Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Pulm Circ ; 10(3): 2045894020952019, 2020.
Article in English | MEDLINE | ID: mdl-32999709

ABSTRACT

Obesity is a prevalent global public health issue characterized by excess body fat. Adipose tissue is now recognized as an important endocrine organ releasing an abundance of bioactive adipokines including, but not limited to, leptin, adiponectin and resistin. Obesity is a common comorbidity amongst pulmonary arterial hypertension patients, with 30% to 40% reported as obese, independent of other comorbidities associated with pulmonary arterial hypertension (e.g. obstructive sleep apnoea). An 'obesity paradox' has been observed, where obesity has been associated with subclinical right ventricular dysfunction but paradoxically may confer a protective effect on right ventricular function once pulmonary hypertension develops. Obesity and pulmonary arterial hypertension share multiple pathophysiological mechanisms including inflammation, oxidative stress, elevated leptin (proinflammatory) and reduced adiponectin (anti-inflammatory). The female prevalence of pulmonary arterial hypertension has instigated the hypothesis that estrogens may play a causative role in its development. Adipose tissue, a major site for storage and metabolism of sex steroids, is the primary source of estrogens and circulating estrogens levels which are elevated in postmenopausal women and men with pulmonary arterial hypertension. This review discusses the functions of adipose tissue in both health and obesity and the links between obesity and pulmonary arterial hypertension. Shared pathophysiological mechanisms and the contribution of specific fat depots, metabolic and sex-dependent differences are discussed.

2.
Eur Respir J ; 53(6)2019 06.
Article in English | MEDLINE | ID: mdl-30923189

ABSTRACT

Obesity is a common comorbidity for pulmonary arterial hypertension (PAH). Additionally, oestrogen and its metabolites are risk factors for the development of PAH. Visceral adipose tissue (VAT) is a major site of oestrogen production; however, the influence of obesity-induced changes in oestrogen synthesis and metabolism on the development of PAH is unclear. To address this we investigated the effects of inhibiting oestrogen synthesis and metabolism on the development of pulmonary hypertension in male and female obese mice.We depleted endogenous oestrogen in leptin-deficient (ob/ob) mice with the oestrogen inhibitor anastrozole (ANA) and determined the effects on the development of pulmonary hypertension, plasma oestradiol and urinary 16α-hydroxyestrone (16αOHE1). Oestrogen metabolism through cytochrome P450 1B1 (CYP1B1) was inhibited with 2,2',4,6'-tetramethoxystilbene (TMS).ob/ob mice spontaneously develop pulmonary hypertension, pulmonary vascular remodelling and increased reactive oxygen species production in the lung; these effects were attenuated by ANA. Oestradiol levels were decreased in obese male mice; however, VAT CYP1B1 and 16αOHE1 levels were increased. TMS also attenuated pulmonary hypertension in male ob/ob mice. Intra-thoracic fat from ob/ob mice and VAT conditioned media produce 16αOHE1 and can contribute to oxidative stress, effects that are attenuated by both ANA and TMS.Obesity can induce pulmonary hypertension and changes in oestrogen metabolism, resulting in increased production of 16αOHE1 from VAT that contributes to oxidative stress. Oestrogen inhibitors are now in clinical trials for PAH. This study has translational consequences as it suggests that oestrogen inhibitors may be especially beneficial in treating obese individuals with PAH.


Subject(s)
Estrogens/metabolism , Intra-Abdominal Fat/metabolism , Obesity/metabolism , Pulmonary Arterial Hypertension/enzymology , Pulmonary Artery/enzymology , Anastrozole , Animals , Cytochrome P-450 CYP1B1/genetics , Estradiol/blood , Female , Hydroxyestrones/urine , Hypoxia/complications , Leptin/metabolism , Lung/enzymology , Lung/pathology , Male , Mice , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Obesity/genetics , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/pathology , Pulmonary Artery/pathology , Stilbenes
3.
Adv Exp Med Biol ; 1065: 511-528, 2018.
Article in English | MEDLINE | ID: mdl-30051404

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive disease characterised by increased pulmonary vascular resistance and pulmonary artery remodelling as result of increased vascular tone and vascular cell proliferation, respectively. Eventually, this leads to right heart failure. Heritable PAH is caused by a mutation in the bone morphogenetic protein receptor-II (BMPR-II). Female susceptibility to PAH has been known for some time, and most recent figures show a female-to-male ratio of 4:1. Variations in the female sex hormone estrogen and estrogen metabolism modify FPAH risk, and penetrance of the disease in BMPR-II mutation carriers is increased in females. Several lines of evidence point towards estrogen being pathogenic in the pulmonary circulation, and thus increasing the risk of females developing PAH. Recent studies have also suggested that estrogen metabolism may be crucial in the development and progression of PAH with studies indicating that downstream metabolites such as 16α-hydroxyestrone are upregulated in several forms of experimental pulmonary hypertension (PH) and can cause pulmonary artery smooth muscle cell proliferation and subsequent vascular remodelling. Conversely, other estrogen metabolites such as 2-methoxyestradiol have been shown to be protective in the context of PAH. Estrogen may also upregulate the signalling pathways of other key mediators of PAH such as serotonin.


Subject(s)
Arterial Pressure , Hypertension, Pulmonary/physiopathology , Pulmonary Artery/physiopathology , Vascular Remodeling , Age Factors , Animals , Antihypertensive Agents/therapeutic use , Arterial Pressure/drug effects , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Female , Genetic Predisposition to Disease , Gonadal Steroid Hormones/metabolism , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/epidemiology , Hypertension, Pulmonary/metabolism , Male , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Receptors, Estrogen/metabolism , Risk Factors , Sex Characteristics , Sex Factors , Signal Transduction , Treatment Outcome , Vascular Remodeling/drug effects
4.
Arterioscler Thromb Vasc Biol ; 37(7): 1361-1370, 2017 07.
Article in English | MEDLINE | ID: mdl-28473438

ABSTRACT

OBJECTIVE: Serotonin can induce human pulmonary artery smooth muscle cell (hPASMC) proliferation through reactive oxygen species (ROS), influencing the development of pulmonary arterial hypertension (PAH). We hypothesize that in PASMCs, serotonin induces oxidative stress through NADPH-oxidase-derived ROS generation and reduced Nrf-2 (nuclear factor [erythroid-derived 2]-like 2) antioxidant systems, promoting vascular injury. APPROACH AND RESULTS: HPASMCs from controls and PAH patients, and PASMCs from Nox1-/- mice, were stimulated with serotonin in the absence/presence of inhibitors of Src kinase, the 5-HT1B receptor, and NADPH oxidase 1 (Nox1). Markers of fibrosis were also determined. The pathophysiological significance of our findings was examined in vivo in serotonin transporter overexpressing female mice, a model of pulmonary hypertension. We confirmed thatserotonin increased superoxide and hydrogen peroxide production in these cells. For the first time, we show that serotonin increased oxidized protein tyrosine phosphatases and hyperoxidized peroxiredoxin and decreased Nrf-2 and catalase activity in hPASMCs. ROS generation was exaggerated and dependent on cellular Src-related kinase, 5-HT1B receptor, and the serotonin transporter in human pulmonary artery smooth muscle cells from PAH subjects. Proliferation and extracellular matrix remodeling were exaggerated in human pulmonary artery smooth muscle cells from PAH subjects and dependent on 5-HT1B receptor signaling and Nox1, confirmed in PASMCs from Nox1-/- mice. In serotonin transporter overexpressing mice, SB216641, a 5-HT1B receptor antagonist, prevented development of pulmonary hypertension in a ROS-dependent manner. CONCLUSIONS: Serotonin can induce cellular Src-related kinase-regulated Nox1-induced ROS and Nrf-2 dysregulation, contributing to increased post-translational oxidative modification of proteins and activation of redox-sensitive signaling pathways in hPASMCs, associated with mitogenic responses. 5-HT1B receptors contribute to experimental pulmonary hypertension by inducing lung ROS production. Our results suggest that 5-HT1B receptor-dependent cellular Src-related kinase-Nox1-pathways contribute to vascular remodeling in PAH.


Subject(s)
Arterial Pressure , Hypertension, Pulmonary/enzymology , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidases/metabolism , Pulmonary Artery/enzymology , Receptor, Serotonin, 5-HT1B/metabolism , Serotonin/metabolism , Adult , Aged , Animals , Antihypertensive Agents/pharmacology , Arterial Pressure/drug effects , Case-Control Studies , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Female , Genetic Predisposition to Disease , Humans , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/prevention & control , Hypoxia/complications , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/deficiency , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , NADPH Oxidases/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Peroxiredoxins/metabolism , Phenotype , Protein Carbonylation , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Reactive Oxygen Species/metabolism , Receptor, Serotonin, 5-HT1B/drug effects , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Signal Transduction , Time Factors , Vascular Remodeling , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
5.
Am J Respir Crit Care Med ; 191(12): 1432-42, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25871906

ABSTRACT

RATIONALE: Females are predisposed to pulmonary arterial hypertension (PAH); evidence suggests that serotonin, mutations in the bone morphogenetic protein receptor (BMPR) II gene, and estrogens influence development of PAH. The 5-hydroxytryptamine 1B receptor (5-HT1BR) mediates human pulmonary artery smooth muscle cell (hPASMC) proliferation. OBJECTIVES: We aimed to determine whether selected microRNAs (miRNAs) expressed in PASMCs are influenced by sex, BMPR-II mutations, and estrogens, and contribute to PASMC proliferation in PAH. METHODS: Expression levels of miRNAs targeting genes related to PAH, estrogen, and serotonin were determined by quantitative RT-PCR in hPASMCs and mouse PASMCs harboring a heterozygous mutation in BMPR-II (BMPR-II(R899X+/-) PASMCs). miRNA-96 targets 5-HT1BR and was selected for further investigation. miRNA target validation was confirmed by luciferase reporter assay. Precursor miRNA-96 was transfected into hPASMCs to examine effects on proliferation and 5-HT1BR expression. The effect of a miRNA-96 mimic on the development of hypoxic pulmonary hypertension in mice was also assessed. MEASUREMENTS AND MAIN RESULTS: miRNA-96 expression was reduced in BMPR-II(R899X+/-) PASMCs from female mice and hPASMCs from female patients with PAH; this was associated with increased 5-HT1BR expression and serotonin-mediated proliferation. 5-HT1BR was validated as a target for miRNA-96. Transfection of precursor miRNA-96 into hPASMCs reduced 5-HT1BR expression and inhibited serotonin-induced proliferation. Restoration of miRNA-96 expression in pulmonary arteries in vivo via administration of an miRNA-96 mimic reduced the development of hypoxia-induced pulmonary hypertension in the mouse. CONCLUSIONS: Increased 5-HT1BR expression may be a consequence of decreased miRNA-96 expression in female patient PASMCs, and this may contribute to the development of PAH.


Subject(s)
Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , MicroRNAs/metabolism , Receptor, Serotonin, 5-HT1B/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type II/metabolism , Cell Proliferation/physiology , Cells, Cultured , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , Real-Time Polymerase Chain Reaction , Sex Characteristics , Signal Transduction/physiology
6.
Am J Respir Crit Care Med ; 191(6): 693-703, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25608111

ABSTRACT

RATIONALE: Major pulmonary arterial hypertension (PAH) registries report a greater incidence of PAH in women; mutations in the bone morphogenic protein type II receptor (BMPR-II) occur in approximately 80% of patients with heritable PAH (hPAH). OBJECTIVES: We addressed the hypothesis that women may be predisposed to PAH due to normally reduced basal BMPR-II signaling in human pulmonary artery smooth muscle cells (hPASMCs). METHODS: We examined the BMPR-II signaling pathway in hPASMCs derived from men and women with no underlying cardiovascular disease (non-PAH hPASMCs). We also determined the development of pulmonary hypertension in male and female mice deficient in Smad1. MEASUREMENTS AND MAIN RESULTS: Platelet-derived growth factor, estrogen, and serotonin induced proliferation only in non-PAH female hPASMCs. Female non-PAH hPASMCs exhibited reduced messenger RNA and protein expression of BMPR-II, the signaling intermediary Smad1, and the downstream genes, inhibitors of DNA binding proteins, Id1 and Id3. Induction of phospho-Smad1/5/8 and Id protein by BMP4 was also reduced in female hPASMCs. BMP4 induced proliferation in female, but not male, hPASMCs. However, small interfering RNA silencing of Smad1 invoked proliferative responses to BMP4 in male hPASMCs. In male hPASMCs, estrogen decreased messenger RNA and protein expression of Id genes. The estrogen metabolite 4-hydroxyestradiol decreased phospho-Smad1/5/8 and Id expression in female hPASMCs while increasing these in males commensurate with a decreased proliferative effect in male hPASMCs. Female Smad1(+/-) mice developed pulmonary hypertension (reversed by ovariectomy). CONCLUSIONS: We conclude that estrogen-driven suppression of BMPR-II signaling in non-PAH hPASMCs derived from women contributes to a pro-proliferative phenotype in hPASMCs that may predispose women to PAH.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/physiology , Muscle, Smooth, Vascular/cytology , Pulmonary Artery/cytology , Animals , Down-Regulation , Estrogens/metabolism , Estrogens/physiology , Female , Humans , Hypertension, Pulmonary , Male , Mice , Sex Factors , Signal Transduction/physiology
7.
Am J Respir Crit Care Med ; 190(4): 456-67, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24956156

ABSTRACT

RATIONALE: The incidence of pulmonary arterial hypertension is greater in women, suggesting estrogens may play a role in the disease pathogenesis. Experimentally, in males, exogenously administered estrogen can protect against pulmonary hypertension (PH). However, in models that display female susceptibility, estrogens may play a causative role. OBJECTIVES: To clarify the influence of endogenous estrogen and sex in PH and assess the therapeutic potential of a clinically available aromatase inhibitor. METHODS: We interrogated the effect of reduced endogenous estrogen in males and females using the aromatase inhibitor, anastrozole, in two models of PH: the hypoxic mouse and Sugen 5416/hypoxic rat. We also determined the effects of sex on pulmonary expression of aromatase in these models and in lungs from patients with pulmonary arterial hypertension. MEASUREMENTS AND MAIN RESULTS: Anastrozole attenuated PH in both models studied, but only in females. To verify this effect was caused by reduced estrogenic activity we confirmed that in hypoxic mice inhibition of estrogen receptor α also has a therapeutic effect specifically in females. Female rodent lung displays increased aromatase and decreased bone morphogenetic protein receptor 2 and Id1 expression compared with male. Anastrozole treatment reversed the impaired bone morphogenetic protein receptor 2 pathway in females. Increased aromatase expression was also detected in female human pulmonary artery smooth muscle cells compared with male. CONCLUSIONS: The unique phenotype of female pulmonary arteries facilitates the therapeutic effects of anastrozole in experimental PH confirming a role for endogenous estrogen in the disease pathogenesis in females and suggests aromatase inhibitors may have therapeutic potential.


Subject(s)
Estrogens/blood , Hypertension, Pulmonary/blood , Anastrozole , Animals , Aromatase Inhibitors/blood , Aromatase Inhibitors/pharmacology , Blotting, Western/methods , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Hypertension, Pulmonary/complications , Hypoxia/blood , Hypoxia/complications , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Nitriles/blood , Nitriles/pharmacology , Polymerase Chain Reaction/methods , Rats , Rats, Wistar , Sex Factors , Triazoles/blood , Triazoles/pharmacology
8.
Pulm Pharmacol Ther ; 29(1): 7-14, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24417910

ABSTRACT

The calgranulin-like protein MTS1/S100A4 and the receptor for advanced glycation end-products (RAGE) have recently been implicated in mediating pulmonary arterial smooth muscle cell proliferation and vascular remodelling in experimental pulmonary arterial hypertension (PH). Here, the effects of RAGE antagonism upon 2 weeks of hypobaric hypoxia (10% O2)-induced PH in mice were assessed. Treatment with sRAGE was protective against hypobaric hypoxia-induced increases in right ventricular pressure but distal pulmonary vascular remodelling was unaffected. Intralobar pulmonary arteries from hypobaric hypoxic mice treated with sRAGE showed protection against a hypoxia-induced reduction in compliance. However, a combination of sRAGE and hypoxia also dramatically increased the force of contractions to KCl and 5-HT observed in these vessels. The acute addition of sRAGE to the organ bath produced a small, sustained contraction in intralobar pulmonary vessels and produced a synergistic enhancement of the maximal force of contraction in subsequent concentration-response curves to 5-HT. sRAGE had no effect on 5-HT-induced proliferation of Chinese hamster lung fibroblasts (CCL39), used since they have a similar pharmacological profile to mouse pulmonary fibroblasts but, surprisingly, produced a marked increase in hypoxia-induced proliferation. These data implicate RAGE as a modulator of both vasoreactivity and of proliferative processes in the response of the pulmonary circulation to chronic-hypoxia.


Subject(s)
Fibroblasts/metabolism , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Receptor for Advanced Glycation End Products/metabolism , Animals , Cell Line , Cell Proliferation/physiology , Cricetinae , Cricetulus , Disease Models, Animal , Hemodynamics , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/physiology , Pulmonary Artery/metabolism , Receptor for Advanced Glycation End Products/administration & dosage , Serotonin/administration & dosage , Serotonin/metabolism , Vascular Remodeling/physiology
9.
Cardiovasc Res ; 99(1): 24-34, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23519266

ABSTRACT

AIMS: Pulmonary arterial hypertension (PAH) occurs more frequently in women than men. Oestrogen and the oestrogen-metabolising enzyme cytochrome P450 1B1 (CYP1B1) play a role in the development of PAH. Anorectic drugs such as dexfenfluramine (Dfen) have been associated with the development of PAH. Dfen mediates PAH via a serotonergic mechanism and we have shown serotonin to up-regulate expression of CYP1B1 in human pulmonary artery smooth muscle cells (PASMCs). Thus here we assess the role of CYP1B1 in the development of Dfen-induced PAH. METHODS AND RESULTS: Dfen (5 mg kg(-1) day(-1) PO for 28 days) increased right ventricular pressure and pulmonary vascular remodelling in female mice only. Mice dosed with Dfen showed increased whole lung expression of CYP1B1 and Dfen-induced PAH was ablated in CYP1B1(-/-) mice. In line with this, Dfen up-regulated expression of CYP1B1 in PASMCs from PAH patients (PAH-PASMCs) and Dfen-mediated proliferation of PAH-PASMCs was ablated by pharmacological inhibition of CYP1B1. Dfen increased expression of tryptophan hydroxylase 1 (Tph1; the rate-limiting enzyme in the synthesis of serotonin) in PAH-PASMCs and both Dfen-induced proliferation and Dfen-induced up-regulation of CYP1B1 were ablated by inhibition of Tph1. 17ß-Oestradiol increased expression of both Tph1 and CYP1B1 in PAH-PASMCs, and Dfen and 17ß-oestradiol had synergistic effects on proliferation of PAH-PASMCs. Finally, ovariectomy protected against Dfen-induced PAH in female mice. CONCLUSION: CYP1B1 is critical in the development of Dfen-induced PAH in mice in vivo and proliferation of PAH-PASMCs in vitro. CYP1B1 may provide a novel therapeutic target for PAH.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Dexfenfluramine , Hypertension, Pulmonary/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Animals , Arterial Pressure , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/deficiency , Aryl Hydrocarbon Hydroxylases/genetics , Cell Proliferation , Cells, Cultured , Cytochrome P-450 CYP1B1 , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Estradiol/pharmacology , Familial Primary Pulmonary Hypertension , Female , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Norfenfluramine/toxicity , Ovariectomy , Pulmonary Artery/enzymology , Pulmonary Artery/physiopathology , Serotonin/metabolism , Sex Factors , Tryptophan Hydroxylase/metabolism , Ventricular Function, Right , Ventricular Pressure
10.
Circulation ; 126(9): 1087-98, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22859684

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension (PAH) is a hyperproliferative vascular disorder observed predominantly in women. Estrogen is a potent mitogen in human pulmonary artery smooth muscle cells and contributes to PAH in vivo; however, the mechanisms attributed to this causation remain obscure. Curiously, heightened expression of the estrogen-metabolizing enzyme cytochrome P450 1B1 (CYP1B1) is reported in idiopathic PAH and murine models of PAH. METHODS AND RESULTS: Here, we investigated the putative pathogenic role of CYP1B1 in PAH. Quantitative reverse transcription-polymerase chain reaction, immunoblotting, and in situ analysis revealed that pulmonary CYP1B1 is increased in hypoxic PAH, hypoxic+SU5416 PAH, and human PAH and is highly expressed within the pulmonary vascular wall. PAH was assessed in mice via measurement of right ventricular hypertrophy, pulmonary vascular remodeling, and right ventricular systolic pressure. Hypoxic PAH was attenuated in CYP1B1(-/-) mice, and the potent CYP1B1 inhibitor 2,3',4,5'-tetramethoxystilbene (TMS; 3 mg · kg(-1) · d(-1) IP) significantly attenuated hypoxic PAH and hypoxic+SU5416 PAH in vivo. TMS also abolished estrogen-induced proliferation in human pulmonary artery smooth muscle cells and PAH-pulmonary artery smooth muscle cells. The estrogen metabolite 16α-hydroxyestrone provoked human pulmonary artery smooth muscle cell proliferation, and this mitogenic effect was greatly pronounced in PAH-pulmonary artery smooth muscle cells. ELISA analysis revealed that 16α-hydroxyestrone concentration was elevated in PAH, consistent with CYP1B1 overexpression and activity. Finally, administration of the CYP1B1 metabolite 16α-hydroxyestrone (1.5 mg · kg(-1) · d(-1) IP) caused the development of PAH in mice. CONCLUSIONS: Increased CYP1B1-mediated estrogen metabolism promotes the development of PAH, likely via the formation of mitogens, including 16α-hydroxyestrone. Collectively, this study reveals a possible novel therapeutic target in clinical PAH.


Subject(s)
Aryl Hydrocarbon Hydroxylases/physiology , Estrogens/metabolism , Hypertension, Pulmonary/enzymology , Pulmonary Artery/enzymology , Animals , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/biosynthesis , Aryl Hydrocarbon Hydroxylases/deficiency , Aryl Hydrocarbon Hydroxylases/genetics , Cell Hypoxia , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chronic Disease , Cytochrome P-450 CYP1B1 , Enzyme Induction , Estradiol/pharmacology , Female , Humans , Hydroxyestrones/metabolism , Hydroxyestrones/pharmacology , Hydroxyestrones/toxicity , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/enzymology , Hypoxia/complications , Lung/enzymology , Lung/pathology , Male , Mice , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/pathology , Reverse Transcriptase Polymerase Chain Reaction , Stilbenes/pharmacology , Up-Regulation
11.
Circ Res ; 111(3): 290-300, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22715469

ABSTRACT

RATIONALE: Despite improved understanding of the underlying genetics, pulmonary arterial hypertension (PAH) remains a severe disease. Extensive remodeling of small pulmonary arteries, including proliferation of pulmonary artery smooth muscle cells (PASMCs), characterizes PAH. MicroRNAs (miRNAs) are noncoding RNAs that have been shown to play a role in vascular remodeling. OBJECTIVE: We assessed the role of miR-145 in PAH. METHODS AND RESULTS: We localized miR-145 in mouse lung to smooth muscle. Using quantitative PCR, we demonstrated increased expression of miR-145 in wild-type mice exposed to hypoxia. PAH was evaluated in miR-145 knockout and mice treated with anti-miRs via measurement of systolic right ventricular pressure, right ventricular hypertrophy, and percentage of remodeled pulmonary arteries. miR-145 deficiency and anti-miR-mediated reduction resulted in significant protection from the development of PAH. In contrast, miR-143 anti-miR had no effect. Furthermore, we observed upregulation of miR-145 in lung tissue of patients with idiopathic and heritable PAH compared with unaffected control subjects and demonstrated expression of miR-145 in SMC of remodeled vessels from such patients. Finally, we show elevated levels of miR-145 expression in primary PASMCs cultured from patients with BMPR2 mutations and also in the lungs of BMPR2-deficient mice. CONCLUSIONS: miR-145 is dysregulated in mouse models of PAH. Downregulation of miR-145 protects against the development of PAH. In patient samples of heritable PAH and idiopathic PAH, miR-145 is expressed in remodeled vessels and mutations in BMPR2 lead to upregulation of miR-145 in mice and PAH patients. Manipulation of miR-145 may represent a novel strategy in PAH treatment.


Subject(s)
Disease Models, Animal , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , MicroRNAs/physiology , Animals , Down-Regulation/genetics , Female , Gene Knock-In Techniques , Humans , Hypertension, Pulmonary/prevention & control , Lung/pathology , Lung/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics
12.
Respir Res ; 12: 159, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22185646

ABSTRACT

BACKGROUND: Idiopathic and familial forms of pulmonary arterial hypertension (PAH) occur more frequently in women than men. However, the reason for this remains unknown. Both the calcium binding protein S100A4/Mts1 (Mts1) and its endogenous receptor (receptor for advanced glycosylation end products; RAGE) have been implicated in the development of PAH. We wished to investigate if the Mts1/RAGE pathway may play a role in the gender bias associated with PAH. METHODS: We investigated the effects of gender on development of PAH in mice over-expressing Mts1 (Mts1+ mice) via measurement of pulmonary arterial remodeling, systolic right ventricular pressure (sRVP) and right ventricular hypertrophy (RVH). Gender differences in pulmonary arterial Mts1 and RAGE expression were assessed by qRT-PCR and immunohistochemistry. Western blotting and cell counts were used to investigate interactions between 17ß-estradiol, Mts1 and RAGE on proliferation of human pulmonary artery smooth muscle cells (hPASMCs). Statistical analysis was by one-way analysis of variance with Dunnetts post test or two-way analysis of variance with Bonferronis post test, as appropriate. RESULTS: Female Mts1+ mice developed increased sRVP and pulmonary vascular remodeling, whereas male Mts1+ mice remained unaffected. The development of plexiform-like lesions in Mts1+ mice was specific to females. These lesions stained positive for both Mts1 and RAGE in the endothelial and adventitial layers. Expression of pulmonary arterial Mts1 was greater in female than male Mts1+ mice, and was localised to the medial and adventitial layers in non plexiform-like pulmonary arteries. RAGE gene expression and immunoreactivity were similar between male and female Mts1+ mice and RAGE staining was localised to the endothelial layer in non plexiform-like pulmonary arteries adjacent to airways. In non-plexiform like pulmonary arteries not associated with airways RAGE staining was present in the medial and adventitial layers. Physiological concentrations of 17ß-estradiol increased Mts1 expression in hPASMCs. 17ß-estradiol-induced hPASMC proliferation was inhibited by soluble RAGE, which antagonises the membrane bound form of RAGE. CONCLUSIONS: Mts1 over-expression combined with female gender is permissive to the development of experimental PAH in mice. Up-regulation of Mts1 and subsequent activation of RAGE may contribute to 17ß-estradiol-induced proliferation of hPASMCs.


Subject(s)
Blood Pressure , Hypertension, Pulmonary/metabolism , Pulmonary Artery/metabolism , S100 Proteins/metabolism , Analysis of Variance , Animals , Blood Pressure/genetics , Blotting, Western , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Estradiol/metabolism , Familial Primary Pulmonary Hypertension , Female , Genetic Predisposition to Disease , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Real-Time Polymerase Chain Reaction , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , S100 Calcium-Binding Protein A4 , S100 Proteins/genetics , Sex Factors , Signal Transduction , Up-Regulation , Ventricular Function, Right , Ventricular Pressure
13.
Mol Cell Biol ; 30(15): 3827-41, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20516217

ABSTRACT

We demonstrate here a new concept termed "oncogene tolerance" whereby human EGF receptor 2 (HER2) increases sphingosine kinase 1 (SK1) expression in estrogen receptor-positive (ER(+)) MCF-7 HER2 cells and SK1, in turn, limits HER2 expression in a negative-feedback manner. The HER2-dependent increase in SK1 expression also limits p21-activated protein kinase 1 (p65 PAK1) and extracellular signal regulated kinase 1/2 (ERK-1/2) signaling. Sphingosine 1-phosphate signaling via S1P(3) is also altered in MCF-7 HER2 cells. In this regard, S1P binding to S1P(3) induces a migratory phenotype via an SK1-dependent mechanism in ER(+) MCF-7 Neo cells, which lack HER2. This involves the S1P stimulated accumulation of phosphorylated ERK-1/2 and actin into membrane ruffles/lamellipodia and migration. In contrast, S1P failed to promote redistribution of phosphorylated ERK-1/2 and actin into membrane ruffles/lamellipodia or migration of MCF-7 HER2 cells. However, a migratory phenotype in these cells could be induced in response to S1P when SK1 expression had been knocked down with a specific siRNA or when recombinant PAK1 was ectopically overexpressed. Thus, the HER2-dependent increase in SK1 expression functions to desensitize the S1P-induced formation of a migratory phenotype. This is correlated with improved prognosis in patients who have a low HER1-3/SK1 expression ratio in their ER(+) breast cancer tumors compared to patients that have a high HER1-3/SK1 expression ratio.


Subject(s)
Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Receptors, Estrogen/metabolism , Breast Neoplasms/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cell Line, Tumor , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/genetics , Estrogen Receptor alpha , Humans , Lysophospholipids , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/pharmacology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/pharmacology , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor) , Protein Binding/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Receptors, Estrogen/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingosine/analogs & derivatives , p21-Activated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...