Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Eur Respir J ; 61(6)2023 06.
Article En | MEDLINE | ID: mdl-37024132

INTRODUCTION: Pulmonary arterial hypertension (PAH) is characterised by loss of microvessels. The Wnt pathways control pulmonary angiogenesis but their role in PAH is incompletely understood. We hypothesised that Wnt activation in pulmonary microvascular endothelial cells (PMVECs) is required for pulmonary angiogenesis, and its loss contributes to PAH. METHODS: Lung tissue and PMVECs from healthy and PAH patients were screened for Wnt production. Global and endothelial-specific Wnt7a -/- mice were generated and exposed to chronic hypoxia and Sugen-hypoxia (SuHx). RESULTS: Healthy PMVECs demonstrated >6-fold Wnt7a expression during angiogenesis that was absent in PAH PMVECs and lungs. Wnt7a expression correlated with the formation of tip cells, a migratory endothelial phenotype critical for angiogenesis. PAH PMVECs demonstrated reduced vascular endothelial growth factor (VEGF)-induced tip cell formation as evidenced by reduced filopodia formation and motility, which was partially rescued by recombinant Wnt7a. We discovered that Wnt7a promotes VEGF signalling by facilitating Y1175 tyrosine phosphorylation in vascular endothelial growth factor receptor 2 (VEGFR2) through receptor tyrosine kinase-like orphan receptor 2 (ROR2), a Wnt-specific receptor. We found that ROR2 knockdown mimics Wnt7a insufficiency and prevents recovery of tip cell formation with Wnt7a stimulation. While there was no difference between wild-type and endothelial-specific Wnt7a -/- mice under either chronic hypoxia or SuHx, global Wnt7a +/- mice in hypoxia demonstrated higher pulmonary pressures and severe right ventricular and lung vascular remodelling. Similar to PAH, Wnt7a +/- PMVECs exhibited an insufficient angiogenic response to VEGF-A that improved with Wnt7a. CONCLUSIONS: Wnt7a promotes VEGF signalling in lung PMVECs and its loss is associated with an insufficient VEGF-A angiogenic response. We propose that Wnt7a deficiency contributes to progressive small vessel loss in PAH.


Pulmonary Arterial Hypertension , Mice , Animals , Pulmonary Arterial Hypertension/complications , Vascular Endothelial Growth Factor A/metabolism , Endothelial Cells/metabolism , Familial Primary Pulmonary Hypertension/metabolism , Hypoxia/metabolism
2.
Angiogenesis ; 23(4): 699-714, 2020 11.
Article En | MEDLINE | ID: mdl-32813135

Imbalanced transforming growth factor beta (TGFß) and bone morphogenetic protein (BMP) signaling are postulated to favor a pathological pulmonary endothelial cell (EC) phenotype in pulmonary arterial hypertension (PAH). BMP9 is shown to reinstate BMP receptor type-II (BMPR2) levels and thereby mitigate hemodynamic and vascular abnormalities in several animal models of pulmonary hypertension (PH). Yet, responses of the pulmonary endothelium of PAH patients to BMP9 are unknown. Therefore, we treated primary PAH patient-derived and healthy pulmonary ECs with BMP9 and observed that stimulation induces transient transcriptional signaling associated with the process of endothelial-to-mesenchymal transition (EndMT). However, solely PAH pulmonary ECs showed signs of a mesenchymal trans-differentiation characterized by a loss of VE-cadherin, induction of transgelin (SM22α), and reorganization of the cytoskeleton. In the PAH cells, a prolonged EndMT signaling was found accompanied by sustained elevation of pro-inflammatory, pro-hypoxic, and pro-apoptotic signaling. Herein we identified interleukin-6 (IL6)-dependent signaling to be the central mediator required for the BMP9-induced phenotypic change in PAH pulmonary ECs. Furthermore, we were able to target the BMP9-induced EndMT process by an IL6 capturing antibody that normalized autocrine IL6 levels, prevented mesenchymal transformation, and maintained a functional EC phenotype in PAH pulmonary ECs. In conclusion, our results show that the BMP9-induced aberrant EndMT in PAH pulmonary ECs is dependent on exacerbated pro-inflammatory signaling mediated through IL6.


Endothelial Cells/metabolism , Growth Differentiation Factor 2/metabolism , Inflammation/metabolism , Inflammation/pathology , Lung/pathology , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Signal Transduction , Adult , Aged , Endothelium, Vascular/pathology , Female , Homeostasis , Humans , Interleukin-6/metabolism , Male , Microvessels/pathology , Middle Aged , Neutralization Tests , Phenotype , Pulmonary Arterial Hypertension/genetics , Transcription, Genetic
3.
Sci Transl Med ; 12(543)2020 05 13.
Article En | MEDLINE | ID: mdl-32404506

Human genetics, biomarker, and animal studies implicate loss of function in bone morphogenetic protein (BMP) signaling and maladaptive transforming growth factor-ß (TGFß) signaling as drivers of pulmonary arterial hypertension (PAH). Although sharing common receptors and effectors with BMP/TGFß, the function of activin and growth and differentiation factor (GDF) ligands in PAH are less well defined. Increased expression of GDF8, GDF11, and activin A was detected in lung lesions from humans with PAH and experimental rodent models of pulmonary hypertension (PH). ACTRIIA-Fc, a potent GDF8/11 and activin ligand trap, was used to test the roles of these ligands in animal and cellular models of PH. By blocking GDF8/11- and activin-mediated SMAD2/3 activation in vascular cells, ACTRIIA-Fc attenuated proliferation of pulmonary arterial smooth muscle cells and pulmonary microvascular endothelial cells. In several experimental models of PH, prophylactic administration of ACTRIIA-Fc markedly improved hemodynamics, right ventricular (RV) hypertrophy, RV function, and arteriolar remodeling. When administered after the establishment of hemodynamically severe PH in a vasculoproliferative model, ACTRIIA-Fc was more effective than vasodilator in attenuating PH and arteriolar remodeling. Potent antiremodeling effects of ACTRIIA-Fc were associated with inhibition of SMAD2/3 activation and downstream transcriptional activity, inhibition of proliferation, and enhancement of apoptosis in the vascular wall. ACTRIIA-Fc reveals an unexpectedly prominent role of GDF8, GDF11, and activin as drivers of pulmonary vascular disease and represents a therapeutic strategy for restoring the balance between SMAD1/5/9 and SMAD2/3 signaling in PAH.


Hypertension, Pulmonary , Activins , Animals , Cell Differentiation , Endothelial Cells , Hypertension, Pulmonary/drug therapy , Signal Transduction
4.
Am J Respir Crit Care Med ; 199(7): 891-902, 2019 04 01.
Article En | MEDLINE | ID: mdl-30312106

RATIONALE: BMP9 (bone morphogenetic protein 9) is a circulating endothelial quiescence factor with protective effects in pulmonary arterial hypertension (PAH). Loss-of-function mutations in BMP9, its receptors, and downstream effectors have been reported in heritable PAH. OBJECTIVES: To determine how an acquired deficiency of BMP9 signaling might contribute to PAH. METHODS: Plasma levels of BMP9 and antagonist soluble endoglin were measured in group 1 PAH, group 2 and 3 pulmonary hypertension (PH), and in patients with severe liver disease without PAH. MEASUREMENTS AND MAIN RESULTS: BMP9 levels were markedly lower in portopulmonary hypertension (PoPH) versus healthy control subjects, or other etiologies of PAH or PH; distinguished PoPH from patients with liver disease without PAH; and were an independent predictor of transplant-free survival. BMP9 levels were decreased in mice with PH associated with CCl4-induced portal hypertension and liver cirrhosis, but were normal in other rodent models of PH. Administration of ALK1-Fc, a BMP9 ligand trap consisting of the activin receptor-like kinase-1 extracellular domain, exacerbated PH and pulmonary vascular remodeling in mice treated with hypoxia versus hypoxia alone. CONCLUSIONS: BMP9 is a sensitive and specific biomarker of PoPH, predicting transplant-free survival and the presence of PAH in liver disease. In rodent models, acquired deficiency of BMP9 signaling can predispose to or exacerbate PH, providing a possible mechanistic link between PoPH and heritable PAH. These findings describe a novel experimental model of severe PH that provides insight into the synergy between pulmonary vascular injury and diminished BMP9 signaling in the pathogenesis of PAH.


Bone Morphogenetic Proteins/metabolism , Hypertension, Portal/metabolism , Hypertension, Portal/physiopathology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Liver Diseases/metabolism , Liver Diseases/physiopathology , Adult , Biomarkers/blood , Biomarkers/metabolism , Female , Humans , Male , Middle Aged
6.
Sci Transl Med ; 10(445)2018 06 13.
Article En | MEDLINE | ID: mdl-29899023

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


Adaptor Proteins, Signal Transducing/metabolism , Collagen Type III/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Lung/metabolism , Lung/pathology , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Collagen Type III/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Lung/physiopathology , Male , Phosphoproteins/genetics , Protein Binding , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Smad3 Protein/genetics , Smad3 Protein/metabolism , Systems Biology/methods
7.
Am J Respir Crit Care Med ; 194(9): 1140-1151, 2016 11 01.
Article En | MEDLINE | ID: mdl-27115515

RATIONALE: Transforming growth factor-ß (TGF-ß) ligands signal via type I and type II serine-threonine kinase receptors to regulate broad transcriptional programs. Excessive TGF-ß-mediated signaling is implicated in the pathogenesis of pulmonary arterial hypertension, based in part on the ability of broad inhibition of activin-like kinase (ALK) receptors 4/5/7 recognizing TGF-ß, activin, growth and differentiation factor, and nodal ligands to attenuate experimental pulmonary hypertension (PH). These broad inhibition strategies do not delineate the specific contribution of TGF-ß versus a multitude of other ligands, and their translation is limited by cardiovascular and systemic toxicity. OBJECTIVES: We tested the impact of a soluble TGF-ß type II receptor extracellular domain expressed as an immunoglobulin-Fc fusion protein (TGFBRII-Fc), serving as a selective TGF-ß1/3 ligand trap, in several experimental PH models. METHODS: Signaling studies used cultured human pulmonary artery smooth muscle cells. PH was studied in monocrotaline-treated Sprague-Dawley rats, SU5416/hypoxia-treated Sprague-Dawley rats, and SU5416/hypoxia-treated C57BL/6 mice. PH, cardiac function, vascular remodeling, and valve structure were assessed by ultrasound, invasive hemodynamic measurements, and histomorphometry. MEASUREMENTS AND MAIN RESULTS: TGFBRII-Fc is an inhibitor of TGF-ß1 and TGF-ß3, but not TGF-ß2, signaling. In vivo treatment with TGFBRII-Fc attenuated Smad2 phosphorylation, normalized expression of plasminogen activator inhibitor-1, and mitigated PH and pulmonary vascular remodeling in monocrotaline-treated rats, SU5416/hypoxia-treated rats, and SU5416/hypoxia-treated mice. Administration of TGFBRII-Fc to monocrotaline-treated or SU5416/hypoxia-treated rats with established PH improved right ventricular systolic pressures, right ventricular function, and survival. No cardiac structural or valvular abnormalities were observed after treatment with TGFBRII-Fc. CONCLUSIONS: Our findings are consistent with a pathogenetic role of TGF-ß1/3, demonstrating the efficacy and tolerability of selective TGF-ß ligand blockade for improving hemodynamics, remodeling, and survival in multiple experimental PH models.


Hypertension, Pulmonary/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Disease Models, Animal , Heart/physiopathology , Hemodynamics/physiology , Hypertension, Pulmonary/physiopathology , Immunoglobulin Fc Fragments/metabolism , Ligands , Male , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Proteins , Signal Transduction/drug effects , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Vascular Remodeling/physiology
8.
FASEB J ; 30(7): 2511-27, 2016 07.
Article En | MEDLINE | ID: mdl-27006450

Activation of the mammalian target of rapamycin complex 1 (mTORC1) subunit Raptor induces cell growth and is a downstream target of Akt. Elevated levels of aldosterone activate Akt, and, in pulmonary arterial hypertension (PAH), correlate with pulmonary arteriole thickening, which suggests that mTORC1 regulation by aldosterone may mediate adverse pulmonary vascular remodeling. We hypothesized that aldosterone-Raptor signaling induces abnormal pulmonary artery smooth muscle cell (PASMC) survival patterns to promote PAH. Remodeled pulmonary arterioles from SU-5416/hypoxia-PAH rats and monocrotaline-PAH rats with hyperaldosteronism expressed increased levels of the Raptor target, p70S6K, which provided a basis for investigating aldosterone-Raptor signaling in human PASMCs. Aldosterone (10(-9) to 10(-7) M) increased Akt/mTOR/Raptor to activate p70S6K and increase proliferation, viability, and apoptosis resistance in PASMCs. In PASMCs transfected with Raptor-small interfering RNA or treated with spironolactone/eplerenone, aldosterone or pulmonary arterial plasma from patients with PAH failed to increase p70S6K activation or to induce cell survival in vitro Optimal inhibition of pulmonary arteriole Raptor was achieved by treatment with Staramine-monomethoxy polyethylene glycol that was formulated with Raptor-small interfering RNA plus spironolactone in vivo, which decreased arteriole muscularization and pulmonary hypertension in 2 experimental animal models of PAH in vivo Up-regulation of mTORC1 by aldosterone is a critical pathobiologic mechanism that controls PASMC survival to promote hypertrophic vascular remodeling and PAH.-Aghamohammadzadeh, R., Zhang, Y.-Y., Stephens, T. E., Arons, E., Zaman, P., Polach, K. J., Matar, M., Yung, L.-M., Yu, P. B., Bowman, F. P., Opotowsky, A. R., Waxman, A. B., Loscalzo, J., Leopold, J. A., Maron, B. A. Up-regulation of the mammalian target of rapamycin complex 1 subunit Raptor by aldosterone induces abnormal pulmonary artery smooth muscle cell survival patterns to promote pulmonary arterial hypertension.


Adaptor Proteins, Signal Transducing/metabolism , Aldosterone/pharmacology , Gene Expression Regulation/physiology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Up-Regulation , Adaptor Proteins, Signal Transducing/genetics , Aldosterone/metabolism , Animals , Apoptosis/physiology , Cell Proliferation , Cell Survival , Cells, Cultured , Humans , Hypertension, Pulmonary , Male , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/genetics , Rats , Rats, Sprague-Dawley , Regulatory-Associated Protein of mTOR , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics
9.
Eur J Nutr ; 55(5): 1963-72, 2016 Aug.
Article En | MEDLINE | ID: mdl-26276555

PURPOSE: Menopause escalates the risk of cardiovascular diseases in women. There is an unmet need for better treatment strategy for estrogen-deficiency-related cardiovascular complications. Here we investigated the impact of chronic black tea extract (BT) consumption on cardiovascular function and lipid metabolism using a rat model of estrogen deficiency. METHODS: Female Sprague-Dawley rats were ovariectomized (OVX) and treated with BT (15 mg/kg/day, 4 weeks; active ingredients: theaflavins) or estrogen (E2) treatment for 4 weeks. Serum was collected for measuring cholesterol, triacylglycerol and estradiol levels. Changes in vascular reactivity were examined. The protein levels of NADPH oxidases were assessed by Western blotting. Reactive oxygen species (ROS) level was measured using dihydroethidium fluorescence imaging. The concentrations of cGMP were measured using ELISA kit. RESULTS: Aortic rings from control, BT-treated and E2-treated OVX rats exhibited a greater increase in Phe-induced contraction after inhibition of NO synthase compared with those from OVX rats. ACh-induced endothelium-dependent relaxations were augmented in aortae and renal arteries in BT/E2-treated OVX rats than in OVX rats. BT/E2 treatment improved flow-mediated dilatation in small mesenteric resistance arteries of OVX rats. BT/E2 treatment restored the eNOS phosphorylation level and reversed the up-regulation of NADPH oxidases and ROS overproduction in OVX rat aortae. ACh-stimulated cGMP production was significantly elevated in the aortae from BT- and E2-treated rats compared with those from OVX rats. BT/E2 treatment reduced circulating levels of total cholesterol. CONCLUSIONS: The present study reveals the novel benefits of chronic BT consumption to reverse endothelial dysfunction and favorably modifying cholesterol profile in a rat model of estrogen deficiency and provides insights into developing BT as beneficial dietary supplements for postmenopausal women.


Endothelium, Vascular/drug effects , Ovariectomy , Plant Extracts/pharmacology , Tea/chemistry , Animals , Antioxidants/pharmacology , Aorta/drug effects , Aorta/metabolism , Biflavonoids/pharmacology , Catechin/pharmacology , Endothelium, Vascular/metabolism , Estrogens/pharmacology , Female , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Up-Regulation
10.
Cardiovasc Res ; 108(2): 278-87, 2015 Nov 01.
Article En | MEDLINE | ID: mdl-26410368

AIMS: Vascular calcification contributes to mortality and morbidity in atherosclerosis, chronic kidney disease, and diabetes. Vascular calcific lesions contain osteoblast- and chondroblast-like cells, suggesting a process of endochondral or membranous ossification thought to result from the phenotypic plasticity of vascular cells. Bone morphogenetic protein (BMP) signalling potentiates atherosclerotic calcification, whereas BMP inhibition attenuates vascular inflammation and calcification in atherogenic mice. We hypothesized endothelial cells (ECs) may undergo osteogenic differentiation in response to BMP signalling and pro-atherogenic stimuli. METHODS AND RESULTS: Among various BMP ligands tested, BMP6 and BMP9 elicited the most potent signalling in bovine aortic endothelial cells (BAEC), however, only BMP6 induced osteogenic differentiation. BMP6 and oxidized low-density lipoprotein (oxLDL) independently and synergistically induced osteogenic differentiation and mineralization, in a manner consistent with endothelial-to-mesenchymal transition. Treatment of ECs with BMP6 or oxLDL individually induced osteogenic and chondrogenic transcription factors Runx2 and Msx2, whereas treatment with BMP6 and oxLDL synergistically up-regulated Osterix and Osteopontin. Production of H2O2 was necessary for oxLDL-induced regulation of Runx2, Msx2, and Osterix in BAEC, and H2O2 was sufficient by itself to up-regulate these genes. Mineralization of ECs in response to BMP6 or oxLDL was abrogated by scavenging reactive oxygen species or inhibiting BMP type I receptor kinases. Similar synergistic effects of BMP and oxLDL upon osteogenic and chondrogenic transcription and phenotypic plasticity in human aortic endothelial cells were observed. CONCLUSION: These findings provide a potential mechanism for the observed interactions of BMP signalling, oxidative stress, and inflammation in recruiting vascular calcification associated with atherosclerosis.


Bone Morphogenetic Protein 6/metabolism , Cell Differentiation , Endothelial Cells/physiology , Lipoproteins, LDL/metabolism , Vascular Calcification/etiology , Animals , Cattle , Cell Line , Humans , Osteogenesis , Vascular Calcification/metabolism
11.
Nat Med ; 21(7): 777-85, 2015 Jul.
Article En | MEDLINE | ID: mdl-26076038

Genetic evidence implicates the loss of bone morphogenetic protein type II receptor (BMPR-II) signaling in the endothelium as an initiating factor in pulmonary arterial hypertension (PAH). However, selective targeting of this signaling pathway using BMP ligands has not yet been explored as a therapeutic strategy. Here, we identify BMP9 as the preferred ligand for preventing apoptosis and enhancing monolayer integrity in both pulmonary arterial endothelial cells and blood outgrowth endothelial cells from subjects with PAH who bear mutations in the gene encoding BMPR-II, BMPR2. Mice bearing a heterozygous knock-in allele of a human BMPR2 mutation, R899X, which we generated as an animal model of PAH caused by BMPR-II deficiency, spontaneously developed PAH. Administration of BMP9 reversed established PAH in these mice, as well as in two other experimental PAH models, in which PAH develops in response to either monocrotaline or VEGF receptor inhibition combined with chronic hypoxia. These results demonstrate the promise of direct enhancement of endothelial BMP signaling as a new therapeutic strategy for PAH.


Bone Morphogenetic Protein Receptors, Type II/metabolism , Endothelial Cells/metabolism , Growth Differentiation Factor 2/pharmacology , Hypertension, Pulmonary/pathology , Pulmonary Artery/pathology , Aging/pathology , Animals , Apoptosis/drug effects , Cell Membrane Permeability/drug effects , Densitometry , Endothelial Cells/drug effects , Endothelial Cells/pathology , Gene Expression Profiling , Gene Knock-In Techniques , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Immunoblotting , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Monocrotaline , Phosphorylation/drug effects , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , Systole/drug effects , Transcription, Genetic/drug effects
12.
Eur J Nutr ; 52(3): 1145-55, 2013 Apr.
Article En | MEDLINE | ID: mdl-22836513

PURPOSE: Postmenopausal women experience higher risks for cardiovascular diseases than age-matched men and pre-menopausal women. There is a need for better treatment strategy for estrogen-deficient-related cardiovascular complications. We and others have recently reported that activated renin-angiotensin system and the associated oxidative stress impaired endothelium-dependent relaxation in ovariectomized rat, while angiotensin receptor blocker rescues endothelial dysfunction. Dietary supplements and lifestyle modifications provide an alternative way to improve cardiovascular health. The present study tests the hypothesis that chronic cranberry juice consumption improves cholesterol profiles and vascular functions in estrogen-deficient animal model. The effect of cranberry consumption on expression and activity of renin-angiotensin system in the vasculature will be determined. METHODS: Ovariectomized rats were treated daily with commercial cranberry juice at 7 mg/kg for 8 weeks, a dosage comparable to recent clinical studies. Serum was collected for measuring cholesterol levels while aorta was isolated for isometric force assay and expression studies. RESULTS: Cranberry juice consumption reduced circulating levels of total cholesterol, triacylglycerols, HDL, nHDL, and nHDL/HDL ratio. Meanwhile, cranberry juice consumption improved endothelium-dependent relaxation in aorta of ovariectomized rats by restoring p-eNOS level (endothelial nitric oxide synthase phosphorylated at ser-1177), reversing the up-regulated levels of renin-angiotensin system markers (angiotensin-converting enzyme, angiotensin II, and angiotensin II type 1 receptor), and normalizing the elevated NAD(P)H oxidase expression and oxidative stress. CONCLUSIONS: Our data demonstrate the novel cardiovascular benefits of cranberry juice consumption in improving both vascular functions and cholesterol profiles, providing insight into developing cranberry products into useful dietary supplements for postmenopausal women.


Aging , Anticholesteremic Agents/therapeutic use , Beverages , Endothelium, Vascular/physiopathology , Fruit/chemistry , Hypercholesterolemia/prevention & control , Vaccinium macrocarpon/chemistry , Animals , Anticholesteremic Agents/analysis , Anticholesteremic Agents/chemistry , Antioxidants/analysis , Antioxidants/chemistry , Antioxidants/therapeutic use , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Beverages/analysis , Cholesterol/blood , Down-Regulation , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Hypercholesterolemia/physiopathology , Nitric Oxide Synthase Type III/metabolism , Ovariectomy/adverse effects , Oxidative Stress , Phosphorylation , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Renin-Angiotensin System , Vasodilation
13.
PLoS One ; 7(9): e44622, 2012.
Article En | MEDLINE | ID: mdl-23028567

Hepcidin is an antimicrobial peptide, which also negatively regulates iron in circulation by controlling iron absorption from dietary sources and iron release from macrophages. Hepcidin is synthesized mainly in the liver, where hepcidin is regulated by iron loading, inflammation and hypoxia. Recently, we have demonstrated that bone morphogenetic protein (BMP)-hemojuvelin (HJV)-SMAD signaling is central for hepcidin regulation in hepatocytes. Hepcidin is also expressed by macrophages. Studies have shown that hepcidin expression by macrophages increases following bacterial infection, and that hepcidin decreases iron release from macrophages in an autocrine and/or paracrine manner. Although previous studies have shown that lipopolysaccharide (LPS) can induce hepcidin expression in macrophages, whether hepcidin is also regulated by BMPs in macrophages is still unknown. Therefore, we examined the effects of BMP signaling on hepcidin expression in RAW 264.7 and J774 macrophage cell lines, and in primary peritoneal macrophages. We found that BMP4 or BMP6 alone did not have any effect on hepcidin expression in macrophages although they stimulated Smad1/5/8 phosphorylation and Id1 expression. In the presence of LPS, however, BMP4 and BMP6 were able to stimulate hepcidin expression in macrophages, and this stimulation was abolished by the NF-κB inhibitor Ro1069920. These results suggest that hepcidin expression is regulated differently in macrophages than in hepatocytes, and that BMPs regulate hepcidin expression in macrophages in a LPS-NF-κB dependent manner.


Bone Morphogenetic Proteins/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Animals , Antimicrobial Cationic Peptides , Blotting, Western , Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Protein 6/pharmacology , Cell Line , Cells, Cultured , Hepcidins , Inhibitor of Differentiation Protein 1/metabolism , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Phosphorylation/drug effects , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Sulfoxides/pharmacology , Tetrazoles/pharmacology
14.
Stroke ; 43(1): 199-204, 2012 Jan.
Article En | MEDLINE | ID: mdl-21980199

BACKGROUND AND PURPOSE: Cerebral preconditioning provides insights into endogenous mechanisms that protect the brain from ischemic injury. Hypoxia and the anesthetic isoflurane are powerful preconditioning agents. Recent data show that sphingosine 1-phosphate receptor stimulation improves outcome in rodent models of stroke. Endogenous sphingosine 1-phosphate levels are controlled by the expression and activity of sphingosine kinases (SPK). We hypothesize that SPK upregulation mediates preconditioning induced by isoflurane and hypoxia and reduces ischemic injury. METHODS: Male wild-type C57BL/J, SPK1(-/-) and SPK2(-/-) mice were exposed to isoflurane or hypoxia preconditioning before transient middle cerebral artery occlusion. Infarct volume and neurological outcome were measured 24 hours later. SPK inhibitors (SKI-II and ABC294640) were used to test the involvement of SPK2. Expressions of SPK1, SPK2, and hypoxia-inducible factor 1α were determined. Primary cultures of mouse cortical neurons were exposed to isoflurane before glutamate- or hydrogen peroxide-induced cell death. RESULTS: Isoflurane preconditioning and hypoxia preconditioning significantly reduced infarct volume and improved neurological outcome in wild-type and SPK1(-/-) mice but not in SPK2(-/-) mice. Pretreatment with SKI-II or ABC294640 abolished the isoflurane preconditioning-induced tolerance. Western blot showed a rapid and sustained increase in SPK2 level, whereas SPK1 level was similar between preconditioned mice and controls. Hypoxia-inducible factor 1α was upregulated in wild-type isoflurane-preconditioned mice but not in SPK2(-/-). Isoflurane preconditioning protected primary neurons against cell death, which was abolished in ABC294640-treated cells. CONCLUSIONS: Applying genetic and pharmacological approaches, we demonstrate that neuronal SPK2 isoform plays an important role in cerebral preconditioning.


Brain Ischemia/metabolism , Brain/metabolism , Ischemic Preconditioning/methods , Neurons/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Brain/pathology , Brain Ischemia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Knockout , Neurons/pathology , Phosphotransferases (Alcohol Group Acceptor)/genetics
15.
Pharmacol Res ; 65(2): 182-8, 2012 Feb.
Article En | MEDLINE | ID: mdl-22005391

Although vascular effects of selective estrogen receptor modulators (SERMs) have been extensively examined in conduit arteries, whether SERMs could favorably modulate myogenic response in resistance arteries is unknown. The impact of raloxifene therapy and cholesterol diet on myogenic constriction during estrogen deficiency is unresolved. This study investigated changes in vascular reactivity and myogenic responses in female ovariectomized (Ovx) hamsters fed high-cholesterol diet (HCD) with and without chronic treatment of raloxifene. Functional studies were performed on hamster septal coronary arteries cannulated in a pressure myograph. Acetylcholine (ACh)-induced dilatation was reduced in arteries from cholesterol-fed Ovx hamsters, but not in those from cholesterol-fed hamsters, while pressure-induced myogenic constriction was unaffected. Chronic treatment with raloxifene restored ACh-induced dilatation in cholesterol-fed Ovx hamsters. U46619-induced constriction was increased in arteries from cholesterol-fed Ovx hamsters but not from cholesterol-fed control hamsters, which was normalized by chronic raloxifene treatment. The pressure-diameter relationship is presented as normalized diameter versus intraluminal pressure, while the effect of ACh or U46619 is expressed as percentage of tone at 80 mm Hg. Two-way analysis of variance (ANOVA) followed by Bonferroni post-tests were used for statistical evaluation among different treatment groups. P<0.05 was taken as statistically significant. The present results show that chronic treatment with raloxifene could benefit myogenically active coronary arteries by (i) restoring ACh-induced dilatation and (ii) reducing U46619-induced constriction without affecting pressure-induced myogenic responses in cholesterol-fed hamsters during estrogen deficiency. If such benefit can be observed in humans, raloxifene and other SERMs may be useful to preserve endothelial function and curtail vascular hypersensitivity in resistance coronary arteries in post-menopausal women with hypercholesterolemia or hyperlipidemia, a lipid condition implicated in the pathogenesis of myocardial infarction.


Cholesterol, Dietary/administration & dosage , Coronary Vessels/drug effects , Endothelium, Vascular/drug effects , Raloxifene Hydrochloride/pharmacology , Acetylcholine/pharmacology , Animals , Cholesterol, Dietary/blood , Cholesterol, Dietary/toxicity , Cholesterol, LDL/blood , Cricetinae , Endothelium, Vascular/physiology , Female , Lipase/blood , Mesocricetus , Muscle Development/drug effects , Muscle Development/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Ovariectomy/methods , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/drug effects , Vasodilation/physiology
16.
J Mol Cell Cardiol ; 52(1): 237-44, 2012 Jan.
Article En | MEDLINE | ID: mdl-22064324

The expression of bone morphogenic protein 4 (BMP4), a new pro-inflammatory marker, is increased by disturbed flow in endothelial cells (ECs). BMP4 stimulates production of reactive oxygen species (ROS) and causes endothelial cell dysfunction. The present study examined BMP4-induced apoptosis in ECs and isolated arteries from rat, mouse, and human, and the signaling pathways mediating BMP4-induced apoptosis. Apoptosis was assessed by flow cytometry to detect Annexin-V positive cells, and terminal deoxynucleotidyl transferase dUTP nick end (TUNEL) labeling. The superoxide production was measured by dihydroethidium fluorescence. BMP4 induced EC apoptosis in human mesenteric arteries, mouse aortic endothelium, rat primary ECs, and human ECs. BMP4-induced EC apoptosis was mediated through ROS production by activation of NADPH oxidase, which led to cleaved caspase-3 expression. BMP4 also induced sequential activation of p38 MAPK and JNK which was upstream of caspase 3 activation. Knockdown of BMP receptor 1A by lentiviral shRNA or NOX4 siRNA transfection inhibited BMP4-induced ROS production, p38 and JNK phosphorylation, and caspase-3 activation in ECs. JNK siRNA inhibited BMP4-induced JNK phosphorylation and caspase-3 activation. The present study delineates that BMP4 causes EC apoptosis through activation of caspase-3 in a ROS/p38MAPK/JNK-dependent signaling cascade.


Apoptosis , Bone Morphogenetic Protein 4/pharmacology , Endothelial Cells/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Oxidative Stress , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type I/metabolism , Caspase 3/metabolism , Endothelial Cells/drug effects , Gene Knockdown Techniques , Humans , In Vitro Techniques , Male , Mesenteric Arteries/cytology , Mesenteric Arteries/metabolism , Mice , NADPH Oxidases/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
17.
PLoS One ; 6(3): e17437, 2011 Mar 29.
Article En | MEDLINE | ID: mdl-21479266

BACKGROUND: Estrogen deficiency increases the cardiovascular risks in postmenopausal women. Inhibition of the renin-angiotensin system (RAS) and associated oxidative stress confers a cardiovascular protection, but the role of RAS in estrogen deficiency-related vascular dysfunction is unclear. The present study investigates whether the up-regulation of RAS and associated oxidative stress contributes to the development of endothelial dysfunction during estrogen deficiency in ovariectomized (OVX) rats. METHODOLOGY/PRINCIPAL FINDINGS: Adult female rats were ovariectomized with and without chronic treatment with valsartan and enalapril. Isometric force measurement was performed in isolated aortae. The expression of RAS components was determined by immunohistochemistry and Western blotting method while ROS accumulation in the vascular wall was evaluated by dihydroethidium fluorescence. Ovariectomy increased the expression of angiotensin-converting enzyme (ACE), angiotensin II type 1 receptor (AT(1)R), NAD(P)H oxidase, and nitrotyrosine in the rat aorta. An over-production of angiotensin II and ROS was accompanied by decreased phosphorylation of eNOS at Ser(1177) in OVX rat aortae. These pathophysiological changes were closely coupled with increased oxidative stress and decreased nitric oxide bioavailability, culminating in markedly impaired endothelium-dependent relaxations. Furthermore, endothelial dysfunction and increased oxidative stress in aortae of OVX rats were inhibited or reversed by chronic RAS inhibition with enalapril or valsartan. CONCLUSIONS/SIGNIFICANCE: The novel findings highlight a significant therapeutic benefit of RAS blockade in the treatment of endothelial dysfunction-related vascular complications in postmenopausal states.


Endothelium, Vascular/physiopathology , Estrogens/deficiency , Oxidative Stress , Renin-Angiotensin System/physiology , Acetylcholine/pharmacology , Angiotensin II/metabolism , Animals , Enalapril/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Estrogens/metabolism , Female , Free Radical Scavengers/metabolism , NADPH Oxidases/metabolism , Nitric Oxide/metabolism , Ovariectomy , Oxidative Stress/drug effects , Peptidyl-Dipeptidase A/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Renin-Angiotensin System/drug effects , Tetrazoles/pharmacology , Valine/analogs & derivatives , Valine/pharmacology , Valsartan , Vasodilation/drug effects
18.
Ann Neurol ; 69(1): 119-29, 2011 Jan.
Article En | MEDLINE | ID: mdl-21280082

OBJECTIVE: The sphingosine-1-phosphate (S1P) receptor agonist fingolimod (FTY720), that has shown efficacy in advanced multiple sclerosis clinical trials, decreases reperfusion injury in heart, liver, and kidney. We therefore tested the therapeutic effects of fingolimod in several rodent models of focal cerebral ischemia. To assess the translational significance of these findings, we asked whether fingolimod improved long-term behavioral outcomes, whether delayed treatment was still effective, and whether neuroprotection can be obtained in a second species. METHODS: We used rodent models of middle cerebral artery occlusion and cell-culture models of neurotoxicity and inflammation to examine the therapeutic potential and mechanisms of neuroprotection by fingolimod. RESULTS: In a transient mouse model, fingolimod reduced infarct size, neurological deficit, edema, and the number of dying cells in the core and periinfarct area. Neuroprotection was accompanied by decreased inflammation, as fingolimod-treated mice had fewer activated neutrophils, microglia/macrophages, and intercellular adhesion molecule-1 (ICAM-1)-positive blood vessels. Fingolimod-treated mice showed a smaller infarct and performed better in behavioral tests up to 15 days after ischemia. Reduced infarct was observed in a permanent model even when mice were treated 4 hours after ischemic onset. Fingolimod also decreased infarct size in a rat model of focal ischemia. Fingolimod did not protect primary neurons against glutamate excitotoxicity or hydrogen peroxide, but decreased ICAM-1 expression in brain endothelial cells stimulated by tumor necrosis factor alpha. INTERPRETATION: These findings suggest that anti-inflammatory mechanisms, and possibly vasculoprotection, rather than direct effects on neurons, underlie the beneficial effects of fingolimod after stroke. S1P receptors are a highly promising target in stroke treatment.


Brain Ischemia/drug therapy , Brain Ischemia/prevention & control , Immunosuppressive Agents/therapeutic use , Propylene Glycols/therapeutic use , Sphingosine/analogs & derivatives , Stroke/drug therapy , Animals , Apoptosis/drug effects , Disease Models, Animal , Fingolimod Hydrochloride , In Vitro Techniques , Inflammation/drug therapy , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/immunology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/prevention & control , Propylene Glycols/pharmacology , Rats , Receptors, Lysosphingolipid/agonists , Sphingosine/pharmacology , Sphingosine/therapeutic use , Stroke/prevention & control
19.
Arterioscler Thromb Vasc Biol ; 30(5): 992-9, 2010 May.
Article En | MEDLINE | ID: mdl-20185791

OBJECTIVE: Selective estrogen receptor modulators (SERMs) inhibit constriction of mammalian conduit arteries. However, it is unknown whether SERMs at therapeutically achievable concentrations could reduce vascular tone in resistance arteries. The present study aimed to examine roles of Ca(2+) influx in endothelium and endothelial nitric oxide synthase (eNOS) activation in dilatations induced by raloxifene, a second-generation SERM in myogenically active arteries. METHODS AND RESULTS: Small mesenteric arteries from Sprague-Dawley rats were isolated and mounted in a pressure myograph for measurement of changes in vessel diameter. [Ca(2+)](i) images on native endothelial cells of intact arteries were determined by the fluorescence imaging technique, and phosphorylation of eNOS was assayed by Western blotting. Raloxifene (0.3 to 10 nmol/L) produced dilatations on established steady myogenic constriction. Female rat arteries dilated significantly more in response to raloxifene than male arteries. Raloxifene-induced dilatations of female arteries were blunted by N(G)-nitro-l-arginine methyl ester but unaffected by 1400W, charybdotoxin plus apamin, wortmannin, or LY294002. Raloxifene (3 nmol/L) triggered rises in endothelial cell [Ca(2+)](i) and increased eNOS phosphorylation at Ser1177. Both effects were greater in arteries from female rats than in arteries from male rats. Increases in endothelial cell [Ca(2+)](i) and in eNOS phosphorylation were prevented by removal of extracellular Ca(2+) ions. Finally, ICI 182,780 did not affect the raloxifene-stimulated rise in endothelial cell [Ca(2+)](i), eNOS phosphorylation, and vasodilatations. Chronic raloxifene treatment reduced myogenic constriction in arteries from female but not male rats. CONCLUSION: Raloxifene at therapeutically relevant concentrations inhibits myogenic constriction by an NO-dependent mechanism that causally involves the elevated [Ca(2+)](i) in endothelial cells and subsequent eNOS activation. Raloxifene dilates resistance arteries more effectively in female rats, indicating its significant gender-related action on endothelial cells in microcirculation.


Calcium Signaling/drug effects , Mesenteric Arteries/drug effects , Nitric Oxide Synthase Type III/metabolism , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Androstadienes/pharmacology , Animals , Apamin/pharmacology , Blood Pressure/drug effects , Charybdotoxin/pharmacology , Chromones/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Fulvestrant , In Vitro Techniques , Male , Mesenteric Arteries/enzymology , Morpholines/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Ovariectomy , Phosphorylation , Potassium Channel Blockers/pharmacology , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Sex Factors , Time Factors , Vascular Resistance/drug effects , Wortmannin
20.
Sports Med ; 39(1): 45-63, 2009.
Article En | MEDLINE | ID: mdl-19093695

There is much evidence extolling the virtues of physical activity on cardiovascular disease (CVD). The evidence derives from different population groups where leisure time physical activity reduced the risk of coronary heart disease and cardiovascular mortality in both men and women. Recent meta-analyses have shown that large risk reductions for both ischaemic and haemorrhagic stroke can be achieved by moderate or intense physical activity. There are many data from human and animal studies confirming a beneficial role for exercise in the prevention and treatment of CVD. Physical inactivity and obesity/overweight are not only associated with a number of health-related risk factors, but are considered to be independent risk factors for CVD, type 2 diabetes mellitus and hypertension. Clinical trials confirm that lifestyle interventions (dietary modification and increased physical activity) reduce the risk of progressing from impaired glucose tolerance to type 2 diabetes. Moreover, epidemiological studies indicate that the risk of hypertension increases by being overweight. Modest increases in exercise intensity and frequency have hypotensive effects in sedentary hypertensive patients. Long-term training improves endothelium-dependent dilatation in the aorta and resistance arteries of the heart, whereas short-term training increases endothelial function in coronary conduit arteries. Overall, more scientific evidence will undoubtedly encourage the widespread advocacy of the clinical benefits of exercise therapy in the prevention and treatment of CVD.


Cardiovascular Diseases/prevention & control , Cardiovascular System , Endothelium, Vascular/physiology , Exercise/physiology , Motor Activity/physiology , Aging , Cardiovascular Diseases/physiopathology , Diabetes Mellitus/physiopathology , Diabetes Mellitus/prevention & control , Humans , Metabolic Syndrome/physiopathology , Risk Factors
...