PPAR{gamma} regulates hypoxia-induced Nox4 expression in human pulmonary artery smooth muscle cells through NF-{kappa}B.

NADPH oxidases are a major source of superoxide production in the vasculature. The constitutively active Nox4 subunit, which is selectively upregulated in the lungs of human subjects and experimental animals with pulmonary hypertension, is highly expressed in vascular wall cells. We demonstrated that rosiglitazone, a synthetic agonist of the peroxisome proliferator-activated receptor-γ (PPARγ), attenuated hypoxia-induced pulmonary hypertension, vascular remodeling, Nox4 induction, and reactive oxygen species generation in the mouse lung. The current study examined the molecular mechanisms involved in PPARγ-regulated, hypoxia-induced Nox4 expression in human pulmonary artery smooth muscle cells (HPASMC). Exposing HPASMC to 1% oxygen for 72 h increased Nox4 gene expression and H(2)O(2) production, both of which were reduced by treatment with rosiglitazone during the last 24 h of hypoxia exposure or by treatment with small interfering RNA (siRNA) to Nox4. Hypoxia also increased HPASMC proliferation as well as the activity of a Nox4 promoter luciferase reporter, and these increases were attenuated by rosiglitazone. Chromatin immunoprecipitation assays demonstrated that hypoxia increased binding of the NF-κB subunit, p65, to the Nox4 promoter and that binding was attenuated by rosiglitazone treatment. The role of NF-κB in Nox4 regulation was further supported by demonstrating that overexpression of p65 stimulated Nox4 promoter activity, whereas siRNA to p50 or p65 attenuated hypoxic stimulation of Nox4 promoter activity. These results provide novel evidence for NF-κB-mediated stimulation of Nox4 expression in HPASMC that can be negatively regulated by PPARγ. These data provide new insights into potential mechanisms by which PPARγ activation inhibits Nox4 upregulation and the proliferation of cells in the pulmonary vascular wall to ameliorate pulmonary hypertension and vascular remodeling in response to hypoxia.

[1]  C. Hart,et al.  Rosiglitazone attenuates chronic hypoxia-induced pulmonary hypertension in a mouse model. , 2010, American journal of respiratory cell and molecular biology.

[2]  G. Laurent,et al.  The quest for the initial lesion in idiopathic pulmonary fibrosis: gene expression differences in IPF fibroblasts. , 2010, American journal of respiratory cell and molecular biology.

[3]  J. Ritzenthaler,et al.  Disruption of endothelial peroxisome proliferator-activated receptor-gamma reduces vascular nitric oxide production. , 2009, American journal of physiology. Heart and circulatory physiology.

[4]  L. Hofbauer,et al.  Long-term cyclic strain downregulates endothelial Nox4. , 2009, Antioxidants & redox signaling.

[5]  V. Demarco,et al.  Rosuvastatin ameliorates the development of pulmonary arterial hypertension in the transgenic (mRen2)27 rat. , 2009, American journal of physiology. Heart and circulatory physiology.

[6]  M. Aschner,et al.  NADPH oxidases and reactive oxygen species at different stages of chronic hypoxia-induced pulmonary hypertension in newborn piglets. , 2009, American journal of physiology. Lung cellular and molecular physiology.

[7]  Dong-Hoon Shin,et al.  Inhibitor of nuclear factor‐kappaB alpha derepresses hypoxia‐inducible factor‐1 during moderate hypoxia by sequestering factor inhibiting hypoxia‐inducible factor from hypoxia‐inducible factor 1α , 2009, The FEBS journal.

[8]  G. Hansmann,et al.  Tie2-mediated loss of peroxisome proliferator-activated receptor- (cid:1) in mice causes PDGF receptor- (cid:2) -dependent pulmonary arterial muscularization , 2009 .

[9]  A. Shah,et al.  Positive regulation of the NADPH oxidase NOX4 promoter in vascular smooth muscle cells by E2F. , 2008, Free radical biology & medicine.

[10]  W. Seeger,et al.  NOX4 regulates ROS levels under normoxic and hypoxic conditions, triggers proliferation, and inhibits apoptosis in pulmonary artery adventitial fibroblasts. , 2008, Antioxidants & redox signaling.

[11]  A. Rahimipour,et al.  Docosahexaenoic acid sensitizes Ramos cells to Gamma-irradiation-induced apoptosis through involvement of PPAR-γ activation and NF-κB suppression , 2008, Molecular and Cellular Biochemistry.

[12]  G. Hansmann,et al.  An antiproliferative BMP-2/PPARgamma/apoE axis in human and murine SMCs and its role in pulmonary hypertension. , 2008, The Journal of clinical investigation.

[13]  R. Mortensen,et al.  Peroxisome Proliferator-Activated Receptor-γ–Mediated Effects in the Vasculature , 2008, Circulation research.

[14]  L. Kennedy Pioglitazone and Risk of Cardiovascular Events in Patients With Type 2 Diabetes Mellitus: A Meta-analysis of Randomized Trials , 2008 .

[15]  W. Farrar,et al.  Inhibition of adhesive interaction between multiple myeloma and bone marrow stromal cells by PPARgamma cross talk with NF-kappaB and C/EBP. , 2007, Blood.

[16]  F. DeLeo,et al.  Role of NF‐κB in transcriptional regulation of the phagocyte NADPH oxidase by tumor necrosis factor‐α , 2007 .

[17]  C. Rosen The rosiglitazone story--lessons from an FDA Advisory Committee meeting. , 2007, The New England journal of medicine.

[18]  K. Krause,et al.  NOX4 activity is determined by mRNA levels and reveals a unique pattern of ROS generation. , 2007, The Biochemical journal.

[19]  W. Seeger,et al.  Hypoxia-Dependent Regulation of Nonphagocytic NADPH Oxidase Subunit NOX4 in the Pulmonary Vasculature , 2007, Circulation research.

[20]  Seung‐Jung Park,et al.  FcgammaRIIa mediates C-reactive protein-induced inflammatory responses of human vascular smooth muscle cells by activating NADPH oxidase 4. , 2007, Cardiovascular Research.

[21]  T. Kawahara,et al.  Regulation of Nox and Duox enzymatic activity and expression. , 2007, Free radical biology & medicine.

[22]  S. Nissen,et al.  Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. , 2007, The New England journal of medicine.

[23]  P. Thulé,et al.  The PPARgamma ligand, rosiglitazone, reduces vascular oxidative stress and NADPH oxidase expression in diabetic mice. , 2007, Vascular pharmacology.

[24]  J. Hoidal,et al.  Nox4 mediates TGF-beta1-induced retinoblastoma protein phosphorylation, proliferation, and hypertrophy in human airway smooth muscle cells. , 2007, American journal of physiology. Lung cellular and molecular physiology.

[25]  B. Walker,et al.  Effects of peroxisome proliferator-activated receptor-alpha and -gamma agonists on 11beta-hydroxysteroid dehydrogenase type 1 in subcutaneous adipose tissue in men. , 2007, The Journal of clinical endocrinology and metabolism.

[26]  Weiling Zhao,et al.  Knocking Out Peroxisome Proliferator-Activated Receptor (PPAR) α Inhibits Radiation-Induced Apoptosis in the Mouse Kidney through Activation of NF-κB and Increased Expression of IAPs , 2007, Radiation research.

[27]  A. Skene,et al.  The effect of pioglitazone on recurrent myocardial infarction in 2,445 patients with type 2 diabetes and previous myocardial infarction: results from the PROactive (PROactive 05) Study. , 2007, Journal of the American College of Cardiology.

[28]  K. Stenmark,et al.  Rosiglitazone attenuates hypoxia-induced pulmonary arterial remodeling. , 2007, American journal of physiology. Lung cellular and molecular physiology.

[29]  T. Kietzmann,et al.  Reactive Oxygen Species Activate the HIF-1&agr; Promoter Via a Functional NF&kgr;B Site , 2007 .

[30]  Ahmad Y. Sheikh,et al.  Pulmonary Arterial Hypertension Is Linked to Insulin Resistance and Reversed by Peroxisome Proliferator–Activated Receptor-&ggr; Activation , 2007 .

[31]  D. Betteridge,et al.  Effects of Pioglitazone in Patients With Type 2 Diabetes With or Without Previous Stroke: Results From PROactive (PROspective pioglitAzone Clinical Trial In macroVascular Events 04) , 2007, Stroke.

[32]  T. Willson,et al.  Parallel SUMOylation-dependent pathways mediate gene- and signal-specific transrepression by LXRs and PPARgamma. , 2007, Molecular cell.

[33]  A. Manea,et al.  Regulation of NADPH oxidase subunit p22phox by NF-kB in human aortic smooth muscle cells , 2007, Archives of physiology and biochemistry.

[34]  F. DeLeo,et al.  Role of NF-kappaB in transcriptional regulation of the phagocyte NADPH oxidase by tumor necrosis factor-alpha. , 2007, Journal of leukocyte biology.

[35]  T. Kietzmann,et al.  Reactive oxygen species activate the HIF-1alpha promoter via a functional NFkappaB site. , 2007, Arteriosclerosis, thrombosis, and vascular biology.

[36]  Ahmad Y. Sheikh,et al.  Pulmonary arterial hypertension is linked to insulin resistance and reversed by peroxisome proliferator-activated receptor-gamma activation. , 2007, Circulation.

[37]  K. Krause,et al.  The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. , 2007, Physiological reviews.

[38]  B. Spiegelman,et al.  International Union of Pharmacology. LXI. Peroxisome Proliferator-Activated Receptors , 2006, Pharmacological Reviews.

[39]  Chuanshu Huang,et al.  Participation of the PI-3K/Akt-NF-κB signaling pathways in hypoxia-induced mitogenic factor-stimulated Flk-1 expression in endothelial cells , 2006, Respiratory research.

[40]  F. Giancotti,et al.  Faculty Opinions recommendation of Cyld inhibits tumor cell proliferation by blocking Bcl-3-dependent NF-kappaB signaling. , 2006 .

[41]  R. Brandes,et al.  Role of reactive oxygen species and gp91phox in endothelial dysfunction of pulmonary arteries induced by chronic hypoxia , 2006, British journal of pharmacology.

[42]  S. Black,et al.  Increased oxidative stress in lambs with increased pulmonary blood flow and pulmonary hypertension: role of NADPH oxidase and endothelial NO synthase. , 2006, American journal of physiology. Lung cellular and molecular physiology.

[43]  Ramin Massoumi,et al.  Cyld Inhibits Tumor Cell Proliferation by Blocking Bcl-3-Dependent NF-κB Signaling , 2006, Cell.

[44]  W. Seeger,et al.  Impact of mitochondria and NADPH oxidases on acute and sustained hypoxic pulmonary vasoconstriction. , 2006, American journal of respiratory cell and molecular biology.

[45]  C. Iadecola,et al.  NF-κB Regulates Phagocytic NADPH Oxidase by Inducing the Expression of gp91phox* , 2006, Journal of Biological Chemistry.

[46]  C. Iadecola,et al.  NF-kappaB regulates phagocytic NADPH oxidase by inducing the expression of gp91phox. , 2006, The Journal of biological chemistry.

[47]  R. Folz,et al.  Hypoxic pulmonary hypertension: role of superoxide and NADPH oxidase (gp91phox). , 2006, American journal of physiology. Lung cellular and molecular physiology.

[48]  Erland Erdmann,et al.  Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial , 2005, The Lancet.

[49]  Satoshi Suzuki,et al.  [Effects of peroxisome proliferator-activated receptor gamma ligands on monocrotaline-induced pulmonary hypertension in rats]. , 2005, Nihon Kokyuki Gakkai zasshi = the journal of the Japanese Respiratory Society.

[50]  K. Griendling,et al.  Peroxisome proliferator-activated receptor-gamma ligands regulate endothelial membrane superoxide production. , 2005, American journal of physiology. Cell physiology.

[51]  G. Angelini,et al.  Acute hypoxia simultaneously induces the expression of gp91phox and endothelial nitric oxide synthase in the porcine pulmonary artery , 2005, Thorax.

[52]  Pravir Kumar,et al.  Direct Interaction of the Novel Nox Proteins with p22phox Is Required for the Formation of a Functionally Active NADPH Oxidase* , 2004, Journal of Biological Chemistry.

[53]  Manu O. Platt,et al.  Bone Morphogenic Protein 4 Produced in Endothelial Cells by Oscillatory Shear Stress Induces Monocyte Adhesion by Stimulating Reactive Oxygen Species Production From a Nox1-Based NADPH Oxidase , 2004, Circulation research.

[54]  J. Rubin,et al.  Integration of the NfκB p65 subunit into the vitamin D receptor transcriptional complex: Identification of p65 domains that inhibit 1,25‐dihydroxyvitamin D3‐stimulated transcription , 2004, Journal of cellular biochemistry.

[55]  Darrell R. Abernethy,et al.  International Union of Pharmacology: Approaches to the Nomenclature of Voltage-Gated Ion Channels , 2003, Pharmacological Reviews.

[56]  L. Harrison,et al.  Phosphorylation of PPARγ via active ERK1/2 leads to its physical association with p65 and inhibition of NF‐κβ , 2003 .

[57]  Karim Benkirane,et al.  Peroxisome proliferator-activated receptors: vascular and cardiac effects in hypertension. , 2003, Hypertension.

[58]  M. Wick,et al.  Peroxisome Proliferator-Activated Receptor Gamma (PPAR&ggr;) Expression Is Decreased in Pulmonary Hypertension and Affects Endothelial Cell Growth , 2003, Circulation research.

[59]  H. Lodish,et al.  Troglitazone Antagonizes Tumor Necrosis Factor-α-induced Reprogramming of Adipocyte Gene Expression by Inhibiting the Transcriptional Regulatory Functions of NF-κB* , 2003, Journal of Biological Chemistry.

[60]  S. Wedgwood,et al.  Increased Superoxide Generation Is Associated With Pulmonary Hypertension in Fetal Lambs: A Role for NADPH Oxidase , 2003, Circulation research.

[61]  H. Lodish,et al.  Troglitazone antagonizes tumor necrosis factor-alpha-induced reprogramming of adipocyte gene expression by inhibiting the transcriptional regulatory functions of NF-kappaB. , 2003, The Journal of biological chemistry.

[62]  L. Harrison,et al.  Phosphorylation of PPARgamma via active ERK1/2 leads to its physical association with p65 and inhibition of NF-kappabeta. , 2003, Journal of cellular biochemistry.

[63]  W. R. Taylor,et al.  Superoxide Production and Expression of Nox Family Proteins in Human Atherosclerosis , 2002, Circulation.

[64]  J. Lambeth Nox/Duox family of nicotinamide adenine dinucleotide (phosphate) oxidases , 2002, Current opinion in hematology.

[65]  B. Spiegelman,et al.  PPARγ: a Nuclear Regulator of Metabolism, Differentiation, and Cell Growth* , 2001, The Journal of Biological Chemistry.

[66]  B. Spiegelman,et al.  PPARgamma : a nuclear regulator of metabolism, differentiation, and cell growth. , 2001, The Journal of biological chemistry.

[67]  D. Sorescu,et al.  NAD(P)H oxidase: role in cardiovascular biology and disease. , 2000, Circulation research.