Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Am J Physiol Heart Circ Physiol ; 316(3): H566-H579, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30499716

ABSTRACT

Although redox processes closely interplay with mechanoresponses to control vascular remodeling, redox pathways coupling mechanostimulation to cellular cytoskeletal organization remain unclear. The peri/epicellular pool of protein disulfide isomerase-A1 (pecPDIA1) supports postinjury vessel remodeling. Using distinct models, we investigated whether pecPDIA1 could work as a redox-dependent organizer of cytoskeletal mechanoresponses. In vascular smooth muscle cells (VSMCs), pecPDIA1 immunoneutralization impaired stress fiber assembly in response to equibiaxial stretch and, under uniaxial stretch, significantly perturbed cell repositioning perpendicularly to stretch orientation. During cyclic stretch, pecPDIA1 supported thiol oxidation of the known mechanosensor ß1-integrin and promoted polarized compartmentalization of sulfenylated proteins. Using traction force microscopy, we showed that pecPDIA1 organizes intracellular force distribution. The net contractile moment ratio of platelet-derived growth factor-exposed to basal VSMCs decreased from 0.90 ± 0.09 (IgG-exposed controls) to 0.70 ± 0.08 after pecPDI neutralization ( P < 0.05), together with an enhanced coefficient of variation for distribution of force modules, suggesting increased noise. Moreover, in a single cell model, pecPDIA1 neutralization impaired migration persistence without affecting total distance or velocity, whereas siRNA-mediated total PDIA1 silencing disabled all such variables of VSMC migration. Neither expression nor total activity of the master mechanotransmitter/regulator RhoA was affected by pecPDIA1 neutralization. However, cyclic stretch-induced focal distribution of membrane-bound RhoA was disrupted by pecPDI inhibition, which promoted a nonpolarized pattern of RhoA/caveolin-3 cluster colocalization. Accordingly, FRET biosensors showed that pecPDIA1 supports localized RhoA activity at cell protrusions versus perinuclear regions. Thus, pecPDI acts as a thiol redox-dependent organizer and noise reducer mechanism of cytoskeletal repositioning, oxidant generation, and localized RhoA activation during a variety of VSMC mechanoresponses. NEW & NOTEWORTHY Effects of a peri/epicellular pool of protein disulfide isomerase-A1 (pecPDIA1) during mechanoregulation in vascular smooth muscle cells (VSMCs) were highlighted using approaches such as equibiaxial and uniaxial stretch, random single cell migration, and traction force microscopy. pecPDIA1 regulates organization of the cytoskeleton and minimizes the noise of cell alignment, migration directionality, and persistence. pecPDIA1 mechanisms involve redox control of ß1-integrin and localized RhoA activation. pecPDIA1 acts as a novel organizer of mechanoadaptation responses in VSMCs.


Subject(s)
Adaptation, Physiological/physiology , Cytoskeleton/physiology , Myocytes, Smooth Muscle/physiology , Protein Disulfide-Isomerases/physiology , Actin Cytoskeleton/physiology , Animals , Biomechanical Phenomena , Cell Movement , Cells, Cultured , Gene Silencing , Integrin beta1/metabolism , Muscle, Smooth, Vascular/metabolism , Oxidants/metabolism , Pressoreceptors , Protein Disulfide-Isomerases/genetics , Rabbits , rhoA GTP-Binding Protein/metabolism
2.
Proc Natl Acad Sci U S A ; 113(36): E5308-17, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27540115

ABSTRACT

Despite numerous reports implicating NADPH oxidases (Nox) in the pathogenesis of many diseases, precise regulation of this family of professional reactive oxygen species (ROS) producers remains unclear. A unique member of this family, Nox1 oxidase, functions as either a canonical or hybrid system using Nox organizing subunit 1 (NoxO1) or p47(phox), respectively, the latter of which is functional in vascular smooth muscle cells (VSMC). In this manuscript, we identify critical requirement of ezrin-radixin-moesin-binding phosphoprotein 50 (EBP50; aka NHERF1) for Nox1 activation and downstream responses. Superoxide (O2 (•-)) production induced by angiotensin II (AngII) was absent in mouse EBP50 KO VSMC vs. WT. Moreover, ex vivo incubation of aortas with AngII showed a significant increase in O2 (•-) in WT but not EBP50 or Nox1 nulls. Similarly, lipopolysaccharide (LPS)-induced oxidative stress was attenuated in femoral arteries from EBP50 KO vs. WT. In silico analyses confirmed by confocal microscopy, immunoprecipitation, proximity ligation assay, FRET, and gain-/loss-of-function mutagenesis revealed binding of EBP50, via its PDZ domains, to a specific motif in p47(phox) Functional studies revealed AngII-induced hypertrophy was absent in EBP50 KOs, and in VSMC overexpressing EBP50, Nox1 gene silencing abolished VSMC hypertrophy. Finally, ex vivo measurement of lumen diameter in mouse resistance arteries exhibited attenuated AngII-induced vasoconstriction in EBP50 KO vs. WT. Taken together, our data identify EBP50 as a previously unidentified regulator of Nox1 and support that it promotes Nox1 activity by binding p47(phox) This interaction is pivotal for agonist-induced smooth muscle ROS, hypertrophy, and vasoconstriction and has implications for ROS-mediated physiological and pathophysiological processes.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , DNA Helicases/metabolism , Hypertrophy/metabolism , NADPH Oxidase 1/genetics , Phosphoproteins/metabolism , Proteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Adaptor Proteins, Signal Transducing , Angiotensin II/administration & dosage , Angiotensin II/adverse effects , Animals , DNA Helicases/genetics , Femoral Artery/drug effects , Femoral Artery/metabolism , Femoral Artery/pathology , Humans , Hypertrophy/chemically induced , Hypertrophy/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , NADPH Oxidase 1/metabolism , Oxidative Stress/drug effects , Phosphoproteins/genetics , Proteins/genetics , Reactive Oxygen Species/metabolism , Sodium-Hydrogen Exchangers/genetics , Superoxides/metabolism , Vasoconstriction/drug effects , Vasoconstriction/genetics
3.
Hypertension ; 67(3): 613-22, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26781284

ABSTRACT

Whole-vessel remodeling critically determines lumen caliber in vascular (patho)physiology, and it is reportedly redox-dependent. We hypothesized that the cell-surface pool of the endoplasmic reticulum redox chaperone protein disulfide isomerase-A1 (peri/epicellular=pecPDI), which is known to support thrombosis, also regulates disease-associated vascular architecture. In human coronary atheromas, PDI expression inversely correlated with constrictive remodeling and plaque stability. In a rabbit iliac artery overdistension model, there was unusually high PDI upregulation (≈25-fold versus basal, 14 days postinjury), involving both intracellular and pecPDI. PecPDI neutralization with distinct anti-PDI antibodies did not enhance endoplasmic reticulum stress or apoptosis. In vivo pecPDI neutralization with PDI antibody-containing perivascular gel from days 12 to 14 post injury promoted 25% decrease in the maximally dilated arteriographic vascular caliber. There was corresponding whole-vessel circumference loss using optical coherence tomography without change in neointima, which indicates constrictive remodeling. This was accompanied by decreased hydrogen peroxide generation. Constrictive remodeling was corroborated by marked changes in collagen organization, that is, switching from circumferential to radial fiber orientation and to a more rigid fiber type. The cytoskeleton architecture was also disrupted; there was a loss of stress fiber coherent organization and a switch from thin to medium thickness actin fibers, all leading to impaired viscoelastic ductility. Total and PDI-associated expressions of ß1-integrin, and levels of reduced cell-surface ß1-integrin, were diminished after PDI antibody treatment, implicating ß1-integrin as a likely pecPDI target during vessel repair. Indeed, focal adhesion kinase phosphorylation, a downstream ß1-integrin effector, was decreased by PDI antibody. Thus, the upregulated pecPDI pool tunes matrix/cytoskeleton reshaping to counteract inward remodeling in vascular pathophysiology.


Subject(s)
Coronary Stenosis/genetics , Coronary Vessels/pathology , Protein Disulfide-Isomerases/genetics , RNA/genetics , Vascular Remodeling , Animals , Cell Membrane/metabolism , Cells, Cultured , Coronary Stenosis/metabolism , Coronary Stenosis/pathology , Coronary Vessels/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Humans , Male , Phosphorylation , Protein Disulfide-Isomerases/biosynthesis , Rabbits
4.
Arterioscler Thromb Vasc Biol ; 35(2): 430-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25550204

ABSTRACT

OBJECTIVE: Blood vessel hemodynamics have profound influences on function and structure of vascular cells. One of the main mechanical forces influencing vascular smooth muscle cells (VSMC) is cyclic stretch (CS). Increased CS stimulates reactive oxygen species (ROS) production in VSMC, leading to their dedifferentiation, yet the mechanisms involved are poorly understood. This study was designed to test the hypothesis that pathological CS stimulates NADPH oxidase isoform 1 (Nox1)-derived ROS via MEF2B, leading to VSMC dysfunction via a switch from a contractile to a synthetic phenotype. APPROACH AND RESULTS: Using a newly developed isoform-specific Nox1 inhibitor and gene silencing technology, we demonstrate that a novel pathway, including MEF2B-Nox1-ROS, is upregulated under pathological stretch conditions, and this pathway promotes a VSMC phenotypic switch from a contractile to a synthetic phenotype. We observed that CS (10% at 1 Hz) mimicking systemic hypertension in humans increased Nox1 mRNA, protein levels, and enzymatic activity in a time-dependent manner, and this upregulation was mediated by MEF2B. Furthermore, we show that stretch-induced Nox1-derived ROS upregulated a specific marker for synthetic phenotype (osteopontin), whereas it downregulated classical markers for contractile phenotype (calponin1 and smoothelin B). In addition, our data demonstrated that stretch-induced Nox1 activation decreases actin fiber density and augments matrix metalloproteinase 9 activity, VSMC migration, and vectorial alignment. CONCLUSIONS: These results suggest that CS initiates a signal through MEF2B that potentiates Nox1-mediated ROS production and causes VSMC to switch to a synthetic phenotype. The data also characterize a new Nox1 inhibitor as a potential therapy for treatment of vascular dysfunction in hypertension.


Subject(s)
MEF2 Transcription Factors/metabolism , Mechanotransduction, Cellular , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NADH, NADPH Oxidoreductases/metabolism , Pressoreceptors/metabolism , Vascular Remodeling , Animals , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Cell Movement , Cells, Cultured , Cytoskeletal Proteins/metabolism , Enzyme Inhibitors/pharmacology , MEF2 Transcription Factors/genetics , Matrix Metalloproteinase 9/metabolism , Mechanotransduction, Cellular/drug effects , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , Osteopontin/metabolism , Phenotype , RNA Interference , RNA, Messenger/metabolism , Rats , Reactive Oxygen Species/metabolism , Time Factors , Transfection , Vascular Remodeling/drug effects , Calponins
5.
J Am Soc Nephrol ; 25(6): 1171-86, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24511121

ABSTRACT

Ischemia reperfusion injury (IRI) causes tissue and organ injury, in part, through alterations in tissue blood flow and the production of reactive oxygen species. The cell surface receptor signal-regulatory protein-α (SIRP-α) is expressed on inflammatory cells and suppresses phagocytosis, but the function of SIRP-α in IRI has not been determined. We reported previously that the matricellular protein thrombospondin-1 is upregulated in IRI. Here, we report a novel interaction between thrombospondin-1 and SIRP-α on nonphagocytic cells. In cell-free experiments, thrombospondin-1 bound SIRP-α. In vascular smooth muscle cells and renal tubular epithelial cells, treatment with thrombospondin-1 led to phosphorylation of SIRP-α and downstream activation of Src homology domain 2-containing phosphatase-1. Thrombospondin-1 also stimulated phosphorylation of p47(phox) (an organizer subunit for nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1/2) and increased production of superoxide, both of which were abrogated by knockdown or antibody blockade of SIRP-α. In rodent aortic rings, treatment with thrombospondin-1 increased the production of superoxide and inhibited nitric oxide-mediated vasodilation in a SIRP-α-dependent manner. Renal IRI upregulated the thrombospondin-1-SIRP-α signaling axis and was associated with increased superoxide production and cell death. A SIRP-α antibody that blocks thrombospondin-1 activation of SIRP-α mitigated the effects of renal IRI, increasing blood flow, suppressing production of reactive oxygen species, and preserving cellular architecture. A role for CD47 in SIRP-α activation in these pathways is also described. Overall, these results suggest that thrombospondin-1 binding to SIRP-α on nonphagocytic cells activates NADPH oxidase, limits vasodilation, and promotes renal IRI.


Subject(s)
Antigens, Differentiation/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Kidney Diseases/metabolism , Receptors, Immunologic/metabolism , Reperfusion Injury/metabolism , Thrombospondin 1/metabolism , Animals , CD47 Antigen/metabolism , Cells, Cultured , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
6.
J Biol Chem ; 288(51): 36437-50, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24187133

ABSTRACT

Excessive vascular and colon epithelial reactive oxygen species production by NADPH oxidase isoform 1 (Nox1) has been implicated in a number of disease states, including hypertension, atherosclerosis, and neoplasia. A peptide that mimics a putative activation domain of the Nox1 activator subunit NOXA1 (NOXA1 docking sequence, also known as NoxA1ds) potently inhibited Nox1-derived superoxide anion (O2·-) production in a reconstituted Nox1 cell-free system, with no effect on Nox2-, Nox4-, Nox5-, or xanthine oxidase-derived reactive oxygen species production as measured by cytochrome c reduction, Amplex Red fluorescence, and electron paramagnetic resonance. The ability of NoxA1ds to cross the plasma membrane was tested by confocal microscopy in a human colon cancer cell line exclusively expressing Nox1 (HT-29) using FITC-labeled NoxA1ds. NoxA1ds significantly inhibited whole HT-29 carcinoma cell-derived O2·- generation. ELISA and fluorescence recovery after photobleaching experiments indicate that NoxA1ds, but not its scrambled control, binds Nox1. FRET experiments conducted using Nox1-YFP and NOXA1-CFP illustrate that NoxA1ds disrupts the binding interaction between Nox1 and NOXA1, whereas a control peptide did not. Moreover, hypoxia-induced human pulmonary artery endothelial cell O2·- production was completely inhibited by NoxA1ds. Human pulmonary artery endothelial cell migration under hypoxic conditions was also reduced by pretreatment with NoxA1ds. Our data indicate that a peptide recapitulating a putative activation subdomain of NOXA1 (NoxA1ds) is a highly efficacious and selective inhibitor of Nox1 activity and establishes a critical interaction site for Nox1-NOXA1 binding required for enzyme activation.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Catalytic Domain , Cell Movement , Endothelial Cells/metabolism , NADPH Oxidases/metabolism , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Conserved Sequence , Endothelial Cells/enzymology , Endothelial Cells/physiology , Enzyme Activation , HT29 Cells , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Sequence Data , Mutation , NADPH Oxidase 1 , Protein Binding , Pulmonary Artery/cytology , Superoxides/metabolism
7.
Cardiovasc Res ; 97(1): 134-42, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22997161

ABSTRACT

AIMS: Reactive oxygen species (ROS)-mediated intracellular signalling is well described in the vasculature, yet the precise roles of ROS in paracrine signalling are not known. Studies implicate interstitial ROS hydrogen peroxide (H(2)O(2)) in vascular disease, and plasma H(2)O(2) levels in the micromolar range are detectable in animal models and humans with hypertension. Recently, H(2)O(2) was shown to cross biological membranes of non-vascular cells via aquaporin (Aqp) water channels. Previous findings suggest that H(2)O(2) activates NADPH oxidase (Nox) enzymes in vascular cells and apoptosis signal-regulating kinase 1 (Ask1) in non-vascular cells. We hypothesized that extracellular H(2)O(2) induces smooth muscle cell (SMC) hypertrophy by a mechanism involving Aqp1, Nox1, and Ask1. METHODS AND RESULTS: Treatment of rat aortic SMCs (rASMC) with exogenous H(2)O(2) resulted in a concentration-dependent increase in Nox-derived superoxide (O(2)(•-)), determined by L-012 chemiluminescence, cytochrome c and electron paramagnetic resonance. Nox1 was verified as the source of O(2)(·-) by siRNA. Aqp1 siRNA attenuated H(2)O(2) cellular entry and H(2)O(2)-induced O(2)(•-) production. H(2)O(2) treatment increased Ask1 activation and induced rASMC hypertrophy in a Nox1-dependent mechanism. Adenoviral-dominant-negative Ask1 attenuated H(2)O(2)-induced rASMC hypertrophy and adenoviral overexpression of Ask1 augmented it. CONCLUSION: Our results demonstrate for the first time that extracellular H(2)O(2), at pathophysiological concentrations, stimulates rASMC Nox1-derived O(2)(•-), subsequent Ask1 activation and SMC hypertrophy. The data demonstrate a novel pathway by which H(2)O(2) enters vascular cells via aquaporins and activates Nox, leading to hypertrophy, and provide multiple novel targets for combinatorial therapeutics development targeting hypertrophy and vascular disease.


Subject(s)
Aquaporin 1/metabolism , Hydrogen Peroxide/pharmacology , MAP Kinase Kinase Kinase 5/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NADH, NADPH Oxidoreductases/metabolism , Oxidants/pharmacology , Animals , Aquaporin 1/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Electron Spin Resonance Spectroscopy , Enzyme Activation , Flow Cytometry , Hypertrophy , MAP Kinase Kinase Kinase 5/genetics , Microscopy, Confocal , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , NADPH Oxidase 1 , Phosphorylation , RNA Interference , Rats , Signal Transduction/drug effects , Superoxides/metabolism , Time Factors , Transfection
8.
Arterioscler Thromb Vasc Biol ; 32(12): 2966-73, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23087362

ABSTRACT

OBJECTIVE: Although the matricellular protein thrombospondin-1 (TSP1) is highly expressed in the vessel wall in response to injury, its pathophysiological role in the development of vascular disease is poorly understood. This study was designed to test the hypothesis that TSP1 stimulates reactive oxygen species production in vascular smooth muscle cells and induces vascular dysfunction by promoting oxidative stress. METHODS AND RESULTS: Nanomolar concentrations of TSP1 found in human vascular disease robustly stimulated superoxide (O(2)(•-)) levels in vascular smooth muscle cells at both cellular and tissue level as measured by cytochrome c and electron paramagnetic resonance. A peptide mimicking the C terminus of TSP1 known to specifically bind CD47 recapitulated this response. Transcriptional knockdown of CD47 and a monoclonal inhibitory CD47 antibody abrogated TSP1-triggered O(2)(•-) in vitro and ex vivo. TSP1 treatment of vascular smooth muscle cells activated phospholipase C and protein kinase C, resulting in phosphorylation of the NADPH oxidase organizer subunit p47(phox) and subsequent Nox1 activation, leading to impairment of arterial vasodilatation ex vivo. Further, we observed that blockade of CD47 and NADPH oxidase 1 gene silencing in vivo in rats improves TSP1-induced impairment of tissue blood flow after ischemia reperfusion. CONCLUSIONS: Our data suggest a highly regulated process of reactive oxygen species stimulation and blood flow regulation promoted through a direct TSP1/CD47-mediated activation of Nox1. This is the first report, to our knowledge, of a matricellular protein acting as a ligand for NADPH oxidase activation and through specific engagement of integrin-associated protein CD47.


Subject(s)
CD47 Antigen/physiology , Muscle, Smooth, Vascular/physiology , NADH, NADPH Oxidoreductases/physiology , Regional Blood Flow/physiology , Thrombospondin 1/physiology , Animals , CD47 Antigen/genetics , Gene Silencing , Male , Mice , Muscle, Smooth, Vascular/drug effects , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , Oxidative Stress/drug effects , Oxidative Stress/physiology , Protein Kinase C/physiology , Rats , Regional Blood Flow/drug effects , Signal Transduction/physiology , Superoxide Dismutase/physiology , Superoxide Dismutase-1 , Thrombospondin 1/pharmacology , Type C Phospholipases/physiology
9.
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
10.
J Neurol Sci ; 309(1-2): 5-8, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21855089

ABSTRACT

BACKGROUND: Information regarding predisposing factors, frequency, and prognostic implications of new onset atrial fibrillation (NOAF) after carotid endarterectomy (CEA) is scarce. We assessed the frequency, risk factors, and the prognostic impact of NOAF after CEA. METHODS: We assessed every patient undergoing CEA (n = 186) at our academic hospital between 2006 and 2009. Patients underwent continuous electrocardiographic monitoring during surgery and during the rest of hospital stay. We performed univariate and multivariate analyses for identifying variables associated with NOAF and for individualizing variables related to four perioperative adverse outcome measures: a) ischemic stroke; b) ischemic stroke and myocardial infarction, c) ischemic stroke and death, and d) ischemic stroke, myocardial infarction, and death. RESULTS: The study cohort comprised 186 patients. Overall, NOAF was detected in 7 cases (3.8%). The only variable associated with NOAF was intraoperative hypotension (OR 9.6, 95% CI 1.9-47.4, P = .006). There were no perioperative deaths. NOAF was associated with perioperative ischemic stroke and with the combined outcome of ischemic stroke and myocardial infarction. CONCLUSIONS: We found a low frequency of NOAF after CEA. Intraoperative hypotension was associated to a higher risk of NOAF. In turn, NOAF was related to adverse postoperative outcome. Further research is needed to clarify the pathophysiological relation between intraoperative hypotension, NOAF, and adverse CEA outcome.


Subject(s)
Atrial Fibrillation/diagnosis , Endarterectomy, Carotid/adverse effects , Hypotension/diagnosis , Intraoperative Complications/diagnosis , Aged , Aged, 80 and over , Atrial Fibrillation/etiology , Atrial Fibrillation/physiopathology , Cohort Studies , Electrocardiography/methods , Female , Follow-Up Studies , Humans , Hypotension/etiology , Hypotension/physiopathology , Intraoperative Complications/etiology , Intraoperative Complications/physiopathology , Male , Middle Aged , Retrospective Studies , Risk Factors , Treatment Outcome
11.
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
12.
Biochem Biophys Res Commun ; 345(2): 652-9, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16696940

ABSTRACT

The loss of endothelial function is the initiating factor in the development of diabetic vascular disease. Kinins control endothelial function by the activation of two receptors: the B2 which is constitutively expressed, and the B1 which is highly induced in pathological conditions. In the present study, we observed that the levels of B1-receptor mRNA and protein are induced in endothelial cells incubated in high glucose. An increase in B1-receptor was also observed in the endothelial layer of aortas, from 4-week diabetic rats. When cells were grown in high glucose, the B1 agonist des-Arg9-BK increased nitrite levels, whereas in normal glucose nitrite levels were unchanged. Nitrite increase was blocked by L-NAME and 1400W indicating the participation of the inducible Nitric Oxide Synthase (iNOS). iNOS protein levels were also increased in high glucose. These results demonstrate the participation of the B1 receptor in the signaling pathways mediated by kinins in high glucose.


Subject(s)
Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Glucose/pharmacology , Nitric Oxide Synthase/drug effects , Receptor, Bradykinin B1/metabolism , Signal Transduction/drug effects , Amidines/pharmacology , Animals , Benzylamines/pharmacology , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Endothelial Cells/physiology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/metabolism , Nitrites/metabolism , RNA, Messenger/metabolism , Rats , Receptor, Bradykinin B1/drug effects , Signal Transduction/physiology , Time Factors , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...