Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 290(5): 2879-87, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25492869

ABSTRACT

The regulation of the cell cycle by the ubiquitin-proteasome system is dependent on the activity of E3 ligases. Skp2 (S-phase kinase associated protein-2) is the substrate recognition subunit of the E3 ligase that ubiquitylates the cell cycle inhibitors p21(cip1) and p27(kip1) thus promoting cell cycle progression. Increased expression of Skp2 is frequently observed in diseases characterized by excessive cell proliferation, such as cancer and neointima hyperplasia. The stability and cellular localization of Skp2 are regulated by Akt, but the molecular mechanisms underlying these effects remain only partly understood. The scaffolding protein Ezrin-Binding Phosphoprotein of 50 kDa (EBP50) contains two PDZ domains and plays a critical role in the development of neointimal hyperplasia. Here we report that EBP50 directly binds Skp2 via its first PDZ domain. Moreover, EBP50 is phosphorylated by Akt on Thr-156 within the second PDZ domain, an event that allosterically promotes binding to Skp2. The interaction with EBP50 causes cytoplasmic localization of Skp2, increases Skp2 stability and promotes proliferation of primary vascular smooth muscle cells. Collectively, these studies define a novel regulatory mechanism contributing to aberrant cell growth and highlight the importance of scaffolding function of EBP50 in Akt-dependent cell proliferation.


Subject(s)
Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Animals , Cell Proliferation , Cells, Cultured , Humans , Mice , Phosphoproteins/chemistry , Phosphorylation , Protein Binding , Protein Stability , Proto-Oncogene Proteins c-akt/chemistry , S-Phase Kinase-Associated Proteins/chemistry , Sodium-Hydrogen Exchangers/chemistry
2.
PLoS One ; 9(5): e96720, 2014.
Article in English | MEDLINE | ID: mdl-24837600

ABSTRACT

Pulmonary hypertension (PH) is a progressive and fatal disease with no cure. Vascular remodeling in PH involves intraluminal growth of endothelial and smooth muscle cells, leading to obliterative vascular lesions. Cell growth in these lesions is quasi-neoplastic, with evidence of monoclonality, apoptosis resistance and cancer-like metabolic derangements. Herein we tested the effect of human interferon alpha 2b (IFNα), a pleiotropic cytokine and anti-cancer therapeutic, on the development and progression of PH in the rat SU5416/hypoxia (SUH) model and mouse hypoxia model of the disease. In both models IFNα attenuated the development of PH and reversed established PH as assessed by measuring right ventricular systolic pressure and right ventricular hypertrophy. The effect of IFNα was dependent on the type I interferon receptor (IFNAR) since mice lacking a subunit of the IFNAR were not protected by IFNα. Morphometric analysis of pulmonary aterioles from hypoxic mice or SUH rats showed that IFNα inhibited pulmonary vascular remodeling in both models and that IFNα reversed remodeling in SUH rats with established disease. Immunohistochemical staining revealed that IFNα decreased the number of PCNA and Tunel positive cells in the wall of pulmonary arterioles. In vitro, IFNα inhibited proliferation of human pulmonary artery smooth muscle cells and as well as human pulmonary artery endothelial cell proliferation and apoptosis. Together these findings demonstrate that IFNα reverses established experimental PH and provide a rationale for further exploration of the use of IFNα and other immunotherpies in PH.


Subject(s)
Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/prevention & control , Hypoxia/complications , Interferon-alpha/pharmacology , Analysis of Variance , Animals , Blotting, Western , Cells, Cultured , Humans , Hypertension, Pulmonary/etiology , Hypertrophy, Right Ventricular/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Interferon alpha-2 , Interferon-alpha/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Vascular Remodeling/drug effects , Ventricular Pressure/physiology
3.
Circ Res ; 114(10): 1596-600, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24637196

ABSTRACT

RATIONALE: Recent studies demonstrate a role for toll-like receptor 4 (TLR4) in the pathogenesis of pulmonary hypertension (PH); however, the cell types involved in mediating the effects of TLR4 remain unknown. OBJECTIVES: The objective of this study was to determine the contribution of TLR4 expressed on nonparenchymal cells to the pathogenesis of PH. METHODS AND RESULTS: TLR4 bone marrow chimeric mice revealed an equal contribution of TLR4 on nonparenchymal and parenchymal cells in the pathogenesis of PH as determined by measuring right ventricular (RV) systolic pressure and RV hypertrophy. However, the deletion of TLR4 from myeloid lineage cells had no effect on the development of PH because we found no difference in RV systolic pressure or RV hypertrophy in wild-type versus LysM-TLR4(-/-) mice. To explore the potential role of platelet TLR4 in the pathogenesis of PH, platelet-specific TLR4(-/-) mice were generated (PF4-TLR4(-/-) mice). TLR4(-/-) platelets from either global TLR4(-/-) or PF4-TLR4(-/-) mice were functional but failed to respond to lipopolysaccharide, demonstrating a lack of TLR4. PF4-TLR4(-/-) mice demonstrated significant protection from hypoxia-induced PH, including attenuated increases in RV systolic pressure and RV hypertrophy, decreased platelet activation, and less pulmonary vascular remodeling. The deletion of TLR4 from platelets attenuated serotonin release after chronic hypoxia, and lipopolysaccharide-stimulated platelets released serotonin and promoted pulmonary artery smooth muscle cell proliferation in a serotonin-dependent manner. CONCLUSIONS: Our data demonstrate that TLR4 on platelets contributes to the pathogenesis of PH and further highlights the role of platelets in PH.


Subject(s)
Blood Platelets/pathology , Gene Deletion , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/prevention & control , Toll-Like Receptor 4/deficiency , Animals , Blood Platelets/metabolism , Coculture Techniques , Humans , Hypertension, Pulmonary/blood , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiology , Radiation Chimera , Toll-Like Receptor 4/blood , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/physiology
4.
PLoS One ; 8(11): e81903, 2013.
Article in English | MEDLINE | ID: mdl-24312378

ABSTRACT

Although a critical role for caveolae-mediated albumin transcytosis in pulmonary endothelium is well established, considerably less is known about caveolae-independent pathways. In this current study, we confirmed that cultured rat pulmonary microvascular (RPMEC) and pulmonary artery (RPAEC) endothelium endocytosed Alexa488-labeled albumin in a saturable, temperature-sensitive mode and internalization resulted in co-localization by fluorescence microscopy with cholera B toxin and caveolin-1. Although siRNA to caveolin-1 (cav-1) in RPAEC significantly inhibited albumin uptake, a remnant portion of albumin uptake was cav-1-independent, suggesting alternative pathways for albumin uptake. Thus, we isolated and cultured mouse lung endothelial cells (MLEC) from wild type and cav-1(-/-) mice and noted that ~ 65% of albumin uptake, as determined by confocal imaging or live cell total internal reflectance fluorescence microscopy (TIRF), persisted in total absence of cav-1. Uptake of colloidal gold labeled albumin was evaluated by electron microscopy and demonstrated that albumin uptake in MLEC from cav-1(-/-) mice was through caveolae-independent pathway(s) including clathrin-coated pits that resulted in endosomal accumulation of albumin. Finally, we noted that albumin uptake in RPMEC was in part sensitive to pharmacological agents (amiloride [sodium transport inhibitor], Gö6976 [protein kinase C inhibitor], and cytochalasin D [inhibitor of actin polymerization]) consistent with a macropinocytosis-like process. The amiloride sensitivity accounting for macropinocytosis also exists in albumin uptake by both wild type and cav-1(-/-) MLEC. We conclude from these studies that in addition to the well described caveolar-dependent pulmonary endothelial cell endocytosis of albumin, a portion of overall uptake in pulmonary endothelial cells is cav-1 insensitive and appears to involve clathrin-mediated endocytosis and macropinocytosis-like process.


Subject(s)
Albumins/metabolism , Caveolae/physiology , Endocytosis , Endothelium, Vascular/metabolism , Lung/blood supply , Animals , Base Sequence , Caveolin 1/genetics , Caveolin 1/metabolism , Cells, Cultured , DNA Primers , Endothelium, Vascular/cytology , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Pinocytosis , RNA, Small Interfering/genetics , Rats
5.
J Biol Chem ; 288(51): 36426-36, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24196963

ABSTRACT

The interaction between vascular cells and macrophages is critical during vascular remodeling. Here we report that the scaffolding protein, ezrin-binding phosphoprotein 50 (EBP50), is a central regulator of macrophage and vascular smooth muscle cells (VSMC) function. EBP50 is up-regulated in intimal VSMC following endoluminal injury and promotes neointima formation. However, the mechanisms underlying these effects are not fully understood. Because of the fundamental role that inflammation plays in vascular diseases, we hypothesized that EBP50 mediates macrophage activation and the response of vessels to inflammation. Indeed, EBP50 expression increased in primary macrophages and VSMC, and in the aorta of mice, upon treatment with LPS or TNFα. This increase was nuclear factor-κB (NF-κB)-dependent. Conversely, activation of NF-κB was impaired in EBP50-null VSMC and macrophages. We found that inflammatory stimuli promote the formation of an EBP50-PKCζ complex at the cell membrane that induces NF-κB signaling. Macrophage activation and vascular inflammation after acute LPS treatment were reduced in EBP50-null cells and mice as compared with WT. Furthermore, macrophage recruitment to vascular lesions was significantly reduced in EBP50 knock-out mice. Thus, EBP50 and NF-κB participate in a feed-forward loop leading to increased macrophage activation and enhanced response of vascular cells to inflammation.


Subject(s)
Feedback, Physiological , NF-kappa B/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Vasculitis/metabolism , Animals , Aorta/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Macrophages/physiology , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Phosphoproteins/genetics , Protein Kinase C/metabolism , Sodium-Hydrogen Exchangers/genetics , Tumor Necrosis Factor-alpha/pharmacology , Vasculitis/etiology
6.
Mol Med ; 18: 1509-18, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23269975

ABSTRACT

Survival rates for patients with pulmonary hypertension (PH) remain low, and our understanding of the mechanisms involved are incomplete. Here we show in a mouse model of chronic hypoxia (CH)-induced PH that the nuclear protein and damage-associate molecular pattern molecule (DAMP) high mobility group box 1 (HMGB1) contributes to PH via a Toll-like receptor 4 (TLR4)-dependent mechanism. We demonstrate extranuclear HMGB1 in pulmonary vascular lesions and increased serum HMGB1 in patients with idiopathic pulmonary arterial hypertension. The increase in circulating HMGB1 correlated with mean pulmonary artery pressure. In mice, we similarly detected the translocation and release of HMGB1 after exposure to CH. HMGB1-neutralizing antibody attenuated the development of CH-induced PH, as assessed by measurement of right ventricular systolic pressure, right ventricular hypertrophy, pulmonary vascular remodeling and endothelial activation and inflammation. Genetic deletion of the pattern recognition receptor TLR4, but not the receptor for advanced glycation end products, likewise attenuated CH-induced PH. Finally, daily treatment of mice with recombinant human HMGB1 exacerbated CH-induced PH in wild-type (WT) but not Tlr4(-/-) mice. These data demonstrate that HMGB1-mediated activation of TLR4 promotes experimental PH and identify HMGB1 and/or TLR4 as potential therapeutic targets for the treatment of PH.


Subject(s)
HMGB1 Protein/metabolism , Hypertension, Pulmonary/pathology , Toll-Like Receptor 4/metabolism , Adult , Animals , Antibodies, Neutralizing/pharmacology , Chronic Disease , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Familial Primary Pulmonary Hypertension , Female , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/physiopathology , Hypoxia/complications , Hypoxia/pathology , Hypoxia/physiopathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Toll-Like Receptor 4/genetics
7.
J Biol Chem ; 288(2): 1365-73, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23148224

ABSTRACT

In pulmonary hypertension the loss of precapillary arterioles results from vascular injury causing endothelial dysfunction. Endothelial cell migration and proliferation are critical for vascular regeneration. This study focused on the effect of high mobility group box 1 protein (HMGB1) on these critical processes. HMGB1 had no effect on human pulmonary artery endothelial cell (HPAEC) proliferation. In contrast, treatment of HPAECs with HMGB1 dose-dependently inhibited VEGF-stimulated HPAEC migration. The effect of HMGB1 on HPAEC migration was TLR4-dependent because it was reversed by TLR4 siRNA or TLR4-neutralizing antibody. Exposure of HPAECs to hypoxia caused translocation and release of HMGB1 and inhibition of HPAEC migration. The effect of hypoxia on HPAEC migration was mediated by HMGB1 because HMGB1-neutralizing antibody but not control IgG restored HPAEC migration. Likewise, TLR4 siRNA but not control siRNA reversed the inhibitory effect of hypoxia in HPAECs. The canonical TLR4 signaling pathway requires the adaptor protein MyD88 and leads to downstream NFκB activation. Interestingly, HMGB1 failed to stimulate NFκB translocation to the nucleus, but instead activated an alternative pathway characterized by activation of interferon response factor 3 (IRF3). This was in contrast to human umbilical vein endothelial cells in which HMGB1 stimulated nuclear translocation of NFκB but not IRF3. IRF3 siRNA, but not MyD88 siRNA, reversed the inhibitory effect of HMGB1 on HPAEC migration. These data demonstrate that HMGB1 inhibits HPAEC migration, a critical process for vascular regeneration, via TLR4- and IRF3-dependent mechanisms.


Subject(s)
Cell Movement/physiology , HMGB1 Protein/physiology , Interferon Regulatory Factor-3/physiology , Pulmonary Artery/cytology , Toll-Like Receptor 4/physiology , Blotting, Western , Cells, Cultured , Fluorescent Antibody Technique , Humans , Interferon Regulatory Factor-3/genetics , Myeloid Differentiation Factor 88/physiology , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 4/genetics , Vascular Endothelial Growth Factor A/physiology
8.
Am J Physiol Heart Circ Physiol ; 302(12): H2518-27, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22505641

ABSTRACT

Caveolin-1 (Cav-1)-/- mice develop mild pulmonary hypertension as they age. In this study, we sought to determine the effect of chronic hypoxia, an established model of pulmonary hypertension, on young Cav-1-/- mice with no measurable signs of pulmonary hypertension. Exposure of Cav-1-/- mice to chronic hypoxia resulted in an initial rise in right ventricular (RV) systolic pressure (RVSP) similar to wild-type (WT) mice. By three weeks RVSP decreased in the Cav-1-/- mice, whereas it was maintained in WT mice. The drop in RVSP in Cav-1-/- mice was accompanied by decreased cardiac output, increased RV hypertrophy, RV interstitial fibrosis, decreased RV sarco(endo)plasmic reticulum Ca(2+)-ATPase 2a mRNA and decreased RV function compared with WT mice. Importantly, minimal differences were noted in pulmonary vascular remodeling between WT and Cav-1-/- mice, and left ventricular function was normal in hypoxic Cav-1-/- mice. Mechanistically, increased endothelial nitric oxide synthase uncoupling and increased tyrosine nitration of protein kinase G were detected in the RV of Cav-1-/- mice. These hemodynamic, histological, and molecular changes were prevented in Cav-1-/- mice expressing an endothelial-specific Cav-1 transgene or by nitric oxide synthase inhibition. These data suggest that, in Cav-1-/- mice, increased oxidative/nitrosative stress due to endothelial nitric oxide synthase uncoupling modifies the response of the RV to pressure overload, accelerating the deterioration of RV function.


Subject(s)
Blood Pressure/physiology , Caveolin 1/genetics , Heart Failure/etiology , Hypoxia/complications , Animals , Cardiac Output/physiology , Cyclic GMP-Dependent Protein Kinases/metabolism , Heart Failure/genetics , Heart Failure/physiopathology , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/genetics , Hypoxia/physiopathology , Lung/blood supply , Lung/metabolism , Lung/physiopathology , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/physiology
9.
PLoS One ; 7(2): e31495, 2012.
Article in English | MEDLINE | ID: mdl-22355372

ABSTRACT

Chronic alterations in blood flow initiate structural changes in vessel lumen caliber to normalize shear stress. The loss of endothelial derived nitric oxide synthase (eNOS) in mice promotes abnormal flow dependent vascular remodeling, thus uncoupling mechanotransduction from adaptive vascular remodeling. However, the mechanisms of how the loss of eNOS promotes abnormal remodeling are not known. Here we show that abnormal flow-dependent remodeling in eNOS knockout mice (eNOS (-/-)) is associated with activation of the platelet derived growth factor (PDGF) signaling pathway leading to the induction of the inhibitor of apoptosis, survivin. Interfering with PDGF signaling or survivin function corrects the abnormal remodeling seen in eNOS (-/-) mice. Moreover, nitric oxide (NO) negatively regulates PDGF driven survivin expression and cellular proliferation in cultured vascular smooth muscle cells. Collectively, our data suggests that eNOS negatively regulates the PDGF-survivin axis to maintain proportional flow-dependent luminal remodeling and vascular quiescence.


Subject(s)
Blood Vessels/physiopathology , Endothelium, Vascular/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Neovascularization, Physiologic/physiology , Nitric Oxide Synthase Type III/physiology , Platelet-Derived Growth Factor/metabolism , Repressor Proteins/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/cytology , Immunoenzyme Techniques , Inhibitor of Apoptosis Proteins/genetics , Male , Mechanotransduction, Cellular/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Nitric Oxide/metabolism , Platelet-Derived Growth Factor/genetics , Protein Binding , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Signal Transduction , Survivin
10.
Cardiovasc Res ; 93(4): 682-93, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22215724

ABSTRACT

AIMS: Pulmonary arterial hypertension (PAH) is a progressive lung disease characterized by pulmonary vasoconstriction and vascular remodelling, leading to increased pulmonary vascular resistance and right heart failure. Loss of nitric oxide (NO) signalling and increased endothelial nitric oxide synthase (eNOS)-derived oxidative stress are central to the pathogenesis of PAH, yet the mechanisms involved remain incompletely determined. In this study, we investigated the role activated CD47 plays in promoting PAH. METHODS AND RESULTS: We report high-level expression of thrombospondin-1 (TSP1) and CD47 in the lungs of human subjects with PAH and increased expression of TSP1 and activated CD47 in experimental models of PAH, a finding matched in hypoxic human and murine pulmonary endothelial cells. In pulmonary endothelial cells CD47 constitutively associates with caveolin-1 (Cav-1). Conversely, in hypoxic animals and cell cultures activation of CD47 by TSP1 disrupts this constitutive interaction, promoting eNOS-dependent superoxide production, oxidative stress, and PAH. Hypoxic TSP1 null mice developed less right ventricular pressure and hypertrophy and markedly less arteriole muscularization compared with wild-type animals. Further, therapeutic blockade of CD47 activation in hypoxic pulmonary artery endothelial cells upregulated Cav-1, increased Cav-1CD47 co-association, decreased eNOS-derived superoxide, and protected animals from developing PAH. CONCLUSION: Activated CD47 is upregulated in experimental and human PAH and promotes disease by limiting Cav-1 inhibition of dysregulated eNOS.


Subject(s)
CD47 Antigen/metabolism , Caveolin 1/metabolism , Hypertension, Pulmonary/metabolism , Lung/metabolism , Signal Transduction/physiology , Up-Regulation/physiology , Animals , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/pathology , Hypoxia/metabolism , Hypoxia/pathology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocrotaline/adverse effects , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Thrombospondin 1/deficiency , Thrombospondin 1/genetics , Thrombospondin 1/metabolism
11.
Arterioscler Thromb Vasc Biol ; 32(1): 33-41, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22034511

ABSTRACT

OBJECTIVE: The Ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50) is a scaffolding protein known to regulate ion homeostasis in the kidney and intestine. Previous work showed that EBP50 expression increases after balloon injury in rat carotids. This study was designed to determine the role of EBP50 on vascular smooth muscle cells (VSMC) proliferation and the development of neointimal hyperplasia. METHODS AND RESULTS: Wire injury was performed in wild type (WT) and EBP50 knockout (KO) mice. Two weeks after injury, neointima formation was 80% lower in KO than in WT mice. Proliferation of KO VSMC was significantly lower than WT cells and overexpression of EBP50 increased VSMC proliferation. Akt activity and expression of S-phase kinase protein2 decreased in KO cells resulting in the stabilization of the cyclin-dependent kinase inhibitor, p21(cip1). Consequently, KO cells were arrested in G(0)/G(1) phase. Consistent with these observations, p21(cip1) was detected in injured femoral arteries of KO but not WT mice. No differences in apoptosis between WT and KO were observed. CONCLUSIONS: EBP50 is critical for neointima formation and induces VSMC proliferation by decreasing S-phase kinase protein2 stability, thereby accelerating the degradation of the cell cycle inhibitor p21(cip1).


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/physiology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/physiology , Neointima/etiology , Phosphoproteins/physiology , S-Phase Kinase-Associated Proteins/physiology , Sodium-Hydrogen Exchangers/physiology , Animals , Cell Proliferation , Femoral Artery/injuries , Femoral Artery/pathology , Femoral Artery/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neointima/pathology , Neointima/physiopathology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Sodium-Hydrogen Exchangers/genetics
12.
PLoS One ; 6(12): e28578, 2011.
Article in English | MEDLINE | ID: mdl-22194859

ABSTRACT

BACKGROUND: Evidence suggests a role of both innate and adaptive immunity in the development of pulmonary arterial hypertension. The complement system is a key sentry of the innate immune system and bridges innate and adaptive immunity. To date there are no studies addressing a role for the complement system in pulmonary arterial hypertension. METHODOLOGY/PRINCIPAL FINDINGS: Immunofluorescent staining revealed significant C3d deposition in lung sections from IPAH patients and C57Bl6/J wild-type mice exposed to three weeks of chronic hypoxia to induce pulmonary hypertension. Right ventricular systolic pressure and right ventricular hypertrophy were increased in hypoxic vs. normoxic wild-type mice, which were attenuated in C3-/- hypoxic mice. Likewise, pulmonary vascular remodeling was attenuated in the C3-/- mice compared to wild-type mice as determined by the number of muscularized peripheral arterioles and morphometric analysis of vessel wall thickness. The loss of C3 attenuated the increase in interleukin-6 and intracellular adhesion molecule-1 expression in response to chronic hypoxia, but not endothelin-1 levels. In wild-type mice, but not C3-/- mice, chronic hypoxia led to platelet activation as assessed by bleeding time, and flow cytometry of platelets to determine cell surface P-selectin expression. In addition, tissue factor expression and fibrin deposition were increased in the lungs of WT mice in response to chronic hypoxia. These pro-thrombotic effects of hypoxia were abrogated in C3-/- mice. CONCLUSIONS: Herein, we provide compelling genetic evidence that the complement system plays a pathophysiologic role in the development of PAH in mice, promoting pulmonary vascular remodeling and a pro-thrombotic phenotype. In addition we demonstrate C3d deposition in IPAH patients suggesting that complement activation plays a role in the development of PAH in humans.


Subject(s)
Complement C3/deficiency , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/prevention & control , Hypoxia/complications , Animals , Arterioles/metabolism , Arterioles/pathology , Biomarkers/metabolism , Cell Proliferation , Chronic Disease , Complement C3/metabolism , Complement C3a/metabolism , Complement C5a/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Fibrin/metabolism , Gene Deletion , Humans , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Platelet Activation , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Thromboplastin/metabolism , Up-Regulation/genetics
13.
J Clin Invest ; 121(9): 3747-55, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21804187

ABSTRACT

Aberrant regulation of eNOS and associated NO release are directly linked with various vascular diseases. Caveolin-1 (Cav-1), the main coat protein of caveolae, is highly expressed in endothelial cells. Its scaffolding domain serves as an endogenous negative regulator of eNOS function. Structure-function analysis of Cav-1 has shown that phenylalanine 92 (F92) is critical for the inhibitory actions of Cav-1 toward eNOS. Herein, we show that F92A-Cav-1 and a mutant cell-permeable scaffolding domain peptide called Cavnoxin can increase basal NO release in eNOS-expressing cells. Cavnoxin reduced vascular tone ex vivo and lowered blood pressure in normal mice. In contrast, similar experiments performed with eNOS- or Cav-1-deficient mice showed that the vasodilatory effect of Cavnoxin is abolished in the absence of these gene products, which indicates a high level of eNOS/Cav-1 specificity. Mechanistically, biochemical assays indicated that noninhibitory F92A-Cav-1 and Cavnoxin specifically disrupted the inhibitory actions of endogenous Cav-1 toward eNOS and thereby enhanced basal NO release. Collectively, these data raise the possibility of studying the inhibitory influence of Cav-1 on eNOS without interfering with the other actions of endogenous Cav-1. They also suggest a therapeutic application for regulating the eNOS/Cav-1 interaction in diseases characterized by decreased NO release.


Subject(s)
Caveolin 1/chemistry , Caveolin 1/genetics , Caveolin 1/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/biosynthesis , Peptides/metabolism , Vasodilation/physiology , Animals , Cells, Cultured , Humans , Mice , Mice, Knockout , Peptides/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
14.
J Biol Chem ; 286(38): 33134-40, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21808054

ABSTRACT

Activation of bone morphogenetic protein (BMP) receptor II (BMPRII) promotes pulmonary artery endothelial cell (PAEC) survival, proliferation, and migration. Mutations to BMPRII are associated with the development of pulmonary arterial hypertension (PAH). Endothelial dysfunction, including decreased endothelial nitric-oxide synthase (eNOS) activity and loss of bioactive nitric oxide (NO), plays a prominent role in the development of PAH. We hypothesized that stimulation of BMPRII promotes normal PAEC function by activating eNOS. We report that BMPRII ligands, BMP2 and BMP4, (i) stimulate eNOS phosphorylation at a critical regulatory site, (ii) increase eNOS activity, and (iii) result in canonical changes in eNOS protein-protein interactions. The stimulation of eNOS activity by BMPRII ligands was largely dependent on protein kinase A (PKA) activation, as demonstrated using the PKA inhibitors H89 and myristoylated PKI(6-22) amide. PAEC migration stimulated by BMP2 and BMP4 was inhibited by the NOS inhibitor l-nitroarginine methyl ester, providing functional evidence of eNOS activation. Furthermore, BMP2 and BMP4 failed to stimulate eNOS phosphorylation when BMPRII was knocked down by siRNA. Most important to the pathophysiology of the disease, BMP2 and BMP4 failed to stimulate eNOS phosphorylation in PAECs isolated from patients with mutations in the BMPR2 gene. These data demonstrate a new action of BMPs/BMPRII in the pulmonary endothelium and provide novel mechanistic insight into the pathogenesis of PAH.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Cattle , Cell Movement/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Activation/drug effects , Humans , Mutation/genetics , Phosphorylation/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/cytology
15.
Cardiovasc Res ; 88(3): 471-81, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20610415

ABSTRACT

AIMS: Thrombospondin-1 (TSP1), via its necessary receptor CD47, inhibits nitric oxide (NO)-stimulated soluble guanylate cyclase activation in vascular smooth muscle cells, and TSP1-null mice have increased shear-dependent blood flow compared with wild-type mice. Yet, the endothelial basement membrane should in theory function as a barrier to diffusion of soluble TSP1 into the arterial smooth muscle cell layer. These findings suggested that endothelial-dependent differences in blood flow in TSP1-null mice may be the result of direct modulation of endothelial NO synthase (eNOS) activation by circulating TSP1. Here we tested the hypothesis that TSP1 inhibits eNOS activation and endothelial-dependent arterial relaxation. METHODS AND RESULTS: Acetylcholine (ACh)-stimulated activation of eNOS and agonist-driven calcium transients in endothelial cells were inhibited by TSP1. TSP1 also inhibited eNOS phosphorylation at serine(1177). TSP1 treatment of the endothelium of wild-type and TSP1-null but not CD47-null arteries inhibited ACh-stimulated relaxation. TSP1-null vessels demonstrated greater endothelial-dependent vasorelaxation compared with the wild type. Conversely, TSP1-null arteries demonstrated less vasoconstriction to phenylephrine compared with the wild type, which was corrected upon inhibition of eNOS. In TSP1-null mice, intravenous TSP1 blocked ACh-stimulated decreases in blood pressure, and both intravenous TSP1 and a CD47 agonist antibody acutely elevated blood pressure in mice. CONCLUSION: TSP1, via CD47, inhibits eNOS activation and endothelial-dependent arterial relaxation and limits ACh-driven decreases in blood pressure. Conversely, intravenous TSP1 and a CD47 antibody increase blood pressure. These findings suggest that circulating TSP1, by limiting endogenous NO production, functions as a pressor agent supporting blood pressure.


Subject(s)
Blood Pressure/physiology , Endothelium, Vascular/physiology , Nitric Oxide Synthase Type III/physiology , Thrombospondin 1/physiology , Vasodilation/physiology , Acetylcholine/pharmacology , Animals , Antibodies/pharmacology , Blood Pressure/drug effects , CD47 Antigen/immunology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Mice , Models, Animal , Nitric Oxide Synthase Type III/drug effects , Phenylephrine/pharmacology , Thrombospondin 1/genetics , Thrombospondin 1/pharmacology , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
16.
Arterioscler Thromb Vasc Biol ; 30(1): 98-104, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19875720

ABSTRACT

OBJECTIVE: Heme oxygenase-1 (HO-1), via its enzymatic degradation products, exhibits cell and tissue protective effects in models of vascular injury and disease. The migration of vascular smooth muscle cells (VSMC) from the medial to the intimal layer of blood vessels plays an integral role in the development of a neointima in these models. Despite this, there are no studies addressing the effect of increased HO-1 expression on VSMC migration. Results and Methods- The effects of increased HO-1 expression, as well as biliverdin, bilirubin, and carbon monoxide (CO), were studied in in vitro models of VSMC migration. Induction of HO-1 or CO, but not biliverdin or bilirubin, inhibited VSMC migration. This effect was mediated by the inhibition of Nox1 as determined by a range of approaches, including detection of intracellular superoxide, nicotinamide adenine dinucleotide phosphate oxidase activity measurements, and siRNA experiments. Furthermore, CO decreased platelet-derived growth factor-stimulated, redox-sensitive signaling pathways. CONCLUSIONS: Herein, we demonstrate that increased HO-1 expression and CO decreases platelet-derived growth factor-stimulated VSMC migration via inhibition of Nox1 enzymatic activity. These studies reveal a novel mechanism by which HO-1 and CO may mediate their beneficial effects in arterial inflammation and injury.


Subject(s)
Cell Movement/physiology , Heme Oxygenase (Decyclizing)/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Vasculitis/metabolism , Adenoviridae/genetics , Animals , Aorta/cytology , Bilirubin/metabolism , Biliverdine/metabolism , Carbon Monoxide/metabolism , Cell Movement/drug effects , Cells, Cultured , Heme Oxygenase (Decyclizing)/genetics , In Vitro Techniques , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , NADPH Oxidases/metabolism , Oxidation-Reduction , Oxygen/metabolism , Platelet-Derived Growth Factor/pharmacology , Rats , Signal Transduction/physiology , Tunica Intima/cytology
17.
Circulation ; 121(1): 98-109, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20026772

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension is a progressive proliferative vasculopathy of the small pulmonary arteries that is characterized by a primary failure of the endothelial nitric oxide and prostacyclin vasodilator pathways, coupled with dysregulated cellular proliferation. We have recently discovered that the endogenous anion salt nitrite is converted to nitric oxide in the setting of physiological and pathological hypoxia. Considering the fact that nitric oxide exhibits vasoprotective properties, we examined the effects of nitrite on experimental pulmonary arterial hypertension. METHODS AND RESULTS: We exposed mice and rats with hypoxia or monocrotaline-induced pulmonary arterial hypertension to low doses of nebulized nitrite (1.5 mg/min) 1 or 3 times a week. This dose minimally increased plasma and lung nitrite levels yet completely prevented or reversed pulmonary arterial hypertension and pathological right ventricular hypertrophy and failure. In vitro and in vivo studies revealed that nitrite in the lung was metabolized directly to nitric oxide in a process significantly enhanced under hypoxia and found to be dependent on the enzymatic action of xanthine oxidoreductase. Additionally, physiological levels of nitrite inhibited hypoxia-induced proliferation of cultured pulmonary artery smooth muscle cells via the nitric oxide-dependent induction of the cyclin-dependent kinase inhibitor p21(Waf1/Cip1). The therapeutic effect of nitrite on hypoxia-induced pulmonary hypertension was significantly reduced in the p21-knockout mouse; however, nitrite still reduced pressures and right ventricular pathological remodeling, indicating the existence of p21-independent effects as well. CONCLUSIONS: These studies reveal a potent effect of inhaled nitrite that limits pathological pulmonary arterial hypertrophy and cellular proliferation in the setting of experimental pulmonary arterial hypertension.


Subject(s)
Hypertension, Pulmonary/drug therapy , Hypoxia/drug therapy , Nitric Oxide/metabolism , Sodium Nitrite/pharmacology , Xanthine Dehydrogenase/metabolism , Administration, Inhalation , Animals , Cell Division/drug effects , Cells, Cultured , Chronic Disease , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocrotaline/toxicity , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Pulmonary Artery/cytology , Rats , Rats, Sprague-Dawley , Sodium Nitrite/pharmacokinetics , Xanthine Dehydrogenase/antagonists & inhibitors
18.
Circ Res ; 105(10): 965-72, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19797175

ABSTRACT

RATIONALE: Fatty acid nitroalkenes are endogenously generated electrophilic byproducts of nitric oxide and nitrite-dependent oxidative inflammatory reactions. Existing evidence indicates nitroalkenes support posttranslational protein modifications and transcriptional activation that promote the resolution of inflammation. OBJECTIVE: The aim of this study was to assess whether in vivo administration of a synthetic nitroalkene could elicit antiinflammatory actions in vivo using a murine model of vascular injury. METHODS AND RESULTS: The in vivo administration (21 days) of nitro-oleic acid (OA-NO(2)) inhibited neointimal hyperplasia after wire injury of the femoral artery in a murine model (OA-NO(2) treatment resulted in reduced intimal area and intima to media ratio versus vehicle- or oleic acid (OA)-treated animals,P<0.0001). Increased heme oxygenase (HO)-1 expression accounted for much of the vascular protection induced by OA-NO(2) in both cultured aortic smooth muscle cells and in vivo. Inhibition of HO by Sn(IV)-protoporphyrin or HO-1 small interfering RNA reversed OA-NO(2)-induced inhibition of platelet-derived growth factor-stimulated rat aortic smooth muscle cell migration. The upregulation of HO-1 expression also accounted for the antistenotic actions of OA-NO(2) in vivo, because inhibition of neointimal hyperplasia following femoral artery injury was abolished in HO-1(-/-) mice (OA-NO(2)-treated wild-type versus HO-1(-/-) mice, P=0.016). CONCLUSIONS: In summary, electrophilic nitro-fatty acids induce salutary gene expression and cell functional responses that are manifested by a clinically significant outcome, inhibition of neointimal hyperplasia induced by arterial injury.


Subject(s)
Femoral Artery/enzymology , Femoral Artery/injuries , Heme Oxygenase (Decyclizing)/biosynthesis , Nitro Compounds/pharmacology , Oleic Acids/pharmacology , Tunica Intima/enzymology , Animals , Cell Movement/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Inflammation/metabolism , Mice , Mice, Knockout , Nitric Oxide/metabolism , Nitro Compounds/metabolism , Oleic Acids/metabolism , Oxidation-Reduction/drug effects , Platelet-Derived Growth Factor/pharmacology , Rats , Up-Regulation/drug effects
19.
Biochem Biophys Res Commun ; 375(4): 557-61, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18725205

ABSTRACT

Recent studies demonstrate the interaction of BMPRII and caveolin-1 in various cell types. In this study we test the hypothesis that caveolin-1 interacts with and regulates BMPRII-dependent signaling in vascular smooth muscle cells. We demonstrate that BMPRII localizes to caveolae and directly interacts with caveolin-1 in mouse aortic smooth muscle cells. We demonstrate that this interaction is mediated by the caveolin-1 scaffolding domain and is regulated by caveolin-1 phosphorylation. Downregulation of caveolin-1 via siRNA resulted in a loss of BMP-dependent SMAD phosphorylation and gene regulation. Further studies revealed that loss of caveolin-1 results in decreased BMPRII membrane localization and decreased association of BMPRII with the type I BMP receptor BMPRIa. Dominant negative caveolin-1 decreased BMPRII membrane localization suggesting a role for caveolin-1 in BMPRII trafficking. Taken together, our findings establish caveolin-1 as an important regulator of downstream signaling and membrane targeting of BMPRII in vascular smooth muscle cells.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/metabolism , Caveolin 1/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Aorta/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Caveolin 1/genetics , Cell Membrane/enzymology , Down-Regulation , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Signal Transduction , Smad Proteins/metabolism
20.
Proc Natl Acad Sci U S A ; 104(42): 16468-73, 2007 Oct 16.
Article in English | MEDLINE | ID: mdl-17921256

ABSTRACT

Little is known about the tyrosine kinase c-Src's function in the systemic circulation, in particular its role in arterial responses to hormonal stimuli. In human aortic and venous endothelial cells, c-Src is indispensable for 17beta-estradiol (E2)-stimulated phosphatidylinositol 3-kinase/Akt/endothelial NO synthase (eNOS) pathway activation, a possible mechanism in E2-mediated vascular protection. Here we show that c-Src supports basal and E2-stimulated NO production and is required for E2-induced vasorelaxation in murine aortas. Only membrane c-Src is structurally and functionally involved in E2-induced eNOS activation. Independent of c-Src kinase activity, c-Src is associated with an N-terminally truncated estrogen receptor alpha variant (ER46) and eNOS in the plasma membrane through its "open" (substrate-accessible) conformation. In the presence of E2, c-Src kinase is activated by membrane ER46 and in turn phosphorylates ER46 for subsequent ER46 and c-Src membrane recruitment, the assembly of an eNOS-centered membrane macrocomplex, and membrane-initiated eNOS activation. Overall, these results provide insights into a critical role for the tyrosine kinase c-Src in estrogen-stimulated arterial responses, and in membrane-initiated rapid signal transduction, for which obligate complex assembly and localization require the c-Src substrate-accessible structure.


Subject(s)
Aorta/enzymology , Estrogen Receptor alpha/metabolism , Nitric Oxide Synthase Type III/metabolism , Protein-Tyrosine Kinases/metabolism , Vasodilation , Animals , Aorta/drug effects , CSK Tyrosine-Protein Kinase , Cell Membrane/chemistry , Cell Membrane/metabolism , Enzyme Activation , Estradiol/pharmacology , Estrogen Receptor alpha/analysis , Estrogen Receptor alpha/genetics , Humans , Mice , Phosphorylation , Protein Structure, Tertiary , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/chemistry , RNA Splicing , src-Family Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...