 mutation. Therapeutic supplementation of miR-181a-5p and miR-324-5p reduces proliferative and angiogenic responses in patient-derived cells and attenuates disease progression in PAH mice. This study shows that reduced KLF2 signalling is a common feature of human PAH and highlights the potential therapeutic role of KLF2-regulated exosomal miRNAs in PAH and other diseases associated with vascular remodelling.


Genetic Therapy/methods , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/therapeutic use , Pulmonary Arterial Hypertension/therapy , Adult , Aged , Animals , Cell Proliferation/genetics , Disease Models, Animal , Disease Progression , Endothelial Cells , Exosomes/genetics , Exosomes/metabolism , Female , Gene Expression Regulation , Humans , Kruppel-Like Transcription Factors/genetics , Lung/blood supply , Lung/cytology , Lung/pathology , Male , Mice , MicroRNAs/metabolism , Middle Aged , Mutation, Missense , Primary Cell Culture , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/cytology , Pulmonary Artery/pathology , Signal Transduction/genetics , Vascular Remodeling/genetics , Young Adult
10.
Nat Chem ; 11(6): 552-561, 2019 06.
Article En | MEDLINE | ID: mdl-30936521

Post-translational farnesylation or geranylgeranylation at a C-terminal cysteine residue regulates the localization and function of over 100 proteins, including the Ras isoforms, and is a therapeutic target in diseases including cancer and infection. Here, we report global and selective profiling of prenylated proteins in living cells enabled by the development of isoprenoid analogues YnF and YnGG in combination with quantitative chemical proteomics. Eighty prenylated proteins were identified in a single human cell line, 64 for the first time at endogenous abundance without metabolic perturbation. We further demonstrate that YnF and YnGG enable direct identification of post-translationally processed prenylated peptides, proteome-wide quantitative analysis of prenylation dynamics and alternative prenylation in response to four different prenyltransferase inhibitors, and quantification of defective Rab prenylation in a model of the retinal degenerative disease choroideremia.


Alkynes/chemistry , Molecular Probes/chemistry , Protein Prenylation , Proteins/analysis , Proteome/analysis , Proteomics/methods , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Gene Knockout Techniques , Humans , Mass Spectrometry , Mice, Knockout , Protein Prenylation/drug effects , Proteins/chemistry , Proteome/chemistry
11.
Circ Res ; 124(1): 52-65, 2019 01 04.
Article En | MEDLINE | ID: mdl-30582444

RATIONALE: Increased expression of CLIC4 (chloride intracellular channel 4) is a feature of endothelial dysfunction in pulmonary arterial hypertension, but its role in disease pathology is not fully understood. OBJECTIVE: To identify CLIC4 effectors and evaluate strategies targeting CLIC4 signaling in pulmonary hypertension. METHODS AND RESULTS: Proteomic analysis of CLIC4-interacting proteins in human pulmonary artery endothelial cells identified regulators of endosomal trafficking, including Arf6 (ADP ribosylation factor 6) GTPase activating proteins and clathrin, while CLIC4 overexpression affected protein regulators of vesicular trafficking, lysosomal function, and inflammation. CLIC4 reduced BMPRII (bone morphogenetic protein receptor II) expression and signaling as a result of Arf6-mediated reduction in gyrating clathrin and increased lysosomal targeting of the receptor. BMPRII expression was restored by Arf6 siRNA, Arf inhibitor Sec7 inhibitor H3 (SecinH3), and inhibitors of clathrin-mediated endocytosis but was unaffected by chloride channel inhibitor, indanyloxyacetic acid 94 or Arf1 siRNA. The effects of CLIC4 on NF-κB (nuclear factor-kappa B), HIF (hypoxia-inducible factor), and angiogenic response were prevented by Arf6 siRNA and SecinH3. Sugen/hypoxia mice and monocrotaline rats showed elevated expression of CLIC4, activation of Arf6 and NF-κB, and reduced expression of BMPRII in the lung. These changes were established early during disease development. Lung endothelium-targeted delivery of CLIC4 siRNA or treatment with SecinH3 attenuated the disease, reduced CLIC4/Arf activation, and restored BMPRII expression in the lung. Endothelial colony-forming cells from idiopathic pulmonary hypertensive patients showed upregulation of CLIC4 expression and Arf6 activity, suggesting potential importance of this pathway in the human condition. CONCLUSIONS: Arf6 is a novel effector of CLIC4 and a new therapeutic target in pulmonary hypertension.


ADP-Ribosylation Factors/antagonists & inhibitors , Antihypertensive Agents/pharmacology , Chloride Channels/metabolism , Endothelial Cells/drug effects , Hypertension, Pulmonary/prevention & control , Mitochondrial Proteins/metabolism , Pulmonary Artery/drug effects , RNAi Therapeutics , Triazoles/pharmacology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type II/metabolism , Cells, Cultured , Chloride Channels/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Molecular Targeted Therapy , Monocrotaline , Proteomics/methods , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction
12.
Nat Nanotechnol ; 14(1): 80-88, 2019 01.
Article En | MEDLINE | ID: mdl-30510280

Much of the functionality of multicellular systems arises from the spatial organization and dynamic behaviours within and between cells. Current single-cell genomic methods only provide a transcriptional 'snapshot' of individual cells. The real-time analysis and perturbation of living cells would generate a step change in single-cell analysis. Here we describe minimally invasive nanotweezers that can be spatially controlled to extract samples from living cells with single-molecule precision. They consist of two closely spaced electrodes with gaps as small as 10-20 nm, which can be used for the dielectrophoretic trapping of DNA and proteins. Aside from trapping single molecules, we also extract nucleic acids for gene expression analysis from living cells without affecting their viability. Finally, we report on the trapping and extraction of a single mitochondrion. This work bridges the gap between single-molecule/organelle manipulation and cell biology and can ultimately enable a better understanding of living cells.


Nanotechnology , Optical Tweezers , Single-Cell Analysis , Animals , Axons/metabolism , Biopsy , Cell Line, Tumor , Cell Nucleus/metabolism , DNA/chemistry , Electricity , Electrodes , Fluorescence , Humans , Mice , Mitochondria/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Solutions
13.
Cardiovasc Res ; 113(3): 276-287, 2017 03 01.
Article En | MEDLINE | ID: mdl-28395021

Aims: RhoB plays a key role in the pathogenesis of hypoxia-induced pulmonary hypertension. Farnesylated RhoB promotes growth responses in cancer cells and we investigated whether inhibition of protein farnesylation will have a protective effect. Methods and results: The analysis of lung tissues from rodent models and pulmonary hypertensive patients showed increased levels of protein farnesylation. Oral farnesyltransferase inhibitor tipifarnib prevented development of hypoxia-induced pulmonary hypertension in mice. Tipifarnib reduced hypoxia-induced vascular cell proliferation, increased endothelium-dependent vasodilatation and reduced vasoconstriction of intrapulmonary arteries without affecting cell viability. Protective effects of tipifarnib were associated with inhibition of Ras and RhoB, actin depolymerization and increased eNOS expression in vitro and in vivo. Farnesylated-only RhoB (F-RhoB) increased proliferative responses in cultured pulmonary vascular cells, mimicking the effects of hypoxia, while both geranylgeranylated-only RhoB (GG-RhoB), and tipifarnib had an inhibitory effect. Label-free proteomics linked F-RhoB with cell survival, activation of cell cycle and mitochondrial biogenesis. Hypoxia increased and tipifarnib reduced the levels of F-RhoB-regulated proteins in the lung, reinforcing the importance of RhoB as a signalling mediator. Unlike simvastatin, tipifarnib did not increase the expression levels of Rho proteins. Conclusions: Our study demonstrates the importance of protein farnesylation in pulmonary vascular remodelling and provides a rationale for selective targeting of this pathway in pulmonary hypertension.


Antihypertensive Agents/pharmacology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Hypertension, Pulmonary/prevention & control , Hypoxia/drug therapy , Pulmonary Artery/drug effects , Quinolones/pharmacology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Farnesyltranstransferase/metabolism , Humans , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/etiology , Hypoxia/complications , Hypoxia/enzymology , Male , Mice, Inbred C57BL , Phenotype , Protein Prenylation , Proteomics/methods , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Time Factors , Transfection , Vasoconstriction/drug effects , Vasodilation/drug effects , rhoB GTP-Binding Protein/genetics , rhoB GTP-Binding Protein/metabolism
14.
Arterioscler Thromb Vasc Biol ; 36(10): 2078-87, 2016 10.
Article En | MEDLINE | ID: mdl-27470511

OBJECTIVE: Inflammation and dysregulated angiogenesis are features of endothelial dysfunction in pulmonary hypertension. Neutrophil extracellular traps (NETs), produced by dying neutrophils, contribute to pathogenesis of numerous vascular disorders but their role in pulmonary hypertension has not been studied. We sought evidence of (NETs) formation in pulmonary hypertension and investigated the effect of NETs on endothelial function. APPROACH AND RESULTS: Plasma and lung tissues of patients with pulmonary hypertension were analyzed for NET markers. The effects of NETs on endothelial function were studied in vitro and in vivo. Patients with chronic thromboembolic pulmonary hypertension and idiopathic pulmonary hypertension showed elevated plasma levels of DNA, neutrophil elastase, and myeloperoxidase. NET-forming neutrophils and extensive areas of NETosis were found in the occlusive plexiform lesions and vascularized intrapulmonary thrombi. NETs induced nuclear factor κB-dependent endothelial angiogenesis in vitro and increased vascularization of matrigel plugs in vivo. Angiogenic responses were associated with increased release of matrix metalloproteinase-9, heparin-binding epidermal growth factor-like growth factor, latency-associated peptide of the transforming growth factor ß1, and urokinase-type plasminogen activator, accompanied by increased endothelial permeability and cell motility. NETs-induced responses depended on myeloperoxidase/H2O2-dependent activation of Toll-like receptor 4/nuclear factor κB signaling. NETs stimulated the release of endothelin-1 in HPAECs (human pulmonary artery endothelial cells) and stimulated pulmonary smooth muscle cell proliferation in vitro. CONCLUSIONS: We are the first to implicate NETs in angiogenesis and provide a functional link between NETs and inflammatory angiogenesis in vitro and in vivo. We demonstrate the potential pathological relevance of this in 2 diseases of disordered vascular homeostasis, pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension.


Endothelial Cells/metabolism , Extracellular Traps/metabolism , Hypertension, Pulmonary/metabolism , Neovascularization, Pathologic , Neutrophils/metabolism , Pulmonary Artery/metabolism , Animals , Case-Control Studies , Cell Movement , Cell Proliferation , Cells, Cultured , Chloride Channels/genetics , Chloride Channels/metabolism , Coculture Techniques , Endothelial Cells/pathology , Humans , Hydrogen Peroxide/metabolism , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Male , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neutrophils/pathology , Peroxidase/metabolism , Pulmonary Artery/pathology , RNA Interference , Signal Transduction , Toll-Like Receptor 4/metabolism , Transfection
15.
J Cardiovasc Transl Res ; 9(2): 162-4, 2016 Apr.
Article En | MEDLINE | ID: mdl-26960567

Pulmonary arterial hypertension (PAH) is a chronic and progressive disease which continues to carry an unacceptably high mortality and morbidity. The nitric oxide (NO) pathway has been implicated in the pathophysiology and progression of the disease. Its extremely short half-life and systemic effects have hampered the clinical use of NO in PAH. In an attempt to circumvent these major limitations, we have developed a new NO-nanomedicine formulation. The formulation was based on hydrogel-like polymeric composite NO-releasing nanoparticles (NO-RP). The kinetics of NO release from the NO-RP showed a peak at about 120 min followed by a sustained release for over 8 h. The NO-RP did not affect the viability or inflammation responses of endothelial cells. The NO-RP produced concentration-dependent relaxations of pulmonary arteries in mice with PAH induced by hypoxia. In conclusion, NO-RP drugs could considerably enhance the therapeutic potential of NO therapy for PAH.


Antihypertensive Agents/pharmacology , Arterial Pressure/drug effects , Hypertension, Pulmonary/drug therapy , Nanoparticles , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Pulmonary Artery/drug effects , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/metabolism , Dose-Response Relationship, Drug , Drug Compounding , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Kinetics , Mice , Nanomedicine , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology
16.
J Biomed Sci ; 23: 4, 2016 Jan 19.
Article En | MEDLINE | ID: mdl-26786759

BACKGROUND: Increased circulating levels of endoglin(+) endothelial microparticles (EMPs) have been identified in several cardiovascular disorders, related to severity. Endoglin is an auxilary receptor for transforming growth factor ß (TGF-ß) important in the regulation of vascular structure. RESULTS: We quantified the number of microparticles in plasma of six patients with chronic thromboembolic pulmonary hypertension (CTEPH) and age- and sex-matched pulmonary embolic (PE) and healthy controls and investigated the role of microparticle endoglin in the regulation of pulmonary endothelial function in vitro. Results show significantly increased levels of endoglin(+) EMPs in CTEPH plasma, compared to healthy and disease controls. Co-culture of human pulmonary endothelial cells with CTEPH microparticles increased intracellular levels of endoglin and enhanced TGF-ß-induced angiogenesis and Smad1,5,8 phosphorylation in cells, without affecting BMPRII expression. In an in vitro model, we generated endothelium-derived MPs with enforced membrane localization of endoglin. Co-culture of these MPs with endothelial cells increased cellular endoglin content, improved cell survival and stimulated angiogenesis in a manner similar to the effects induced by overexpressed protein. CONCLUSIONS: Increased generation of endoglin(+) EMPs in CTEPH is likely to represent a protective mechanism supporting endothelial cell survival and angiogenesis, set to counteract the effects of vascular occlusion and endothelial damage.


Cell-Derived Microparticles/metabolism , Endothelium, Vascular/metabolism , Hypertension, Pulmonary/metabolism , Neovascularization, Pathologic/metabolism , Pulmonary Embolism/metabolism , Adult , Aged , Aged, 80 and over , Cell-Derived Microparticles/pathology , Endothelium, Vascular/pathology , Female , Humans , Hypertension, Pulmonary/pathology , Male , Middle Aged , Neovascularization, Pathologic/pathology , Pulmonary Embolism/pathology
17.
Biochem J ; 462(1): 103-12, 2014 Aug 15.
Article En | MEDLINE | ID: mdl-24895913

The NOS (nitric oxide synthase) inhibitor ADMA (asymmetric dimethylarginine) contributes to the pathogenesis of pulmonary hypertension. Reduced levels of the enzymes metabolizing ADMA, dimethylarginine dimethylaminohydrolases (DDAH1 and DDAH2) and increased levels of miR-21 are linked to disease pathology, but the mechanisms are not understood. In the present study we assessed the potential role of miR-21 in the regulation of hypoxia-induced changes in ADMA metabolism in vitro and in vivo. Hypoxia inhibited DDAH1 and DDAH2 expression and increased ADMA levels in cultured human pulmonary endothelial cells. In contrast, in human pulmonary smooth muscle cells, only DDAH2 was reduced whereas ADMA levels remained unchanged. Endothelium-specific down-regulation of DDAH1 by miR-21 in hypoxia induced endothelial dysfunction and was prevented by overexpression of DDAH1 and miR-21 blockade. DDAH1, but not DDAH2, mRNA levels were reduced, whereas miR-21 levels were elevated in lung tissues from patients with pulmonary arterial hypertension and mice with pulmonary hypertension exposed to 2 weeks of hypoxia. Hypoxic mice treated with miR-21 inhibitors and DDAH1 transgenic mice showed elevated lung DDAH1, increased cGMP levels and attenuated pulmonary hypertension. Regulation of DDAH1 by miR-21 plays a role in the development of hypoxia-induced pulmonary hypertension and may be of broader significance in pulmonary hypertension.


Amidohydrolases/metabolism , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , MicroRNAs/physiology , Animals , Arginine/analogs & derivatives , Cells, Cultured , Cyclic GMP/metabolism , Endothelial Cells/metabolism , Familial Primary Pulmonary Hypertension , Humans , Hypertension, Pulmonary/genetics , Lung/blood supply , Lung/metabolism , Male , Mice , Mice, Transgenic , Myocytes, Smooth Muscle/metabolism
18.
Cell Tissue Res ; 355(3): 675-85, 2014 Mar.
Article En | MEDLINE | ID: mdl-24599334

Pulmonary endothelial permeability is an important determinant of vascular adaptation to changes in oxygen tension, blood pressure, levels of growth factors or inflammatory cytokines. The Ras homologous (Rho) family of guanosine triphosphate phosphatases (Rho GTPases), key regulators of the actin cytoskeleton, regulate endothelial barrier function in response to a variety of environmental factors and signalling agents via the reorganization of the actin cytoskeleton, changes in receptor trafficking or the phosphorylation of junctional proteins. This review provides a brief summary of recent knowledge on Rho-GTPase-mediated effects on pulmonary endothelial barrier function and focuses in particular on their role in pulmonary vascular disorders, including pulmonary hypertension, chronic obstructive pulmonary disease, acute lung injury and acute respiratory distress syndrome.


Actins/physiology , Capillary Permeability/physiology , Endothelium, Vascular/physiology , Lung/physiology , rho GTP-Binding Proteins/physiology , Actins/metabolism , Animals , Endothelium, Vascular/metabolism , Humans , Lung/cytology , Lung/enzymology , rho GTP-Binding Proteins/metabolism
19.
Circulation ; 129(17): 1770-80, 2014 Apr 29.
Article En | MEDLINE | ID: mdl-24503951

BACKGROUND: Chloride intracellular channel 4 (CLIC4) is highly expressed in the endothelium of remodeled pulmonary vessels and plexiform lesions of patients with pulmonary arterial hypertension. CLIC4 regulates vasculogenesis through endothelial tube formation. Aberrant CLIC4 expression may contribute to the vascular pathology of pulmonary arterial hypertension. METHODS AND RESULTS: CLIC4 protein expression was increased in plasma and blood-derived endothelial cells from patients with idiopathic pulmonary arterial hypertension and in the pulmonary vascular endothelium of 3 rat models of pulmonary hypertension. CLIC4 gene deletion markedly attenuated the development of chronic hypoxia-induced pulmonary hypertension in mice. Adenoviral overexpression of CLIC4 in cultured human pulmonary artery endothelial cells compromised pulmonary endothelial barrier function and enhanced their survival and angiogenic capacity, whereas CLIC4 shRNA had an inhibitory effect. Similarly, inhibition of CLIC4 expression in blood-derived endothelial cells from patients with idiopathic pulmonary arterial hypertension attenuated the abnormal angiogenic behavior that characterizes these cells. The mechanism of CLIC4 effects involves p65-mediated activation of nuclear factor-κB, followed by stabilization of hypoxia-inducible factor-1α and increased downstream production of vascular endothelial growth factor and endothelin-1. CONCLUSION: Increased CLIC4 expression is an early manifestation and mediator of endothelial dysfunction in pulmonary hypertension.


Chloride Channels/physiology , Endothelium, Vascular/physiopathology , Hypertension, Pulmonary/physiopathology , Mitochondrial Proteins/physiology , Pulmonary Artery/physiopathology , Animals , Cells, Cultured , Chloride Channels/genetics , Endothelium, Vascular/cytology , Familial Primary Pulmonary Hypertension , Humans , Hypertension, Pulmonary/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Mitochondrial Proteins/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiopathology , Pulmonary Artery/cytology , Rats , Rats, Sprague-Dawley , Transcription Factor RelA/physiology
20.
Vascul Pharmacol ; 58(3): 202-10, 2013 Mar.
Article En | MEDLINE | ID: mdl-22982942

Rho proteins, best known for their regulatory role in actin dynamics, stimulate a variety of processes important in the control of vascular function, including morphogenesis, migration, cell proliferation and adhesion, cell survival, gene expression, vesicle transport and microparticle formation. Rho GTPases have been implicated in several pulmonary vascular pathologies. Here we give an overview of the current knowledge of the role of Rho GTPases in vascular dysfunction, and pulmonary diseases such as pulmonary hypertension, pulmonary embolism, chronic obstructive pulmonary disease, acute lung injury and acute respiratory distress syndrome.


Lung Diseases/physiopathology , Vascular Diseases/physiopathology , rho GTP-Binding Proteins/metabolism , Actins/metabolism , Animals , Humans , Hypertension, Pulmonary/physiopathology , Pulmonary Circulation
...